Chapter 14

Biofunctionalization of Hydrogels for Engineering the Cellular Microenvironment

Maniraj Bhagawati and Sanjay Kumar,    Department of Bioengineering, University of California, Berkeley, CA, USA

The in vivo cellular microenvironment is a complex network of proteins and carbohydrates that not only acts as a scaffold for cell adhesion but also provides a wide range of biochemical and physical signals that are important for directing cell behavior. This extracellular matrix (ECM) is in turn also a substrate for cell-induced degradation and remodeling. Current advances in tissue engineering and regenerative medicine have inspired the fabrication of in vitro material systems that can mimic biologically instructive properties of the ECM. In this chapter, we focus on strategies that have been developed for the functionalization of hydrogels with a wide range of biological motifs including ECM-derived proteins and growth factors. First, we consider approaches based on irreversible covalent immobilization as well as reversible and affinity-based immobilization of biomolecules onto polymer scaffolds. Next, we address approaches for the introduction of biodegradable elements such as substrates for cell-secreted enzymes into hydrogel matrices. We conclude the discussion with a short overview of techniques for synthesizing hydrogel nanoparticles, which can be used for functionalizing the surfaces of cells.

Keywords

Extracellular matrix; growth factor; hydrogel; protein immobilization; biodegradation; matrix metalloproteinase; nanoparticle

14.1 The 3D Extracellular Milieu

Tissues are composed of cells and the extracellular matrix (ECM), which provides not only physical support in the form of a scaffold but also a variety of biochemical and biomechanical signals essential for cellular function and homeostasis. The ECM is an intricate network of biopolymers that is secreted and subsequently modified, degraded, and organized by the cells adherent to it. Therefore, cells and their ECM form a closed, bidirectional loop of information transfer in which the ECM provides biochemical and mechanical signals that are conveyed from the cell membrane through the cytoskeleton to the nucleus to direct cell behavior, with cells in turn remodeling the ECM under the regulation of ECM-derived inputs [1]. Although all ECMs are nominally composed of proteins and polysaccharides, each tissue has an ECM that is distinct not only in its biochemical composition but also with respect to its physical properties, including stiffness, microstructure, and porosity. A large and still-growing body of work has demonstrated the functional importance of the ECM in regulating morphogenesis and organogenesis and mediating disease processes that occur as a result of abnormal or deregulated ECM [25]. Thus, the ability to fabricate in vitro cellular environments that can mimic the complex properties of the ECM holds profound value for a wide variety of applications ranging from basic cell biological studies to regenerative medicine and drug delivery.

The biochemical components of the ECM have been extensively described in several previous reviews [68] (Figure 14.1), and a variety of ECM proteins have been incorporated into in vitro systems for cell culture applications [912]. These include fibrous proteins such as collagen and fibrin, which play important roles in providing mechanical support in tissue. More specialized and tissue-specific ECM proteins, such as fibronectin (FN) and laminin, have also been included in these culture systems [13,14]. Importantly, these proteins are not merely inert scaffolds but also specifically bind cell surface adhesion receptors. The most notable among these are the integrins, which are heterodimeric transmembrane proteins that physically link the ECM and the cellular cytoskeleton [1520]. Domain-mapping analyses of various ECM proteins have yielded the identity of several short peptide sequences that define integrin-binding specificity [21]. The best-known such sequence, and the one that has most often been included in synthetic matrices for enabling cell adhesion, is the arginine–glycine–aspartate (RGD) sequence, which was originally derived from FN repeat III10 [22]. Since then this sequence has also been found and shown to mediate cell adhesion in several other ECM proteins such as vitronectin, fibrinogen, laminin, and tenascin [21].

image
Figure 14.1 A highly simplified representation of some of the major components of the ECM and their interactions with cells. Source: Reproduced with permission from Ref. [44].

A second category of ECM macromolecules that has also been extensively used for fabrication of in vitro cell culture matrices is glycosaminoglycans (GAGs). GAGs are large linear polysaccharides consisting of disaccharide units containing an amino sugar (either GlcNAc or GalNAc) and an uronic acid (either glucuronic acid and/or iduronic acid). The amino sugar is very often sulfated, and thus GAGs are highly negatively charged polymers. GAGs, which are defined by the constituent sugar monomers and the bonds linking them together, include hyaluronans, heparan sulfates (HSs), chondroitin sulfate, dermatan sulfate, and keratan sulfate. Except for hyaluronan, all GAGs can be found covalently conjugated with proteins to form proteoglycans. In vivo, GAGs and proteoglycans perform multiple structural and mechanical roles and can trigger cell signaling by engaging cell surface receptors [23]. Another major function of GAGs, especially HS, is to locally sequester and concentrate a diverse set of growth factors and cytokines [24]. This property has inspired the use of HS (and the structurally similar molecule heparin) for enabling the entrapment of GFs in several synthetic matrices [25,26].

As alluded to earlier, the composition of the ECM is not static but is instead subject to active cell-driven remodeling, which is an important mechanism for dynamically regulating various aspects of both morphogenesis and tissue homeostasis and disease [27,28]. The major mechanism for this remodeling is degradation of the various components of the ECM by the action of cell-secreted enzymes. ECM proteins including collagens, FN, and laminin are degraded by enzymes such as matrix metalloproteinases (MMPs) [29,30] and plasmin [31]. Proteoglycans can be degraded by ADAMTS proteinases [32,33], and enzymes like hyaluronidases [34] and heparanase are specialized to degrade specific GAGs. In the context of synthetic cell culture scaffolds, cell-mediated matrix degradation can both create room for tissue growth and angiogenesis and provide a convenient mechanism for clearing the material following in vivo implantation. For these reasons, as we discuss later, there has been a significant effort to develop synthetic ECMs that are amenable to cell-mediated degradation.

14.2 Mimicking the ECM

To summarize, the ECM represents a complex, cell-interactive meshwork of proteins, proteoglycans, and GAGs that sequesters and organizes growth factors and cytokines, offers structural support to tissue, and transduces biochemical signals through specialized adhesion receptors. Thus, developing in vitro materials systems that replicate key structural and functional properties of the ECM could both facilitate the study of cell biology in tissue-like settings and provide important platforms for tissue engineering, drug discovery, and delivery of therapeutic cells and proteins in vivo [3541]. Hydrogels, which are water-swollen, 3D networks of polymers, have demonstrated great potential to accomplish these goals. Hydrogel networks can be structurally stabilized by a number of mechanisms such as covalent cross-linking, physical entanglement, formation of crystallites, and assembly through a variety of noncovalent interactions including hydrogen bonding and electrostatic forces [42,43]. The highly water-swollen nature of hydrogels confers important mechanical properties and facilitates the encapsulation of cells. Hydrogel materials often exhibit strong biocompatibility and high permeability for oxygen, nutrients, and other water-soluble metabolites [4449].

Hydrogels have been fabricated from both natural and synthetic polymers. Natural polymers are typically biocompatible, inherently biodegradable, and often contain motifs that direct specific biological functions. ECM-derived proteins such as elastin, fibrin, collagen, and the collagen derivative gelatin have been traditionally used to form hydrogels [5053]. Despite their advantages, ECM proteins derived from tissue can suffer from concerns over immunogenicity, retention of infectious agents, poor scalability, and high batch-to-batch variability. Furthermore, hydrogels based on natural polymers offer limited opportunity for bottom-up design of desired physicochemical properties or biological functionalities. For these reasons, hydrogels based on recombinantly expressed proteins have recently emerged as a complementary class of biological matrices. One of the most well-known examples of such systems is inspired from recurring amino acid sequences found in tropoelastin and are thus known as elastin-like polypeptides (ELPs) [54]. ELP-based hydrogels have proved promising for both experimentally modeling and serving as a therapeutic platform for a wide variety of tissues including nerve, skeletal muscle, and cartilage [5559]. Peptide hydrogels have also been fabricated using amphiphilic diblock copeptides [60,61] and peptides that fold to form amphiphilic β-hairpins that subsequently self-assemble into a hydrogel network [6264]. Advances in protein- and peptide-based hydrogels have been covered in several recent reviews [65,66].

Tissue-derived GAGs and a variety of non-ECM polysaccharides have also been extensively used for fabricating hydrogels. These polysaccharides can be modified with diverse functional groups such as acrylates, thiols, and amines to allow crosslinking. Hyaluronic acid (HA), which is ubiquitous in the ECM of many tissues, has been extensively used to fabricate hydrogels for application in a wide range of study systems [67,68]. Since HA levels have been shown to be elevated in the stroma of cancers [69,70], hydrogels based on HA have been used for modeling the tumor microenvironment in breast, colon, ovarian, and lung cancers [7173]. HA also forms a large fraction of the ECM in brain, and so HA-based hydrogels have also been used to model brain ECM in vitro. Such hydrogels have found particular use in the study of the brain tumor glioblastoma multiforme [74,75]. Other polysaccharides that have been utilized for fabricating hydrogels include heparin, chitosan, dextran, and alginate [7680].

Synthetic polymer hydrogel systems have a long history of use in medical devices, implants, and other materials, and remain of significant interest for use as ECM scaffolds. In general, synthetic polymers offer greater batch-to-batch consistency and significantly more physicochemical tailorability than native biological materials. For example, poly(ethyleneglycol) (PEG) is perhaps the most widely used synthetic polymer hydrogel system. PEG has several unique properties such as high solubility in water and various organic solvents, low intrinsic toxicity and immunogenicity, and comparatively low nonspecific protein adsorption [8185]. In addition, the hydroxyl end groups of PEG are easily modified with other chemical functionalities that can be used for crosslinking as well as for functionalization with a variety of cell-reactive moieties. While a detailed discussion of all synthetic polymer systems currently under use for hydrogel fabrication is beyond the scope of this chapter, several extensive reviews have been written on this subject [44,86,87].

While native, recombinant, and synthetic hydrogels have their relative advantages and disadvantages, many of these systems share the need for chemical “postprocessing” to achieve maximal biological function. For example, whereas native protein hydrogel systems such as those based on collagen and fibrin encode a rich palette of biological instruction, they may require chemical crosslinking to achieve the desired mechanical properties for a given application or conjugation of growth factors to promote specific phenotypes. Conversely, whereas a PEG hydrogel may have ideal physical properties for a given biological application, its bio-inert nature requires all adhesive and biodegradation properties to be built into the system, typically by attachment of proteins and peptides to the polymer backbone. We now discuss these conjugation strategies (Figure 14.2) in greater detail.

image
Figure 14.2 Strategies for capturing exogenous and cell-derived proteins on hydrogel matrices.

14.2.1 Covalent Immobilization of ECM-Derived Proteins and Growth Factors Through Reaction of Amino Acid Side Chains

The side chains of several amino acids offer chemically reactive handles for bioconjugation. These include the amine group of lysine, the thiol group of cysteine, and the carboxylic acid groups of aspartic and glutamic acid. With the development of orthogonal genetic codes and synthetic tRNA systems that enable the expression of recombinant proteins containing unnatural amino acids, the reactive functionalities available on proteins have expanded even further [8890]. Therefore, covalent coupling of these functionalities with reactive groups on hydrogels or hydrogel precursors is one of the most heavily utilized strategies for immobilization of proteins and peptides on hydrogels. Full-length FN, laminin, and collagen, as well as adhesive peptide motifs derived from these and other proteins have been incorporated into hydrogels using this strategy [9193]. Growth factors and derived peptide motifs have also been immobilized on a variety of matrices using covalent reaction with lysine and cysteine residues (Figure 14.2) [94,95].

Lysines can be directly conjugated to scaffold carboxylic acids or their N-hydroxysuccinimide (NHS) derivatives leading to the formation of stable amide bonds. In this strategy, the carboxylic acid has to be incorporated into the hydrogel prior to protein immobilization. If the polymer does not already contain carboxylate functionality, it can be introduced either by copolymerizing carboxylic acid containing monomers with the polymer backbone or by post hoc carboxylate functionalization of the assembled hydrogel. The first strategy has proved valuable for modifying acrylamide-based hydrogels copolymerized with NHS-ester moieties with ECM proteins such as FN, vitronectin, and collagen [91]. Similarly, copolymerization of acrylic acid with PEG-diacrylate (PEGDA) has facilitated the formation of hydrogels which could be efficiently functionalized with several short peptide motifs derived from FN, laminin, and collagen [92]. The carboxylic acid groups already present in HA have also been used to immobilize rat tail collagen I [96]. As examples of the second strategy, poly(vinylalcohol) (PVA) hydrogels have been carboxylate functionalized with 11-bromoundecanoicacid, activated to yield NHS-esters, and subsequently conjugated with FN [97]. The amine-reactive photo-linkable crosslinker sulfo-SANPAH has also been widely used to activate hydrogels, especially defined stiffness polyacrylamide hydrogels, for immobilization of ECM proteins through lysine residues [96,98]. Sulfo-SANPAH has also been used to conjugate laminin to methylcellulose hydrogels [99].

As an alternative to directly reacting the amine groups of the target protein to the polymer backbone, these amines can instead be conjugated to a small molecule that can react/interact with chemical functionalities available on the polymer backbone. For example, proteins have frequently been modified with acrylate groups, which allow these proteins to be co-photopolymerized with synthetic polymers containing acrylate groups to create bioactive hydrogels. This strategy has been used to conjugate FN to hydrogels made from PEG [100] and HA [101], thus demonstrating the flexibility of its application. Acryl-PEG-NHS has also been used to modify integrin-binding peptides such as RGD and YIGSR [102,103], as well as a variety of growth factors including fibroblast growth factor-2 (FGF-2, also known as basic FGF) [94] and platelet-derived growth factor-BB (PDGF-BB) [104], in order to facilitate the incorporation of these bioactive species into PEGDA hydrogels. Similarly, lysine residues in epidermal growth factor (EGF) have been functionalized with iodoacetamide-NHS to enable conjugation of this growth factor on HA hydrogels carrying thiol groups [105]. Another secondary functionality that enables high-affinity noncovalent conjugation is biotin, which can be used to link the modified biomolecule to avidin-functionalized hydrogels [106108].

Although the abundance of lysines in proteins makes lysine an attractive residue to use for immobilization, it is challenging if not impossible to precisely control which lysine (or lysines) in a given protein reacts with the hydrogel matrix. This leads to an inhomogeneous distribution of protein orientation, which could lead to some fraction of the immobilized protein molecules to be denatured or sterically shielded from their interaction partners on the cell surface. Furthermore, conjugation of functionally critical lysine residues could compromise or eliminate the bioactivity of the protein. Therefore, cysteine residues, which are significantly rarer in proteins, can be used for more selective immobilization. Using recombinant DNA technology, cysteines can also be introduced into target proteins at precisely specified positions along the polypeptide chain, which enables a high degree of control over the orientation of immobilization. The glutathione S-transferase fusion tag which is often used for affinity purification of recombinantly expressed proteins also contains cysteine residues that have been used for immobilization on thiol-reactive matrices [109].

Thiols from cysteine residues can take part in several reactions that have been demonstrated to be very useful for biofunctionalization of hydrogels. One of the most widely used strategies is the Michael addition reaction in which a thiol reacts with an alkene, often in the context of an acrylate or methacrylate moiety. This chemistry has been used to attach FN to PEGDA, which was then photo-crosslinked to yield a hydrogel network [109]. A similar scheme has also been used to first generate FN-functionalized PEGDA and then crosslink thiolated-HA into the PEG network to create a three-component hydrogel [93]. A similar strategy has also been used to fabricate injectable HA hydrogels decorated with RGD peptide for the in vivo delivery of encapsulated cells [110]. In this study, fibroblasts were added to a mixture of thiolated-HA and RGD-modified PEGDA. The resulting mixture was then subcutaneously injected into the flanks of nude mice, leading to the formation of hydrogels in vivo. In a strategy employing the same chemistry but in an opposite topology, HA has been methacrylated using methacrylic anhydride and subsequently reacted with RGD peptides containing a cysteine residue [75,111]. PEGDA has also been conjugated with a thiolated transforming growth factor-β (TGF-β), which enabled the fabrication of TGF-β-functionalized PEG hydrogels [112]. Vinylsulfone moieties, which react more selectively and rapidly with thiols [113], have also been used for functionalization of hydrogels. PEG macromers carrying vinylsulfone terminal groups have been reacted with cysteine-terminated RGD and other peptides to introduce integrin-binding sites in hydrogels [114]. The same reaction has also been utilized to incorporate vascular endothelial growth factor (VEGF) carrying a genetically engineered cysteine residue into PEG hydrogels [115]. Thiol groups can also react with maleimides in a variation of the Michael addition reaction which proceeds with significantly faster kinetics [116]. This reaction has been extensively used for immobilizing peptides derived from ECM proteins onto PEG-based hydrogels [117]. Maleimide-terminated multiarm PEGs (often referred to as star-PEGs) have been modified with RGD and FN through cysteine residues. PEG arms with unreacted maleimide groups could be subsequently reacted with thiol modified PEGs thus crosslinking the polymers into a hydrogel [118]. Heparin functionalized with maleimide groups could also be incorporated into such hydrogels to enhance the bioactivity of the hydrogel system and endow it with growth-factor-binding properties [24]. PEG-maleimide has also been reacted with VEGF in order to allow its incorporation in PEG hydrogels [95]. Another fast and thiol-specific reaction that may be carried out in the presence of living cells is the photoactivated thiolene reaction [119], which has been utilized for immobilization of FN- and laminin-derived peptides on PEG hydrogels [120]. A major advantage of this technique over the earlier strategies is its photosensitive nature, which makes it amenable for spatial patterning of peptides and proteins on hydrogels. Indeed, 3D micropatterning of RGD peptides on PEG-based hydrogels has been achieved using this chemistry [121]. In an elegant approach utilizing click chemistry for hydrogel crosslinking and the thiolene reaction for peptide immobilization, PEG hydrogels with independently controllable mechanical and biochemical parameters could be fabricated [122].

Use of reactive amine and thiol groups already present in the protein sequence is a very attractive approach due to its flexibility and ease of application. However, in addition to the limitations described earlier, a shortcoming of these strategies is their lack of selectivity against cell- and medium-derived proteins, which might themselves couple to the polymer backbone. In addition to compromising yield by consuming reactive sites, this could confound studies that seek to delineate the effect of specific ECM proteins or derived peptides upon cellular processes. Therefore, several methods have been established that enable the selective immobilization of target proteins of interest onto hydrogel platforms, some of which are covered below.

14.2.2 Immobilization of Growth Factors Through Interactions with Other Biomolecules

Growth factors interact with several components of the ECM. In this section we describe ECM proteins and GAGs that exhibit such interactions and hydrogel systems that have taken advantage of some of these interactions to entrap cell-derived factors (Figure 14.2).

As described earlier, the interactions between GAGs and growth factors have been extensively characterized and leveraged for biomaterial design. GAG–protein interactions are largely based on electrostatic attraction between cationic residues on the proteins and anionic sulfate and carboxyl groups on the GAGs, although hydrogen bonding and van der Waals interaction are also believed to contribute [123,124]. Through these interactions GAGs are thought to sequester growth factors, providing a depot effect that regulates these proteins’ local concentration, diffusion, and signaling intensity [125]. Moreover, the association of growth factors with heparin and HS has been shown to increase growth factor lifetime by protecting these proteins from enzymatic degradation [126,127].

Among the GAGs, heparin and HS are probably the most extensively studied molecules based on their interaction with diverse growth factors such as FGFs, TGF-β, VEGF, and PDGF-A and -B [24] (Table 14.1). FGFs are among the best-studied heparin-binding proteins, and structural studies have shown that heparin and HS interact with both FGF and its receptor, while simultaneously promoting receptor dimerization [128,129]. Hydrogels based on heparin and HS have therefore found extensive use as platforms for sustained release of growth factors. A heparin-crosslinked PEG-based hydrogel loaded with VEGF demonstrated gradual release of active VEGF over a period of 3 weeks [130]. Subcutaneous implantation of these hydrogels in mice was shown to induce angiogenesis around the implantation area. Taking this a step further, a similar strategy was used to enable the simultaneous sustained delivery of VEGF and FGF-2, which demonstrated superior pro-angiogenic activity over administration of single growth factors in both in vitro and in vivo models [131]. PEG-heparin hybrid hydrogels loaded with hepatocyte growth factor (HGF) were shown to not only support the culture of primary rat hepatocytes but also promote urea and albumin synthesis, which has motivated the application of this platform for in vitro differentiation of hepatocytes and as vehicles for transplantation [132]. Heparin has also been combined with natural polymers in order to synthesize sustained growth factor-releasing hydrogels. In a related study, heparin was incorporated into chemically crosslinked alginate hydrogels and used to entrap FGF-2 [133]. Here, regenerated axons from rat sciatic nerves grew much faster on heparin-alginate hydrogels than on alginate-only hydrogels, suggesting that heparin promoted retention and delivery of the growth factor. In another study, heparin was incorporated into photo-crosslinked alginate hydrogels and the hydrogels were loaded with bone morphogenetic protein-2 (BMP-2). It was demonstrated that BMP-2-loaded photo-crosslinked heparin-alginate hydrogels induced significantly more osteogenesis than unmodified alginate hydrogels loaded with BMP-2 [25]. Hydrogels formed from HA have also been functionalized with heparin in order to achieve controlled release of BMP-2 [26]. GAGs other than heparin are known to interact with specific growth factors [134136], but these materials have not yet been as extensively explored as matrices for controlled release.

Table 14.1

Interactions Between Growth Factors and ECM Components

ECM Component Interacting GF
Heparin/heparan sulfate FGFs [24,128], TGF-βs [283], VEGF [284,285], PDGFs [286,287], HGF [136,288], BMPs [289291]
Collagens PDGFs [151], HGF [152], TGF-β1 [153]
Vitronectin IGF-II [154], TGF-β1 and 2, EGF, VEGF, FGF-2 [156], HGF [141]
Fibronectin HGF [141], TGF-β1 [140], VEGF [137,144,145], CTGF [142], FGFs [137,146], PDGFs [137,143], PIGF-2, 3, BMP-2, 7, NGF, BDNF [137]
Fibrinogen VEGF [138,149], FGFs [138,148], PIGF-2,3, PDGFs, BMP-2,2/7, TGF-β1,2, BDNF [138]
Tenascin-C PDGFs, VEGF, PIGF-2,3, FGFs, TGF-β1,2, BMP-2, BDNF, HGF [150]

Growth factors have also been shown to interact with ECM protein components. Recently, several ECM proteins such as FN, fibrinogen, and tenascin-C were shown to bind a large number of diverse growth factors (Table 14.1) [137,138]. The regions of these proteins responsible for binding growth factors were localized to their heparin-binding motifs, although binding surprisingly does not require the presence of heparin. Interestingly, mutation of the positively charged residues in these motifs into serines abolishes growth factor binding indicating that the interactions may be driven by electrostatic forces.

FN is an important component of the ECM in wounds and its interaction with growth factors is believed to be important for wound healing [139]. FN is known to interact with TGF-β1, HGF, connective tissue growth factor (CTGF), PDGF-A, and VEGF with high affinity [140144]. Using recombinant FN domains, the C-terminal heparin-binding Hep-II domain of FN (consisting of type III repeats 13 to 14) was identified as the key site for binding VEGF and FGF-2 [145,146]. Interestingly, peptide fragments containing both the α5β1 integrin-binding domain (FN type III repeats 9 to 10) and VEGF-binding Hep-II domain significantly enhanced VEGF-induced cell responses, suggesting strong crosstalk between bound integrins and the VEGF receptor mediated by FN and possibly by other ECM-derived proteins [145]. In a high-throughput study, it was observed that the heparin-binding FN III12-14 domain binds most of the growth factors from the PDGF, VEGF, and FGF families, and some growth factors from the TGF-β and neurotrophin families with nanomolar affinity and without inhibiting growth factor activity [137]. A total of 25 previously unknown interactions with growth factors were identified in this report. Growth factor binding was enhanced in the presence of heparin, but the extent of enhancement was variable.

The therapeutic implications of this discovery were clearly demonstrated in a study in which the multiple interactions of this domain were utilized to sequester a variety of growth factors in a fibrin-based hydrogel [147]. In this study, a fusion peptide consisting of the integrin-binding FN III9-10 domain and the growth factor-binding FN III12-14 domain previously shown to have a synergistic effect on enhancing VEGF-induced cell signaling [145] was used to functionalize fibrin hydrogels. These hydrogels were shown to sequester VEGF-A165, PDGF-BB, and BMP-2, and enhanced the morphogenetic effects of these growth factors in vitro. Significantly, it was further demonstrated that the FN III9-10/12-14 peptide greatly enhanced the regenerative effects of the growth factors in vivo in a diabetic mouse model of chronic wounds and in a rat model of critical-size bone defects at doses where the growth factors delivered within fibrin only had no significant effect (Figure 14.3).

image
Figure 14.3 Enhancement of skin wound healing in diabetic mice and bone regeneration in rats by incorporation of growth factor-binding recombinant FN domains into fibrin hydrogels. (A) Histological examination of wound tissue in diabetic mice 10 days after implantation of the specified fibrin matrices. Much higher wound closure is evident for the fibrin matrices functionalized with FN III9-10/12-14 containing growth factors VEGF-A165 and PDGF-BB. Scale bar is 1 mm. (B) Quantitative analysis of the wound closure and granulation area from histological examination. (C) Representative micro-computed tomography (μCT) images of the skull of rats with critical-size calvarial defects 4 weeks after implantation of the specified fibrin matrices. The growth factors used in this study were BMP-2 and PDGF-BB. FN III9*-10/12-14 contains a point mutation in the 9th repeat which makes it specific to the α5β1 integrin, the engagement of which is known to enhance osteoblastic differentiation of MSCs. (D) Quantitative analysis of the μCT data for implantation of four different fibrin matrices. Source: From Ref. [147]. Reprinted with permission from AAAS.

Another protein system that has been extensively studied for its interactions with growth factors is fibrinogen and its enzymatic degradation product fibrin. Fibrinogen is found in blood plasma and plays an important function in clotting by acting as a scaffold for platelet adhesion and by promoting endothelial cell mitogenesis [139]. FGF-2, which is released by disrupted blood vessels during wound formation, can bind both fibrin and fibrinogen [148], and this sequestration is necessary for endothelial cell migration, mitogenesis, and ultimately angiogenesis during wound repair. VEGF, another growth factor involved in wound healing, also interacts with both fibrinogen and fibrin [149]. A recent high-throughput study identified 15 unique interactions between the ECM proteins fibrin and fibrinogen and growth factors from the PDGF, VEGF, FGF, TGF-β, and neurotrophin families [138]. This study then demonstrated that similar binding characteristics are shown by a dimer of the heparin-binding domain of fibrinogen. Significantly, binding with this domain did not alter the ability of growth factors to trigger cell proliferation. The researchers then incorporated this domain into a synthetic PEG-based hydrogel and showed that the domain effectively sequestered FGF-2 and placental growth factor-2 (PlGF-2) (Figure 14.4). Furthermore, this synthetic bioactive hydrogel could fully recapitulate the efficacy of a fibrin matrix in promoting wound healing in a mouse model.

image
Figure 14.4 Entrapment of growth factors and enhancement of wound healing in diabetic mice by a fibrin-mimetic PEG matrix. (A) Schematic representation of the components for the fibrin-mimetic PEG hydrogel. The matrix contains eight-arm PEG molecules conjugated to two peptide sequences: (i) α2PI1–8 peptide, and (ii) a protease-sensitive sequence fused to the FKGG peptide, which act as substrates for the transglutaminase FXIIIa. RGD fused to α2PI1–8, the growth factor binding fibrinogen domain fused to α2PI1–8, and growth factors are added to the PEG formulation prior to cross-linking. The transglutamination reaction catalyzed by FXIIIa forms isopeptide bonds between a glutamine residue in α2PI1–8 and the lysine in FKGG, thus crosslinking the PEG macromers and also conjugating the RGD peptide and the fibrinogen domain to the matrix. (B) Retention of fibrin-binding growth factors in fibrin-mimetic PEG matrix functionalized with the growth-factor-binding domain of fibrinogen. PIGF-2 and FGF-2, which interact with this domain, are retained in the hydrogel only in the presence of the domain. (C) Representative histological specimens of the wound area 10 days after implantation of the specified matrices in diabetic mice. Delivering growth factors within the PEG matrix functionalized with α2PI1–8-Fg β15–66(2) enhances wound healing to an extent comparable with a fibrin matrix loaded with the growth factors. Scale bar is 1 mm. Source: Adapted with permission from Ref. [138].

Similar promiscuous growth factor-binding behavior was recently demonstrated for the heparin-binding domain of Tenascin-C (TNC) [150]. In this study, recombinant peptide fragments of TNC representing the first five TNCIII repeats (TNCIII1–5) were used to study interactions with various growth factors. Multiple growth factors of the PDGF, FGF, and TGF-β families were found to interact with this region of TNC, specifically to TNCIII5.

Collagens I–VI have been shown to bind PDGF-AA, BB, and AB with high affinity without affecting the activity of these growth factors [151]. Collagens can also bind HGF with nanomolar affinity, and HGF bound to collagen has been shown to retain its pro-motility and proliferative functions in culture [152]. Similarly, collagen was also shown to bind TGF-β [153].

Finally, insulin-like growth factor-II (IGF-II) is known to interact with vitronectin [154]. It was shown that IGF-II bound to vitronectin significantly enhanced migration of breast cancer cells in transwell assays, while unbound IGF-II did not [155]. Vitronectin is also known to interact with a variety of other growth factors such as TGF-β, EGF, FGF-2, VEGF, and HGF [141,156].

Thus, intrinsic interactions between growth factors and various components of the ECM can be utilized to entrap these factors in hydrogels based on both natural and synthetic polymers, with retention of biological activity. Furthermore, the typically reversible nature of the scaffold–factor interaction makes such strategies very attractive for therapeutic applications requiring sustained in vivo delivery of growth factors. A major shortcoming of such approaches is that they are not universally generalizable but are instead limited to specific growth factors with high affinity for a given scaffold. The following sections describe some strategies that can be generically applied for the immobilization of any target protein on hydrogels.

14.2.3 Protein/Peptide Tags for Immobilization

Recombinant DNA technology has been used to tag growth factors and ECM-derived proteins with peptides that enable their specific immobilization onto hydrogels (Figure 14.2). In this way, amino acids in the native sequence that may be critical to function are spared from chemical functionalization. Furthermore, placement of the tag in a known position relative to the bioactive portion(s) of the protein increases the likelihood that protein orientation is homogeneous. Such strategies are also more generic in that most proteins can be recombinantly fused to such tags without affecting their native function as well as expressed in material-scale quantities using bacterial, yeast, and insect cell expression systems.

14.2.3.1 Collagen-Binding Domain-Mediated Immobilization

One of the most common families of peptide tags that have been used for targeted protein immobilization on hydrogels are derived from the collagen-binding domains (CBDs) of proteins. Such domains have been obtained from four sources—the von Willebrand factor (vWF), mammalian collagenase, bacterial collagenase, and FN [157].

The CBDs derived from vWF and human collagenase are ten and seven residues long, respectively. Due to their short lengths, these peptides are preferred fusion tags for growth factors in order to impart them with collagen-binding properties. The CBD derived from vWF has been fused to several growth factors such as TGF-β [158,159], EGF [160], FGF-2 [161], BMP-2 [162,163], and BMP-3 [164]. In vitro experiments demonstrated that collagen matrices functionalized with the TGF-β-CBD fusion protein, but not commercial TGF-β, could promote the survival, proliferation, differentiation, and colony mineralization of osteogenic precursor cells present in rat bone marrow [159]. In an in vivo rat ectopic bone formation assay, implantation of collagen hydrogels functionalized with BMP-2 fused to this CBD induced the production of new bone tissue inside the implants [163]. These tissues displayed the hallmarks of mature bone such as trabecular morphology and medullary cavities. CBD-BMP-3 fusion proteins affixed to collagen hydrogels also stimulated ingrowth of new bone in a rat cranial defect model, underlining the high therapeutic potential of such growth factor fusion constructs [164]. The CBD derived from human collagenase has also been fused to a large number of growth factors in order to endow them with collagen-binding properties. Both in vitro and in vivo experiments using collagen scaffolds functionalized with growth factors such as PDGF-BB [165,166], nerve growth factor-β (NGF-β) [167], neurotrophin-3 [168], VEGF [169,170], EGF [171], and FGF-2 [172] fused to this CBD have demonstrated the great potential of this strategy for tissue engineering and medical applications. A comparison of the fusion tags derived from vWF and human collagenase showed that the latter enhanced collagen affinity. This also translated to in vivo activity, as collagen scaffolds functionalized with FGF-2 fused to the collagenase-derived peptide were able to stimulate greater vascularization upon subcutaneous implantation in rats [172]. The CBD of bacterial collagenase and FN are comparatively larger proteins (24 and 27 kDa respectively) and can thus affect the functionality of growth factors fused to them. Despite this, the bacterial collagenase-derived domain has been fused with FGF-1 [173,174], FGF-2 [175], EGF [175], and VEGF [176], while the CBD of FN has been combined with HGF [177], BMP-4 [178], EGF [179], and VEGF [180] to endow these growth factors with collagen-binding ability. The BMP-4 fusion protein was strongly osteoinductive in 3D cultures of human bone-marrow-derived mesenchymal stem cells in a hybrid scaffold based on collagen and poly(lactic-co-glycolic acid) [178]. In vivo implantation of such scaffolds demonstrated that the immobilized CBD-BMP4 retained its osteoinductive activity, as evidenced by its ability to upregulate osteogenic gene expression and biomineralization.

14.2.3.2 Factor XIIIA Transglutaminase Catalyzed Immobilization

During wound healing, fibrin clots are stabilized by the transglutaminase factor XIIIA (FXIIIa), which crosslinks glutamine residues on one fibrin molecule to lysine residues on another fibrin molecule through isopeptide bonds. This reaction is also used in the in vitro fabrication of fibrin hydrogels and has been extended to the conjugation of target proteins and peptides to such hydrogels [181183]. This strategy is based on a peptide derived from the 12 N-terminal residues of α2-plasmin inhibitor (α2-PI), which was found to be a good substrate for FXIIIa and could be readily crosslinked with fibrin via the transglutamination reaction catalyzed by FXIIIa [184]. Thus, proteins or peptides fused to this FXIIIA substrate peptide could be covalently conjugated onto fibrin matrices as demonstrated for two integrin-binding peptides FN-derived RGD and collagen-derived DGEA [185]. This α2-PI-derived peptide domain was also fused with heparin biding peptides, which were then immobilized on fibrin scaffolds in order to enable the incorporation of heparin into the hydrogels [181]. This strategy was then utilized to sequester the heparin interacting growth factor, FGF-2. It was demonstrated that FGF-2 immobilized within heparin-modified fibrin enhanced neurite extension by up to 100% relative to unmodified fibrin [182]. The promiscuous growth factor-binding domain of FN was also immobilized onto fibrin hydrogels using this reaction [137,147].

Growth factors can also be recombinantly fused to the α2-PI-derived peptide sequence to allow immobilization onto fibrin matrices (Table 14.2). This was first demonstrated for NGF-β [186]. A peptide sequence that could be cleaved by the cell-secreted protease plasmin was included between the α2-PI-derived peptide and NGF-β to ensure that the growth factor, which was irreversibly immobilized onto the matrix, would be released and be able to stimulate signaling only in the vicinity of plasmin-secreting invading cells. This NGF fusion protein immobilized onto a fibrin matrix enhanced neurite extension from embryonic chick dorsal root ganglia by 50% relative to soluble NGF. Other growth factors that have been recombinantly fused to the α2-PI-derived peptide in order to enable incorporation into fibrin hydrogels include VEGF [183,187], BMP-2 [188], and IGF-1 [189]. For growth factors not amenable to recombinant expression, the peptide sequence can also be covalently reacted with surface lysines as has been done for keratinocyte growth factor [190].

Table 14.2

Protein and Peptide Tags That Can Be Used for Site-Specific Immobilization of Target Fusion Proteins/Peptides

Scaffold Material Source and Size of Tag Fused Protein/Peptide
Collagen vWF, 10 residues TGF-β [158,159], EGF [160], FGF-2 [161,172], BMP-2 [162,163], BMP-3 [164]
Human collagenase, 7 residues PDGF-BB [165,166], NGF-β [167], Neurotrophin-3 [168], VEGF [169,170], EGF [171], FGF-2 [172]
Bacterial collagenase, 24 kDa FGF-1 [173,174], FGF-2 [175], EGF [175], VEGF [176]
Fibronectin, 27 kDa/40 kDa HGF [177], BMP-4 [178], EGF [179], VEGF [180]
Fibrin, PEG conjugated to FXIIIA substrate peptide α2-plasmin inhibitor, 12 residues Cell-adhesive peptides [185,193], heparin-binding peptides [181], GF-binding FN domains [137,147], GF-binding fibrinogen domain [138], NGF-β [186], VEGF [115,183,187,193], BMP-2 [188], IGF-1 [189]
HA-MC functionalized with SH3-binding peptides SH3 domains, 60–70 residues FGF-2 [195]
Agarose, chitosan, HA-MC. All functionalized with avidin/streptavidin Screen of peptide libraries, 15 residues (Avitag™) CNTF [199], interferon-γ [200], PDGF-A [201]
Agarose functionalized with barnase Barstar, 89 residues SHH [199]

Image

Notes: (I) Not all listed proteins with CBD fusion have been immobilized on hydrogels. Relevant proteins that have been successfully expressed and purified in bioactive collagen-binding forms have been included and are expected to bind to collagen scaffolds.
(II) The matrix material is not limiting for the other tags as long as they are modified with the interacting peptide/protein tag.

Interestingly, this enzymatic immobilization strategy has been now extended to enable functionalization of synthetic PEG-based matrices as well (Table 14.2). In this approach, multiarm PEG molecules are functionalized with either the α2-PI-derived peptide (acceptor peptide) or with the sequence FKGG (donor peptide), which was previously optimized as an efficient substrate for the transglutamination reaction catalyzed by FXIIIa [191]. Cross-linking of these two PEG formulations by FXIIIa yields a hydrogel, with inclusion of peptides or proteins fused to the acceptor peptide sequence during the crosslinking reaction incorporating these sequences into the hydrogel. Using this strategy, peptides containing the RGD sequence were immobilized onto PEG hydrogels which allowed mammalian cells to adhere and proliferate on the hydrogels [192]. The heparin-binding domain of fibrinogen, which is known to bind several growth factors, was also incorporated into PEG hydrogels using this technique in order to get fibrin-mimetic synthetic hydrogels which efficiently retained growth factors that showed interactions with the fibrinogen domain [138] (Figure 14.4). Such hydrogels could be efficiently loaded with the pro-angiogenic growth factors, FGF-2 and PIGF-2, and could fully mimic the effect of fibrin in promoting wound repair in a diabetic mouse model of impaired wound healing. Recombinant VEGF was also quantitatively incorporated into PEG-based hydrogels using this strategy [193].

14.2.3.3 Src Homology 3 Domain-Mediated Immobilization

Src homology 3 (SH3) domains are independently folding 60–70-residue peptide domains that bind polyproline sequences in proteins [194]. This interaction was used to immobilize FGF-2 fused to an SH3 domain on hyaluronic acid-methyl cellulose (HA-MC) composite hydrogels functionalized with SH3-binding polyproline peptide sequences [195]. Notably, this study also showed that hydrogels functionalized with two different polyproline peptide sequences having distinct affinities to the SH3 domain showed markedly different release profiles of FGF-2, which demonstrated the utility of this strategy to enable tunable release of growth factors and other protein drugs for therapeutic applications.

14.2.3.4 Enzymatic Biotinylation of Proteins for Immobilization

The Escherichia coli biotin ligase BirA catalyzes the transfer of biotin to the amino group of a lysine residue of the biotin carboxyl carrier protein subunit of acetyl-CoA carboxylase. Screening of peptide libraries has led to the discovery of a 14-residue peptide sequence that can be biotinylated by BirA at rates similar to the native substrate [196]. This sequence has since been modified by the addition of a C-terminal glutamate residue and termed the Avitag™ system. Proteins recombinantly fused to this peptide can be site-specifically biotinylated and then modified with streptavidin. This strategy has been used for conjugating recombinant proteins with fluorescent probes [197] and nanoparticles [198]. Recently, this enzymatic reaction was utilized for immobilization of recombinantly expressed growth factors and cytokines on hydrogels as well. In one report, thiolated-agarose hydrogels were first reacted with maleimide-functionalized streptavidin, which was subsequently used to capture ciliary neurotrophic factor (CNTF) fused to the AviTag that was biotinylated prior to immobilization [199]. In another paper, the pro-neuronal differentiation factor interferon-γ was recombinantly fused to the AviTag™ and immobilized on streptavidin-modified chitosan hydrogels [200]. Neural stem/progenitor cells (NSPCs) cultured on these matrices showed preferential differentiation into neurons confirming the retention of bioactivity of the cytokine even after immobilization. In order to promote differentiation of NSPCs into oligodendrocytes, the oligodendrocyte-differentiating factor PDGF-A was biotinylated using this reaction and then immobilized on hybrid HA-MC hydrogels that were previously modified with streptavidin [201]. As expected, rat NSPCs differentiated into significantly more oligodendrocytes when cultured in such hydrogels.

14.2.3.5 Barstar-Mediated Immobilization

The bacterial ribonuclease barnase and its inhibitor barstar bind each other with extremely high affinity [202,203]. This strong binding has been utilized to immobilize the stem cell differentiation factor sonic hedgehog (SHH) recombinantly fused to barstar on barnase-functionalized hydrogels [199]. In this work, photosensitive caging of thiol groups on agarose hydrogels was used to pattern SHH and CNTF with a very high spatial resolution. Here, two-photon irradiation was used to uncage thiol groups in selected regions of the hydrogel followed by immobilization of barnase modified with maleimide. Next, thiol groups in another region were uncaged and reacted with streptavidin conjugated to maleimide groups. Upon immersing the hydrogel in a solution of SHH-barstar and biotin-CNTF, the proteins were independently immobilized into the target regions (Figure 14.5). The high binding affinity of both interactions ensured that binding was almost irreversible. Mouse retinal precursor cells cultured in these matrices expressed downstream effectors of both SHH and CNTF signaling pathways, indicating that the immobilized proteins retained their activity.

image
Figure 14.5 3D patterning of SHH and CNTF in agarose hydrogels. (A) Scheme for sequential patterning of maleimide-barnase and maleimide-streptavidin on agarose hydrogels. (B) A confocal micrograph demonstrating the localization of barstar-SHH-488 (green) and biotin-CNTF-633 (red) to the volumes patterned. (C) 3D projection of a reconstructed stack demonstrating 3D patterning capability. Scale bar is 100 μm. Source: Adapted from Ref [199] with permission from Macmillan Publishers Ltd.

14.2.4 Aptamer-Mediated Immobilization

Aptamers are single stranded DNA or RNA sequences that bind with target proteins with a specificity and affinity comparable to that of antibodies [204]. These sequences are most often selected in vitro from a pool of random oligonucleotides, and selection conditions can be manipulated to obtain aptamers with different specificities and affinities [205]. Aptamers have been identified that bind a variety of proteins including ECM components [206209] and growth factors [210225] (Table 14.3).

Table 14.3

Growth Factor, Cytokine, and ECM Protein Targets of Aptamers

Target Affinity (nM)
VEGF [216] 0.14
PDGF-AB, PDGF-BB [210] 0.1
FGF-2 [212] 0.13
IGF-II [217] 2
TGF-β1 [218] 90
TGF-β2 [219] 5
KGF [214] 0.0003
Angiopoietin-1 [220] 2.8
Angiopoietin-2 [221] 2.2
Erythropoietin-α [225] 33
TNF-α [223] 7
Interferon-γ [224] 1.8
Interleukin-17A [225] 0.05
Tenascin-C [208] 5
Osteopontin [209] 18
Fibronectin [206] Not reported

Taking advantage of the enormous flexibility in designing and synthesizing aptamers, researchers have functionalized synthetic hydrogels with aptamers having different affinities to PDGF-BB to enable sustained release of the growth factor with a high degree of control over the release kinetics (Figure 14.2) [226,227]. Another special feature of aptamer-based protein conjugation is that binding may be readily reversed by introducing the oligonucleotide sequence complementary to the aptamer. This feature was elegantly utilized to achieve specific and triggered release of multiple growth factors from a microparticle-hydrogel hybrid system [228]. Here, polystyrene microparticles were functionalized with aptamers directed against VEGF and PDGF-BB. The microparticles were then loaded with the growth factors and incorporated into hydrogels. Addition of the specific complementary sequences led to the independent release of the target growth factor without any significant effect on the release of the other growth factor. The high degree of control coupled with the low immunogenicity and toxicity of oligonucleotides makes aptamer-functionalized hydrogels a very attractive targeted drug delivery system.

Aptamers can also be selected to engage specific cell types [229]. This is based on cell-specific variations in surface proteins, including receptors and other transmembrane proteins. For example, aptamers that specifically bind to leukemia cells with sub-nanomolar affinity were recently generated and conjugated to PEG hydrogels to facilitate separation and capture of leukemic cells (Figure 14.6) [230]. Adherent cells could also be selectively released under biocompatible conditions by the addition of an endonuclease that cleaved the aptamers [231] or of an oligonucleotide sequence complementary to the aptamer [232].

image
Figure 14.6 Cell type-specific adhesion on PEG hydrogels using aptamers. (A) Flow cytometry analysis demonstrating selective binding of the aptamer Sgc8c (fluorescently labeled) to the human leukemic cell line CCRF-CEM. In contrast, the aptamer does not bind to the nonleukemic cell line Ramos. (B) Microscopy images of PEG hydrogels functionalized with either Sgc8c or a control aptamer establishes that only CCRF-CEM cells adhere to the hydrogels. (C) A quantitative analysis of the images in (B). Source: Reprinted from Ref. [230], with permission from Elsevier.

14.3 Engineering Degradability into Hydrogels

Degradation of hydrogels is essential for many purposes. In tissue engineering applications, degradation is required to create space in the matrix for cell migration and proliferation as well as to allow infiltration of blood vessels. When hydrogels are used for in vivo delivery of drugs, degradation is important for both drug release and removal of the hydrogel scaffold from the body. Importantly, the optimal degradation rate may vary significantly from one application to another, creating the need for tunable degradation kinetics. Moreover, as previously mentioned, the ECM is continuously remodeled by the action of cell-secreted enzymes, so for applications involving tissue regeneration, hydrogels must be amenable to this cell-directed degradation, with the rate of degradation ideally mirroring the rate of new tissue outgrowth. For applications requiring controlled release of therapeutic molecules, the kinetics of degradation should be adequate to maintain effective dosages of the bioactive molecule. A major consideration for hydrogels intended for in vivo implantation is that the degradation products should be nontoxic and amenable to clearance from the body. Major mechanisms of hydrogel degradation that are currently used include hydrolysis and enzymatic degradation.

14.3.1 Hydrolytic Degradation of Hydrogels

Hydrolysis causes homogenous degradation of hydrogels carrying hydrolytically sensitive macromers and crosslinkers. This leads to changes in the overall network properties and can significantly influence the diffusivity of entrapped drugs and proteins [233]. Hydrolytic degradation is spontaneous and is often preferred over enzymatic degradation in drug delivery applications, where patient-to-patient variability in the levels of circulating enzymes can lead to unpredictability in the release of drugs from enzymatically degradable hydrogels [234236].

Hydrolytically degradable hydrogels most commonly utilize the hydrolysis of ester linkages. This mechanism has been applied for fabrication of both natural and synthetic polymer-based matrices. HA functionalized with hydrolysis-sensitive poly(lactic acid)-methacrylate (PLA-MA) has been crosslinked using the polymerization reaction of methacrylate to fabricate hydrolytically degradable hydrogels that allowed controlled release of VEGF [237]. HA has also been crosslinked with other hydrolyzable crosslinkers such as glycidyl methacrylate [238] and PEGDA [96] in order to make hydrogels susceptible to hydrolysis. Hydrogels based on synthetic polymers such as PEG have also been made sensitive to hydrolysis by incorporating hydrolyzable polymers like poly(lactic acid) (PLA) and poly(glycolic acid) into the scaffolds. The most notable example of such networks has been made using triblock polymers of the form PLA–PEG–PLA with acrylate end groups for crosslinking [81]. The ratio of PEG to PLA segments can be used to manipulate the degradation rate as well as permeability [81,239]. Degradable PEG hydrogels have also been fabricated by reacting an ester containing, amine-reactive PEG derivative with a branched PEG amine [240]. Proteins could be covalently immobilized onto this completely hydrophilic hydrogel via reaction of lysine residues with the amine-reactive groups. Multiarm PEGs terminated with hydrolyzable acrylate moieties have also been crosslinked with a PEG dithiol using the Michael addition reaction [241]. Multiarm PEGs terminated with vinylsulfone have also been often used to form hydrogels [242]. Unlike the acrylate group used previously, the vinylsulfone moiety is not hydrolyzable, and so dithiol crosslinkers containing ester groups have been used in order to render these hydrogels susceptible to hydrolytic degradation. It has been further demonstrated that the hydrolytic degradation rates of such PEG-based synthetic hydrogels can be independently modulated without affecting other physical properties. This was first accomplished by using acrylate-terminated PEG macromers incorporated with ester linkages with variable alkyl chain length (acetyl, propionyl, and butyryl esters) [243]. While the mechanical properties of the three formulations were very similar, the degradation rates were significantly higher for shorter alkyl chain lengths. Hydrolysis rates of PEG-based hydrogels have also been controlled by varying the charge of the cross-linker [244]. In this study, acrylate-terminated multiarm PEG was crosslinked using peptides containing either arginine (positively charged) or aspartate (negatively charged) residues. The arginine-based gels underwent hydrolysis 12 times faster than the aspartate-based gels.

14.3.2 Enzymatic Degradation of Hydrogels

Degradation of hydrogels by cell-secreted enzymes is often desirable in tissue engineering systems where there is an interest in locally restricting degradation to regions of cell invasion and coupling the rate of degradation with the rate of tissue formation.

Hydrogels based on natural polymers are sensitive to several cell-secreted enzymes; for example, collagen is degraded by collagenases MMP-1 and MMP-8 [245]. Therefore, hydrogels combining collagen with synthetic polymers such as PEG are also susceptible to cell-mediated degradation [246]. Fibrin hydrogels can be degraded by the action of cell-secreted MMPs and plasmin [247]. HA can be degraded by hyaluronidases [248] and the degradation rate of HA hydrogels can be controlled by covalently modifying the carboxylic groups of the macromer prior to gelation [249]. Hydrogels based on heparin can be degraded by the action of heparanase [250].

Hydrogels based on synthetic polymers can be made susceptible to enzymatic degradation as well. This has been accomplished by both functionalizing constituent macromers with enzymatic substrates or using crosslinkers consisting of peptide sequences that contain substrates for enzymes such as MMPs. These strategies have been successfully applied for creating degradable hydrogels using PEG molecules incorporating enzymatic substrates at both termini [251]. These hydrogels were crosslinked through photopolymerization of acrylate moieties at the ends of the enzyme substrate peptides, including substrates for the collagenase MMP-1 and plasmin. In another study, inclusion of an elastase-sensitive peptide in a similar model made the hydrogel sensitive to this enzyme [252]. The bioactivity of the hydrogels was further enhanced by including PEG macromers containing the cell-binding peptide KQAGDV into the polymerization reaction. This led to the formation of hydrogels with cell adhesive function and enzymatic degradability. Smooth muscle cells (SMCs), which are known to secrete elastase during migration, could be viably encapsulated in the hydrogels during polymerization. It was demonstrated that the SMCs produced more collagen in the degradable hydrogels than in nondegradable hydrogels. Similar methodologies have also been applied to multiarm PEGs, where the PEG arms were functionalized with collagenase substrates prior to crosslinking [253]. Preadipocytes cultured in these degradable hydrogels formed extensive networks resembling adipose tissue while only isolated cells could be seen in the control nondegradable hydrogels. Furthermore, the intracellular lipid droplets in degradable hydrogels were considerably enlarged, resulting in many unilocular cells, a typical feature of mature adipocytes. Multiarm PEGs with vinylsulfone functionalities have also been crosslinked with MMP-substrate peptides with terminal cysteine residues to fabricate degradable hydrogels [254]. This system allowed precise control over the degradability, matrix elasticity, and the concentration of matrix-conjugated RGD cell adhesive peptides.

MMP-1 substrate peptides have also been combined with the FXIIIa-mediated cross-linking strategy described earlier in order to fabricate MMP-1 and plasmin-sensitive PEG-based hydrogels under highly biocompatible conditions [193,255]. In this strategy, the enzymatically degradable peptide is combined with the FXIIIA substrate peptide (containing the donor FKGG sequence) in order to incorporate degradable motifs into the crosslinks. The ability to independently control biofunctionality, proteolytic degradability and crosslinking density in such hydrogels has allowed the study of the role of matrix stiffness and remodeling on 3D cell migration and formation of cellular networks (Figure 14.7) [255]. This study showed that preosteoblastic cells migrated equally efficiently in soft matrices irrespective of their degradability. However, in stiffer matrices, proteolytic degradation was necessary to facilitate migration. A similar trend was also observed in the ability of the cells to form networks in the hydrogels.

image
Figure 14.7 Matrix stiffness and MMP-sensitivity influence the formation of cellular networks. Single dispersed mouse preosteoblastic cells form dense homogeneous networks within 3 weeks in soft MMP-degradable PEG hydrogels. The networks are sparser and less uniform as matrix stiffness increases. Network formation is substantially reduced in soft nondegradable hydrogels and completely absent in stiff ones. Source: Reprinted from Ref. [255], with permission from Elsevier.

Interestingly, natural polymers have also been crosslinked using peptide sequences containing protease substrates in order to make the hydrogels susceptible to different enzymes. HA has been crosslinked using peptides containing MMP-2 substrate sequences, which rendered the hydrogels degradable by this protease [256]. For this, HA was first modified with both maleimide and methacrylate groups. The maleimides were subsequently reacted with a mixture of RGD peptides with one cysteine residue and MMP-substrate peptides with cysteine residues at each end to enable crosslinking. The methacrylate groups could also be photo-crosslinked to make the hydrogel matrix less sensitive to MMP-mediated degradation. This strategy of having two different crosslinkers allowed independent control over matrix mechanics and degradability. It was demonstrated that human mesenchymal stem cells (hMSCs) which do not secrete hyaluronidase could efficiently degrade the hydrogel by cleaving the MMP-sensitive peptides. Interestingly, the differentiation of these cells was directed by the degradability of the hydrogels independent of the matrix stiffness, with more osteogenic differentiation in the degradable hydrogels. A similar MMP-2 substrate peptide sequence has also been used to crosslink dextran to fabricate cell-degradable hydrogels which are otherwise nonhydrolyzable [257].

14.4 Hydrogel Nanoparticles

Hydrogels can thus be modified with a variety of biomolecules in order to fabricate a biologically instructive microenvironment for cells. Such bioactive hydrogels are used for tissue engineering applications as well as for encapsulating cells for in vivo delivery. Another major area of application of hydrogels has been in the field of controlled delivery of protein drugs such as antibodies and growth factors. Hydrophilic hydrogels provide a biocompatible environment for proteins that can protect them from denaturation, enzymatic degradation, and increase their in vivo circulation time. These properties make hydrogels an attractive medium for engineering the surface of therapeutic cells with exogenous proteins. Synthetic nanoparticles loaded with cytokines have been used to engineer the surface of therapeutic cells in order to increase their in vivo viability and function [258]. Drug-loaded synthetic nanoparticles have also been used for functionalization of cells for targeted delivery using the natural homing behavior of immune cells such as T cells and macrophages [259,260]. Synthetic nanoparticles have also been independently used as delivery vehicles for a variety of therapeutic proteins [261263]. The following section will describe a few approaches that have been used for the fabrication of hydrogel nanoparticles or nanogels.

14.4.1 Water-in-Oil Heterogeneous Emulsion

Water-in-oil (W/O or inverse emulsion) methods involve the formation of aqueous droplets (micelles) of hydrophilic polymers in a continuous oil phase by using oil-soluble surfactants, followed by crosslinking of the polymers using water-soluble crosslinkers (Figure 14.8). By using a relatively high concentration of the surfactant, micelles in the submicron range can be obtained, which then form nanogels after crosslinking [264]. Inverse emulsion methods have been used to fabricate chitosan nanogels loaded with the antitumor drug doxorubicin coupled with dextran [265]. In this study, a nanogel diameter of ~100 nm was discovered to favor tissue delivery via the enhanced permeability and retention (EPR) effect commonly exploited to deliver contrast agents and drugs to solid tumors. A similar strategy has also been applied for the preparation of HA-based nanogels [266]. Specifically, thiolated-HA was crosslinked in reverse micelles containing small interfering RNA (siRNA) to form nanogels with a diameter of 198 nm in order to enable intracellular delivery of these molecules for gene silencing. Inverse emulsion techniques have been extensively applied for the production of nanogels based on synthetic polymers such as poly(N-isopropylacrylamide) [267,268], polyacrylamide (PAAm) [269,270], poly(dimethylacrylamide) [271], poly(vinylpyrrolidone) [272,273], and the amphiphilic polymer Pluronic® [274]. Inverse emulsion has also been combined with controlled radical polymerization strategies such as atom transfer radical polymerization (ATRP) and reversible addition-fragmentation chain transfer (RAFT) polymerization to produce nanogels with well-controlled size distributions. This approach was used to fabricate stable nanogels of well-controlled water-soluble poly(oligo(ethylene glycol) monomethyl ether methacrylate) (POEOMA) with a very narrow size distribution [275,276]. Similar strategies have been used to synthesize nanogels based on PAAm [277] and PEO-b-PHEMA block copolymers [278].

image
Figure 14.8 A schematic representation of inverse emulsion methods used for fabrication of nanogels. Aqueous micelles containing monomers, initiators, crosslinkers, etc., are stabilized in a continuous oil phase using surfactants. Polymerization and crosslinking in these submicron-sized micelles results in the formation of nanogels.

14.4.2 Dispersion/Precipitation Polymerization

In dispersion/precipitation polymerization, the monomer, the polymerization-mediating species and the initiator are initially soluble in water to form a homogeneous solution. As the polymerization proceeds to a critical point following initiation, the generated polymers become insoluble in water and thus form a separate phase. RAFT precipitation/dispersion polymerization of N-isopropylacrylamide using poly(N,N′-dimethylacrylamide) has been used to synthesize core–shell thermoresponsive nanogels [279], which could then be selectively functionalized in the core and the shell using orthogonal chemistry [280]. Aqueous dispersion RAFT polymerization has been used for the synthesis of biocompatible, antifouling, and thermosensitive core–shell nanogels based on copolymerization of oligo(ethylene glycol) methacrylates of different side chain length [281]. A similar strategy based on RAFT-controlled radical crosslinking copolymerization of N,N-diethylacrylamide (DEAAm) and N,N′-methylene bisacrylamide in aqueous dispersion polymerization was also used to synthesize PEGylated thermoresponsive core–shell nanogels [282].

14.5 Conclusion

Hydrogels, with their water-filled network structure and biocompatible nature, are excellent in vitro structural mimics of the cellular microenvironment. However, they often lack the enormous biochemical diversity that is present in the native ECM. In this chapter, we have described traditional and emerging bioconjugation strategies that have been developed to functionalize hydrogels with a variety of biomolecules in order to convert them from inert scaffolds to complex microenvironments that can not only interact with cells but can also be modified by them. This has in turn fueled many successful efforts to integrate growth factors and ECM-derived proteins and peptides to confer important bioactivity to otherwise inert materials. More recently, the ability of specific ECM components (whether native or synthetic) to sequester growth factors and cytokines has been exploited to concentrate cell-derived factors and enhance autocrine and paracrine signaling to promote tissue function. All of these strategies may be aided in the future by new peptide, protein, and aptameric tags that can be fused to target proteins and hydrogels in order to enable site-specific and selective immobilization of factors onto matrices. Moreover, the newest generation of scaffolds has begun to accommodate and direct biodegradability in highly specific ways, most notably by incorporation of crosslinks that may be enzymatically degraded to permit tissue growth and vascularization. Finally, the fabrication of hydrogel nanoparticles with “intelligent” material properties represents an important new frontier in drug delivery systems. A key recurring theme in these newest approaches is the desire to design material properties that optimally interface with biological systems. Whereas traditional biomaterial design may have traditionally focused on choosing a material that “does the least harm” in a given biological system, we are rapidly entering an era in which materials can be designed to both facilitate desirable biological processes and actively direct them.

List of Abbreviations

α2-PI α2-plasmin inhibitor

ADAMTS A disintegrin and metalloproteinase with thrombospondin motifs

BMP Bone morphogenetic protein

CBD Collagen-binding domain

CTGF Connective tissue growth factor

ECM Extracellular matrix

FGF Fibroblast growth factor

FN Fibronectin

GAG Glycosaminoglycan

HA Hyaluronic acid

HGF Hepatocyte growth factor

IGF Insulin-like growth factor

MC Methyl cellulose

MMP Matrix metalloproteinase

NGF Nerve growth factor

NHS N-hydroxysuccinimide

PDGF Platelet-derived growth factor

PEG Poly(ethyleneglycol)

PEGDA Poly(ethyleneglycol)-diacrylate

PLA Poly(lactic acid)

PlGF Placental growth factor

PVA Poly(vinylalcohol)

SHH Sonic hedgehog

SMC Smooth muscle cell

TGF Transforming growth factor

TNC Tenascin-C

VEGF Vascular endothelial growth factor

vWF von Willebrand factor

References

1. Gjorevski N, Nelson CM. Bidirectional extracellular matrix signaling during tissue morphogenesis. Cytokine Growth Factor Rev. 2009;20(5–6):459–465.

2. Tsang KY, Cheung MC, Chan D, Cheah KS. The developmental roles of the extracellular matrix: beyond structure to regulation. Cell Tissue Res. 2010;339(1):93–110.

3. Jarvelainen H, Sainio A, Koulu M, Wight TN, Penttinen R. Extracellular matrix molecules: potential targets in pharmacotherapy. Pharmacol Rev. 2009;61(2):198–223.

4. Denys H, Braems G, Lambein K, et al. The extracellular matrix regulates cancer progression and therapy response: implications for prognosis and treatment. Curr Pharm Des. 2009;15(12):1373–1384.

5. Lu P, Weaver VM, Werb Z. The extracellular matrix: a dynamic niche in cancer progression. J Cell Biol. 2012;196(4):395–406.

6. Tanzer ML. Current concepts of extracellular matrix. J Orthop Sci. 2006;11(3):326–331.

7. Bosman FT, Stamenkovic I. Functional structure and composition of the extracellular matrix. J Pathol. 2003;200(4):423–428.

8. Frantz C, Stewart KM, Weaver VM. The extracellular matrix at a glance. J Cell Sci. 2010;123(24):4195–4200.

9. Lu Q, Ganesan K, Simionescu DT, Vyavahare NR. Novel porous aortic elastin and collagen scaffolds for tissue engineering. Biomaterials. 2004;25(22):5227–5237.

10. Simionescu DT, Lu Q, Song Y, et al. Biocompatibility and remodeling potential of pure arterial elastin and collagen scaffolds. Biomaterials. 2006;27(5):702–713.

11. Buijtenhuijs P, Buttafoco L, Poot AA, et al. Tissue engineering of blood vessels: characterization of smooth-muscle cells for culturing on collagen-and-elastin-based scaffolds. Biotechnol Appl Biochem. 2004;39(Pt 2):141–149.

12. Chan-Park MB, Shen JY, Cao Y, et al. Biomimetic control of vascular smooth muscle cell morphology and phenotype for functional tissue-engineered small-diameter blood vessels. J Biomed Mater Res A. 2009;88A(4):1104–1121.

13. Plow EF, Haas TA, Zhang L, Loftus J, Smith JW. Ligand binding to integrins. J Biol Chem. 2000;275(29):21785–21788.

14. Colognato H, Yurchenco PD. Form and function: the laminin family of heterotrimers. Dev Dyn. 2000;218(2):213–234.

15. Leitinger B, Hohenester E. Mammalian collagen receptors. Matrix Biol. 2007;26(3):146–155.

16. Harburger DS, Calderwood DA. Integrin signalling at a glance. J Cell Sci. 2009;122(2):159–163.

17. Rodgers UR, Weiss AS. Cellular interactions with elastin. Pathol Biol. 2005;53(7):390–398.

18. Rodgers UR, Weiss AS. Integrin αvβ3 binds a unique non-RGD site near the C-terminus of human tropoelastin. Biochimie. 2004;86(3):173–178.

19. Bax DV, Rodgers UR, Bilek MM, Weiss AS. Cell adhesion to tropoelastin is mediated via the C-terminal GRKRK motif and integrin αVβ3. J Biol Chem. 2009;284(42):28616–28623.

20. Hynes RO. Integrins: bidirectional, allosteric signaling machines. Cell. 2002;110(6):673–687.

21. Ruoslahti E. RGD and other recognition sequences for integrins. Annu Rev Cell Dev Biol. 1996;12:697–715.

22. Pierschbacher MD, Ruoslahti E. Cell attachment activity of fibronectin can be duplicated by small synthetic fragments of the molecule. Nature. 1984;309(5963):30–33.

23. Esko JD, Kimata K, Lindahl U. Proteoglycans and sulfated glycosaminoglycans. In: Varki A, Cummings RD, Esko JD, eds. Essentials of glycobiology. 2nd ed. New York, NY: Cold Spring Harbor Laboratory Press; 2009.

24. Taipale J, Keski-Oja J. Growth factors in the extracellular matrix. FASEB J. 1997;11(1):51–59.

25. Jeon O, Powell C, Solorio LD, Krebs MD, Alsberg E. Affinity-based growth factor delivery using biodegradable, photocrosslinked heparin–alginate hydrogels. J Control Release. 2011;154(3):258–266.

26. Xu X, Jha AK, Duncan RL, Jia X. Heparin-decorated, hyaluronic acid-based hydrogel particles for the controlled release of bone morphogenetic protein 2. Acta Biomater. 2011;7(8):3050–3059.

27. Lu P, Takai K, Weaver VM, Werb Z. Extracellular matrix degradation and remodeling in development and disease. Cold Spring Harb Perspect Biol. 2011;3:12.

28. Daley WP, Peters SB, Larsen M. Extracellular matrix dynamics in development and regenerative medicine. J Cell Sci. 2008;121(3):255–264.

29. Stamenkovic I. Extracellular matrix remodelling: the role of matrix metalloproteinases. J Pathol. 2003;200(4):448–464.

30. Page-McCaw A, Ewald AJ, Werb Z. Matrix metalloproteinases and the regulation of tissue remodelling. Nat Rev Mol Cell Biol. 2007;8(3):221–233.

31. Smith HW, Marshall CJ. Regulation of cell signalling by uPAR. Nat Rev Mol Cell Biol. 2010;11(1):23–36.

32. Apte SS. A disintegrin-like and metalloprotease (reprolysin type) with thrombospondin type 1 motifs: the ADAMTS family. Int J Biochem Cell Biol. 2004;36(6):981–985.

33. Tang BL. ADAMTS: a novel family of extracellular matrix proteases. Int J Biochem Cell Biol. 2001;33(1):33–44.

34. Stern R, Jedrzejas MJ. Hyaluronidases: their genomics, structures, and mechanisms of action. Chem Rev. 2006;106(3):818–839.

35. Chan BP, Leong KW. Scaffolding in tissue engineering: general approaches and tissue-specific considerations. Eur Spine J. 2008;17(4):467–479.

36. Zhu J. Bioactive modification of poly(ethylene glycol) hydrogels for tissue engineering. Biomaterials. 2010;31(17):4639–4656.

37. Prestwich GD. Evaluating drug efficacy and toxicology in three dimensions: using synthetic extracellular matrices in drug discovery. Acc Chem Res. 2007;41(1):139–148.

38. Prestwich GD. Hyaluronic acid-based clinical biomaterials derived for cell and molecule delivery in regenerative medicine. J Control Release. 2011;155(2):193–199.

39. Perale G, Rossi F, Sundstrom E, et al. Hydrogels in spinal cord injury repair strategies. ACS Chem Neurosci. 2011;2(7):336–345.

40. Soler-Botija C, Bagó JR, Bayes-Genis A. A bird’s-eye view of cell therapy and tissue engineering for cardiac regeneration. Ann NY Acad Sci. 2012;1254(1):57–65.

41. Johnson TD, Christman KL. Injectable hydrogel therapies and their delivery strategies for treating myocardial infarction. Expert Opin Drug Deliv. 2013;10(1):59–72.

42. Peppas NA, Hilt JZ, Khademhosseini A, Langer R. Hydrogels in biology and medicine: from molecular principles to bionanotechnology. Adv Mater. 2006;18(11):1345–1360.

43. Hoare TR, Kohane DS. Hydrogels in drug delivery: progress and challenges. Polymer (Guildf). 2008;49(8):1993–2007.

44. Zhu J, Marchant RE. Design properties of hydrogel tissue-engineering scaffolds. Expert Rev Med Devices. 2011;8(5):607–626.

45. Seliktar D. Designing cell-compatible hydrogels for biomedical applications. Science. 2012;336(6085):1124–1128.

46. Geckil H, Xu F, Zhang X, Moon S, Demirci U. Engineering hydrogels as extracellular matrix mimics. Nanomedicine. 2010;5(3):469–484.

47. Slaughter BV, Khurshid SS, Fisher OZ, Khademhosseini A, Peppas NA. Hydrogels in regenerative medicine. Adv Mater. 2009;21(32–33):3307–3329.

48. Cushing MC, Anseth KS. Hydrogel cell cultures. Science. 2007;316(5828):1133–1134.

49. Brandl F, Sommer F, Goepferich A. Rational design of hydrogels for tissue engineering: impact of physical factors on cell behavior. Biomaterials. 2007;28(2):134–146.

50. Glowacki J, Mizuno S. Collagen scaffolds for tissue engineering. Biopolymers. 2008;89(5):338–344.

51. Sakai S, Hirose K, Taguchi K, Ogushi Y, Kawakami K. An injectable, in situ enzymatically gellable, gelatin derivative for drug delivery and tissue engineering. Biomaterials. 2009;30(20):3371–3377.

52. Daamen WF, Veerkamp JH, van Hest JCM, van Kuppevelt TH. Elastin as a biomaterial for tissue engineering. Biomaterials. 2007;28(30):4378–4398.

53. Mol A, van Lieshout MI, Dam-de Veen CG, et al. Fibrin as a cell carrier in cardiovascular tissue engineering applications. Biomaterials. 2005;26(16):3113–3121.

54. MacEwan SR, Chilkoti A. Elastin-like polypeptides: biomedical applications of tunable biopolymers. Pept Sci. 2010;94(1):60–77.

55. Lampe KJ, Antaris AL, Heilshorn SC. Design of three-dimensional engineered protein hydrogels for tailored control of neurite growth. Acta Biomater. 2013;9(3):5590–5599.

56. Benitez PL, Sweet JA, Fink H, et al. Sequence-specific crosslinking of electrospun, elastin-like protein preserves bioactivity and native-like mechanics. Adv Healthcare Mater. 2013;2(1):114–118.

57. Sengupta D, Gilbert PM, Johnson KJ, Blau HM, Heilshorn SC. Protein-engineered biomaterials to generate human skeletal muscle mimics. Adv Healthcare Mater. 2012;1(6):785–789.

58. McHale MK, Setton LA, Chilkoti A. Synthesis and in vitro evaluation of enzymatically cross-linked elastin-like polypeptide gels for cartilaginous tissue repair. Tissue Eng. 2005;11(11–12):1768–1779.

59. Lim DW, Nettles DL, Setton LA, Chilkoti A. Rapid cross-linking of elastin-like polypeptides with (hydroxymethyl)phosphines in aqueous solution. Biomacromolecules. 2007;8(5):1463–1470.

60. Nowak AP, Breedveld V, Pakstis L, et al. Rapidly recovering hydrogel scaffolds from self-assembling diblock copolypeptide amphiphiles. Nature. 2002;417(6887):424–428.

61. Pakstis LM, Ozbas B, Hales KD, Nowak AP, Deming TJ, Pochan D. Effect of chemistry and morphology on the biofunctionality of self-assembling diblock copolypeptide hydrogels. Biomacromolecules. 2003;5(2):312–318.

62. Pochan DJ, Schneider JP, Kretsinger J, Ozbas B, Rajagopal K, Haines L. Thermally reversible hydrogels via intramolecular folding and consequent self-assembly of a de novo designed peptide. J Am Chem Soc. 2003;125(39):11802–11803.

63. Ozbas B, Kretsinger J, Rajagopal K, Schneider JP, Pochan DJ. Salt-triggered peptide folding and consequent self-assembly into hydrogels with tunable modulus. Macromolecules. 2004;37(19):7331–7337.

64. Schneider JP, Pochan DJ, Ozbas B, Rajagopal K, Pakstis L, Kretsinger J. Responsive hydrogels from the intramolecular folding and self-assembly of a designed peptide. J Am Chem Soc. 2002;124(50):15030–15037.

65. Altunbas A, Pochan DJ. Peptide-based and polypeptide-based hydrogels for drug delivery and tissue engineering. Top Curr Chem. 2012;310:135–167.

66. DiMarco RL, Heilshorn SC. Multifunctional materials through modular protein engineering. Adv Mater. 2012;24(29):3923–3940.

67. Xu X, Jha AK, Harrington DA, Farach-Carson MC, Jia X. Hyaluronic acid-based hydrogels: from a natural polysaccharide to complex networks. Soft Matter. 2012;8(12):3280–3294.

68. Burdick JA, Prestwich GD. Hyaluronic acid hydrogels for biomedical applications. Adv Mater. 2011;23(12):H41–H56.

69. Toole BP, Wight TN, Tammi MI. Hyaluronan–cell interactions in cancer and vascular disease. J Biol Chem. 2002;277(7):4593–4596.

70. Toole BP, Hascall VC. Hyaluronan and tumor growth. Am J Pathol. 2002;161(3):745–747.

71. Liu Y, Shu XZ, Prestwich GD. Tumor engineering: orthotopic cancer models in mice using cell-loaded, injectable, cross-linked hyaluronan-derived hydrogels. Tissue Eng. 2007;13(5):1091–1101.

72. Xu X, Prestwich GD. Inhibition of tumor growth and angiogenesis by a lysophosphatidic acid antagonist in an engineered three-dimensional lung cancer xenograft model. Cancer. 2010;116(7):1739–1750.

73. Serban MA, Scott A, Prestwich GD. Use of hyaluronan-derived hydrogels for three-dimensional cell culture and tumor xenografts. Curr Protoc Cell Biol 2008; Chapter 10: Unit 10 14.

74. Jin SG, Jeong YI, Jung S, Ryu HH, Jin YH, Kim IY. The effect of hyaluronic acid on the invasiveness of malignant glioma cells: comparison of invasion potential at hyaluronic acid hydrogel and matrigel. J Korean Neurosurg Soc. 2009;46(5):472–478.

75. Ananthanarayanan B, Kim Y, Kumar S. Elucidating the mechanobiology of malignant brain tumors using a brain matrix-mimetic hyaluronic acid hydrogel platform. Biomaterials. 2011;32(31):7913–7923.

76. Kirker KR, Prestwich GD. Physical properties of glycosaminoglycan hydrogels. J Polym Sci B Polym Phys. 2004;42(23):4344–4356.

77. Cheng Y, Luo X, Payne GF, Rubloff GW. Biofabrication: programmable assembly of polysaccharide hydrogels in microfluidics as biocompatible scaffolds. J Mater Chem. 2012;22(16):7659–7666.

78. Ramamurthi A, Vesely I. Ultraviolet light-induced modification of crosslinked hyaluronan gels. J Biomed Mater Res A. 2003;66(2):317–329.

79. Kuo CK, Ma PX. Ionically crosslinked alginate hydrogels as scaffolds for tissue engineering: part 1 Structure, gelation rate and mechanical properties. Biomaterials. 2001;22(6):511–521.

80. Kim IY, Seo SJ, Moon HS, et al. Chitosan and its derivatives for tissue engineering applications. Biotechnol Adv. 2008;26(1):1–21.

81. Sawhney AS, Pathak CP, Hubbell JA. Bioerodible hydrogels based on photopolymerized poly(ethylene glycol)-co-poly(.alpha-hydroxy acid) diacrylate macromers. Macromolecules. 1993;26(4):581–587.

82. Burdick JA, Anseth KS. Photoencapsulation of osteoblasts in injectable RGD-modified PEG hydrogels for bone tissue engineering. Biomaterials. 2002;23(22):4315–4323.

83. Bryant SJ, Anseth KS. Hydrogel properties influence ECM production by chondrocytes photoencapsulated in poly(ethylene glycol) hydrogels. J Biomed Mater Res. 2002;59(1):63–72.

84. Adelow C, Segura T, Hubbell JA, Frey P. The effect of enzymatically degradable poly(ethylene glycol) hydrogels on smooth muscle cell phenotype. Biomaterials. 2008;29(3):314–326.

85. Peyton SR, Raub CB, Keschrumrus VP, Putnam AJ. The use of poly(ethylene glycol) hydrogels to investigate the impact of ECM chemistry and mechanics on smooth muscle cells. Biomaterials. 2006;27(28):4881–4893.

86. Tessmar JK, Göpferich AM. Matrices and scaffolds for protein delivery in tissue engineering. Adv Drug Deliv Rev. 2007;59(4–5):274–291.

87. Sokolsky-Papkov M, Agashi K, Olaye A, Shakesheff K, Domb AJ. Polymer carriers for drug delivery in tissue engineering. Adv Drug Deliv Rev. 2007;59(4–5):187–206.

88. Xie J, Schultz PG. Adding amino acids to the genetic repertoire. Curr Opin Chem Biol. 2005;9(6):548–554.

89. Wang L, Brock A, Herberich B, Schultz PG. Expanding the genetic code of Escherichia coli. Science. 2001;292(5516):498–500.

90. Sakamoto K, Hayashi A, Sakamoto A, et al. Site-specific incorporation of an unnatural amino acid into proteins in mammalian cells. Nucleic Acids Res. 2002;30(21):4692–4699.

91. Cretu A, Castagnino P, Assoian R. Studying the effects of matrix stiffness on cellular function using acrylamide-based hydrogels. J Vis Exp 2010.

92. Drumheller PD, Hubbell JA. Polymer networks with grafted cell adhesion peptides for highly biospecific cell adhesive substrates. Anal Biochem. 1994;222(2):380–388.

93. Ghosh K, Ren XD, Shu XZ, Prestwich GD, Clark RA. Fibronectin functional domains coupled to hyaluronan stimulate adult human dermal fibroblast responses critical for wound healing. Tissue Eng. 2006;12(3):601–613.

94. DeLong SA, Moon JJ, West JL. Covalently immobilized gradients of bFGF on hydrogel scaffolds for directed cell migration. Biomaterials. 2005;26(16):3227–3234.

95. Phelps EA, Landázuri N, Thulé PM, Taylor WR, García AJ. Bioartificial matrices for therapeutic vascularization. Proc Natl Acad Sci USA. 2010;107(8):3323–3328.

96. Young JL, Engler AJ. Hydrogels with time-dependent material properties enhance cardiomyocyte differentiation in vitro. Biomaterials. 2011;32(4):1002–1009.

97. Nuttelman CR, Mortisen DJ, Henry SM, Anseth KS. Attachment of fibronectin to poly(vinyl alcohol) hydrogels promotes NIH3T3 cell adhesion, proliferation, and migration. J Biomed Mater Res. 2001;57(2):217–223.

98. Trappmann B, Gautrot JE, Connelly JT, et al. Extracellular-matrix tethering regulates stem-cell fate. Nat Mater. 2012;11(7):642–649.

99. Stabenfeldt SE, Munglani G, Garcia AJ, LaPlaca MC. Biomimetic microenvironment modulates neural stem cell survival, migration, and differentiation. Tissue Eng Part A. 2010;16(12):3747–3758.

100. Chan V, Collens MB, Jeong JH, Park K, Kong H, Bashir R. Directed cell growth and alignment on protein-patterned 3D hydrogels with stereolithography. Virtual Phys Prototyp. 2012;7(3):219–228.

101. Seidlits SK, Drinnan CT, Petersen RR, Shear JB, Suggs LJ, Schmidt CE. Fibronectin–hyaluronic acid composite hydrogels for three-dimensional endothelial cell culture. Acta Biomater. 2011;7(6):2401–2409.

102. Underhill GH, Chen AA, Albrecht DR, Bhatia SN. Assessment of hepatocellular function within PEG hydrogels. Biomaterials. 2007;28(2):256–270.

103. Patel PN, Gobin AS, West JL, Patrick Jr CW. Poly(ethylene glycol) hydrogel system supports preadipocyte viability, adhesion, and proliferation. Tissue Eng. 2005;11(9–10):1498–1505.

104. Saik JE, Gould DJ, Watkins EM, Dickinson ME, West JL. Covalently immobilized platelet-derived growth factor-BB promotes angiogenesis in biomimetic poly(ethylene glycol) hydrogels. Acta Biomater. 2011;7(1):133–143.

105. Owen SC, Fisher SA, Tam RY, Nimmo CM, Shoichet MS. Hyaluronic acid click hydrogels emulate the extracellular matrix. Langmuir. 2013;29(24):7393–7400.

106. Hynd MR, Frampton JP, Dowell-Mesfin N, Turner JN, Shain W. Directed cell growth on protein-functionalized hydrogel surfaces. J Neurosci Methods. 2007;162(1–2):255–263.

107. Hynd MR, Frampton JP, Burnham MR, et al. Functionalized hydrogel surfaces for the patterning of multiple biomolecules. J Biomed Mater Res A. 2007;81(2):347–354.

108. Cosson S, Kobel SA, Lutolf MP. Capturing complex protein gradients on biomimetic hydrogels for cell-based assays. Adv Funct Mater. 2009;19(21):3411–3419.

109. Zhang C, Hekmatfer S, Karuri NW. A comparative study of polyethylene glycol hydrogels derivatized with the RGD peptide and the cell-binding domain of fibronectin. J Biomed Mater Res A. 2014;102A:170–179.

110. Shu XZ, Ghosh K, Liu Y, Palumbo FS, Luo Y, Clark RA. Attachment and spreading of fibroblasts on an RGD peptide-modified injectable hyaluronan hydrogel. J Biomed Mater Res A. 2004;68(2):365–375.

111. Marklein RA, Burdick JA. Spatially controlled hydrogel mechanics to modulate stem cell interactions. Soft Matter. 2010;6(1):136–143.

112. McCall JD, Luoma JE, Anseth KS. Covalently tethered transforming growth factor beta in PEG hydrogels promotes chondrogenic differentiation of encapsulated human mesenchymal stem cells. Drug Deliv Transl Res. 2012;2(5):305–312.

113. Chatani S, Nair DP, Bowman CN. Relative reactivity and selectivity of vinyl sulfones and acrylates towards the thiol-Michael addition reaction and polymerization. Polym Chem. 2013;4(4):1048–1055.

114. Wacker BK, Alford SK, Scott EA, Das Thakur M, Longmore GD, Elbert DL. Endothelial cell migration on RGD-peptide-containing PEG hydrogels in the presence of sphingosine 1-phosphate. Biophys J. 2008;94(1):273–285.

115. Zisch AH, Lutolf MP, Ehrbar M, et al. Cell-demanded release of VEGF from synthetic, biointeractive cell-ingrowth matrices for vascularized tissue growth. FASEB J. 2003;17(15):2260–2262.

116. Phelps EA, Enemchukwu NO, Fiore VF, et al. Maleimide cross-linked bioactive PEG hydrogel exhibits improved reaction kinetics and cross-linking for cell encapsulation and in situ delivery. Adv Mater. 2012;24(1):64–70 2.

117. Fu Y, Kao WJ. In situ forming poly(ethylene glycol)-based hydrogels via thiol-maleimide Michael-type addition. J Biomed Mater Res A. 2011;98A(2):201–211.

118. Nie T, Akins Jr RE, Kiick KL. Production of heparin-containing hydrogels for modulating cell responses. Acta Biomater. 2009;5(3):865–875.

119. Lin CC, Raza A, Shih H. PEG hydrogels formed by thiol-ene photo-click chemistry and their effect on the formation and recovery of insulin-secreting cell spheroids. Biomaterials. 2011;32(36):9685–9695.

120. Raza A, Ki CS, Lin CC. The influence of matrix properties on growth and morphogenesis of human pancreatic ductal epithelial cells in 3D. Biomaterials. 2013;34(21):5117–5127.

121. Polizzotti BD, Fairbanks BD, Anseth KS. Three-dimensional biochemical patterning of click-based composite hydrogels via thiolene photopolymerization. Biomacromolecules. 2008;9(4):1084–1087.

122. Deforest CA, Sims EA, Anseth KS. Peptide-functionalized click hydrogels with independently tunable mechanics and chemical functionality for 3D cell culture. Chem Mater. 2010;22(16):4783–4790.

123. Capila I, Linhardt RJ. Heparin–protein interactions. Angew Chem Int Ed. 2002;41(3):391–412.

124. Thompson LD, Pantoliano MW, Springer BA. Energetic characterization of the basic fibroblast growth factor–heparin interaction: identification of the heparin binding domain. Biochemistry. 1994;33(13):3831–3840.

125. Schultz GS, Wysocki A. Interactions between extracellular matrix and growth factors in wound healing. Wound Repair Regen. 2009;17(2):153–162.

126. Saksela O, Moscatelli D, Sommer A, Rifkin DB. Endothelial cell-derived heparan sulfate binds basic fibroblast growth factor and protects it from proteolytic degradation. J Cell Biol. 1988;107(2):743–751.

127. Sommer A, Rifkin DB. Interaction of heparin with human basic fibroblast growth factor: protection of the angiogenic protein from proteolytic degradation by a glycosaminoglycan. J Cell Physiol. 1989;138(1):215–220.

128. Pellegrini L. Role of heparan sulfate in fibroblast growth factor signalling: a structural view. Curr Opin Struct Biol. 2001;11(5):629–634.

129. Mohammadi M, Olsen SK, Ibrahimi OA. Structural basis for fibroblast growth factor receptor activation. Cytokine Growth Factor Rev. 2005;16(2):107–137.

130. Tae G, Scatena M, Stayton PS, Hoffman AS. PEG-cross-linked heparin is an affinity hydrogel for sustained release of vascular endothelial growth factor. J Biomater Sci Polym Ed. 2006;17(1–2):187–197.

131. Zieris A, Chwalek K, Prokoph S, et al. Dual independent delivery of pro-angiogenic growth factors from starPEG-heparin hydrogels. J Control Release. 2011;156(1):28–36.

132. Kim M, Lee JY, Jones CN, Revzin A, Tae G. Heparin-based hydrogel as a matrix for encapsulation and cultivation of primary hepatocytes. Biomaterials. 2010;31(13):3596–3603.

133. Ohta M, Suzuki Y, Chou H, et al. Novel heparin/alginate gel combined with basic fibroblast growth factor promotes nerve regeneration in rat sciatic nerve. J Biomed Mater Res A. 2004;71(4):661–668.

134. Locci P, Marinucci L, Lilli C, Martinese D, Becchetti E. Transforming growth factor beta 1-hyaluronic acid interaction. Cell Tissue Res. 1995;281(2):317–324.

135. Lyon M, Deakin JA, Rahmoune H, Fernig DG, Nakamura T, Gallagher JT. Hepatocyte growth factor/scatter factor binds with high affinity to dermatan sulfate. J Biol Chem. 1998;273(1):271–278.

136. Catlow K, Deakin JA, Delehedde M, et al. Hepatocyte growth factor/scatter factor and its interaction with heparan sulphate and dermatan sulphate. Biochem Soc Trans. 2003;31(2):352–353.

137. Martino MM, Hubbell JA. The 12th–14th type III repeats of fibronectin function as a highly promiscuous growth factor-binding domain. FASEB J. 2010;24(12):4711–4721.

138. Martino MM, Briquez PS, Ranga A, Lutolf MP, Hubbell JA. Heparin-binding domain of fibrin(ogen) binds growth factors and promotes tissue repair when incorporated within a synthetic matrix. Proc Natl Acad Sci USA. 2013;110(12):4563–4568.

139. Macri L, Silverstein D, Clark RAF. Growth factor binding to the pericellular matrix and its importance in tissue engineering. Adv Drug Deliv Rev. 2007;59(13):1366–1381.

140. Mooradian DL, Lucas RC, Weatherbee JA, Furcht LT. Transforming growth factor-beta 1 binds to immobilized fibronectin. J Cell Biochem. 1989;41(4):189–200.

141. Rahman S, Patel Y, Murray J, et al. Novel hepatocyte growth factor (HGF) binding domains on fibronectin and vitronectin coordinate a distinct and amplified Met-integrin induced signalling pathway in endothelial cells. BMC Cell Biol. 2005;6(1):8.

142. Hoshijima M, Hattori T, Inoue M, et al. CT domain of CCN2/CTGF directly interacts with fibronectin and enhances cell adhesion of chondrocytes through integrin α5β1. FEBS Lett. 2006;580(5):1376–1382.

143. Smith EM, Mitsi M, Nugent MA, Symes K. PDGF-A interactions with fibronectin reveal a critical role for heparan sulfate in directed cell migration during Xenopus gastrulation. Proc Natl Acad Sci USA. 2009;106(51):21683–21688.

144. Wijelath ES, Murray J, Rahman S, et al. Novel vascular endothelial growth factor binding domains of fibronectin enhance vascular endothelial growth factor biological activity. Circ Res. 2002;91(1):25–31.

145. Wijelath ES, Rahman S, Namekata M, et al. Heparin-II domain of fibronectin is a vascular endothelial growth factor-binding domain: enhancement of VEGF biological activity by a singular growth factor/matrix protein synergism. Circ Res. 2006;99(8):853–860.

146. Bossard C, Van den Berghe L, Laurell H, et al. Antiangiogenic properties of fibstatin, an extracellular FGF-2-binding polypeptide. Cancer Res. 2004;64(20):7507–7512.

147. Martino MM, Tortelli F, Mochizuki M, et al. Engineering the growth factor microenvironment with fibronectin domains to promote wound and bone tissue healing. Sci Transl Med. 2011;3(100):100ra89.

148. Sahni A, Odrljin T, Francis CW. Binding of basic fibroblast growth factor to fibrinogen and fibrin. J Biol Chem. 1998;273(13):7554–7559.

149. Sahni A, Francis CW. Vascular endothelial growth factor binds to fibrinogen and fibrin and stimulates endothelial cell proliferation. Blood. 2000;96(12):3772–3778.

150. De Laporte L, Rice JJ, Tortelli F, Hubbell JA. Tenascin C promiscuously binds growth factors via its fifth fibronectin type III-like domain. PLoS One. 2013;8(4):e62076.

151. Somasundaram R, Schuppan D. Type I, II, III, IV, V, and VI collagens serve as extracellular ligands for the isoforms of platelet-derived growth factor (AA, BB, and AB). J Biol Chem. 1996;271(43):26884–26891.

152. Schuppan D, Schmid M, Somasundaram R, et al. Collagens in the liver extracellular matrix bind hepatocyte growth factor. Gastroenterology. 1998;114(1):139–152.

153. Paralkar VM, Vukicevic S, Reddi AH. Transforming growth factor β type 1 binds to collagen IV of basement membrane matrix: implications for development. Dev Biol. 1991;143(2):303–308.

154. Upton Z, Webb H, Hale K, et al. Identification of vitronectin as a novel insulin-like growth factor-II binding protein. Endocrinology. 1999;140(6):2928–2931.

155. Noble A, Towne C, Chopin L, Leavesley D, Upton Z. Insulin-like growth factor-II bound to vitronectin enhances MCF-7 breast cancer cell migration. Endocrinology. 2003;144(6):2417–2424.

156. Schoppet M, Chavakis T, Al-Fakhri N, Kanse SM, Preissner KT. Molecular interactions and functional interference between vitronectin and transforming growth factor-beta. Lab Invest. 2002;82(1):37–46.

157. Tada S, Kitajima T, Ito Y. Design and synthesis of binding growth factors. Int J Mol Sci. 2012;13(5):6053–6072.

158. Tuan TL, Cheung DT, Wu LT, et al. Engineering, expression and renaturation of targeted TGF-beta fusion proteins. Connect Tissue Res. 1996;34(1):1–9.

159. Andrades JA, Han B, Becerra J, Sorgente N, Hall FL, Nimni ME. A recombinant human TGF-beta1 fusion protein with collagen-binding domain promotes migration, growth, and differentiation of bone marrow mesenchymal cells. Exp Cell Res. 1999;250(2):485–498.

160. Hall FL, Kaiser A, Liu L, et al. Design, expression, and renaturation of a lesion-targeted recombinant epidermal growth factor–von Willebrand factor fusion protein: efficacy in an animal model of experimental colitis. Int J Mol Med. 2000;6(6):635–643.

161. Andrades JA, Santamaria JA, Wu LT, Hall FL, Nimni ME, Becerra J. Production of a recombinant human basic fibroblast growth factor with a collagen binding domain. Protoplasma. 2001;218(1–2):95–103.

162. Chen B, Lin H, Zhao Y, et al. Activation of demineralized bone matrix by genetically engineered human bone morphogenetic protein-2 with a collagen binding domain derived from von Willebrand factor propolypeptide. J Biomed Mater Res A. 2007;80(2):428–434.

163. Visser R, Arrabal PM, Becerra J, Rinas U, Cifuentes M. The effect of an rhBMP-2 absorbable collagen sponge-targeted system on bone formation in vivo. Biomaterials. 2009;30(11):2032–2037.

164. Han B, Perelman N, Tang B, Hall F, Shors EC, Nimni ME. Collagen-targeted BMP3 fusion proteins arrayed on collagen matrices or porous ceramics impregnated with Type I collagen enhance osteogenesis in a rat cranial defect model. J Orthop Res. 2002;20(4):747–755.

165. Lin H, Chen B, Sun W, Zhao W, Zhao Y, Dai J. The effect of collagen-targeting platelet-derived growth factor on cellularization and vascularization of collagen scaffolds. Biomaterials. 2006;27(33):5708–5714.

166. Sun W, Lin H, Xie H, et al. Collagen membranes loaded with collagen-binding human PDGF-BB accelerate wound healing in a rabbit dermal ischemic ulcer model. Growth Factors. 2007;25(5):309–318.

167. Sun W, Lin H, Chen B, Zhao W, Zhao Y, Dai J. Promotion of peripheral nerve growth by collagen scaffolds loaded with collagen-targeting human nerve growth factor-beta. J Biomed Mater Res A. 2007;83(4):1054–1061.

168. Fan J, Xiao Z, Zhang H, et al. Linear ordered collagen scaffolds loaded with collagen-binding neurotrophin-3 promote axonal regeneration and partial functional recovery after complete spinal cord transection. J Neurotrauma. 2010;27(9):1671–1683.

169. Zhang J, Ding L, Zhao Y, et al. Collagen-targeting vascular endothelial growth factor improves cardiac performance after myocardial infarction. Circulation. 2009;119(13):1776–1784.

170. Lin N, Li X, Song T, et al. The effect of collagen-binding vascular endothelial growth factor on the remodeling of scarred rat uterus following full-thickness injury. Biomaterials. 2012;33(6):1801–1807.

171. Yang Y, Zhao Y, Chen B, et al. Collagen-binding human epidermal growth factor promotes cellularization of collagen scaffolds. Tissue Eng Part A. 2009;15(11):3589–3596.

172. Zhao W, Chen B, Li X, et al. Vascularization and cellularization of collagen scaffolds incorporated with two different collagen-targeting human basic fibroblast growth factors. J Biomed Mater Res A. 2007;82(3):630–636.

173. Pang Y, Wang X, Ucuzian AA, et al. Local delivery of a collagen-binding FGF-1 chimera to smooth muscle cells in collagen scaffolds for vascular tissue engineering. Biomaterials. 2010;31(5):878–885.

174. Brewster LP, Washington C, Brey EM, et al. Construction and characterization of a thrombin-resistant designer FGF-based collagen binding domain angiogen. Biomaterials. 2008;29(3):327–336.

175. Nishi N, Matsushita O, Yuube K, Miyanaka H, Okabe A, Wada F. Collagen-binding growth factors: production and characterization of functional fusion proteins having a collagen-binding domain. Proc Natl Acad Sci USA. 1998;95(12):7018–7023.

176. Akimoto M, Takeda A, Matsushita O, et al. Effects of CB-VEGF-A injection in rat flap models for improved survival. Plast Reconstr Surg. 2013;131(4):717–725.

177. Kitajima T, Terai H, Ito Y. A fusion protein of hepatocyte growth factor for immobilization to collagen. Biomaterials. 2007;28(11):1989–1997.

178. Lu H, Kawazoe N, Kitajima T, et al. Spatial immobilization of bone morphogenetic protein-4 in a collagen-PLGA hybrid scaffold for enhanced osteoinductivity. Biomaterials. 2012;33(26):6140–6146.

179. Ishikawa T, Terai H, Kitajima T. Production of a biologically active epidermal growth factor fusion protein with high collagen affinity. J Biochem. 2001;129(4):627–633.

180. Ishikawa T, Eguchi M, Wada M, et al. Establishment of a functionally active collagen-binding vascular endothelial growth factor fusion protein in situ. Arterioscler Thromb Vasc Biol. 2006;26(9):1998–2004.

181. Sakiyama SE, Schense JC, Hubbell JA. Incorporation of heparin-binding peptides into fibrin gels enhances neurite extension: an example of designer matrices in tissue engineering. FASEB J. 1999;13(15):2214–2224.

182. Sakiyama-Elbert SE, Hubbell JA. Development of fibrin derivatives for controlled release of heparin-binding growth factors. J Control Release. 2000;65(3):389–402.

183. Zisch AH, Schenk U, Schense JC, Sakiyama-Elbert SE, Hubbell JA. Covalently conjugated VEGF—fibrin matrices for endothelialization. J Control Release. 2001;72(1–3):101–113.

184. Ichinose A, Tamaki T, Aoki N. Factor XIII-mediated cross-linking of NH2-terminal peptide of α2-plasmin inhibitor to fibrin. FEBS Lett. 1983;153(2):369–371.

185. Schense JC, Hubbell JA. Cross-linking exogenous bifunctional peptides into fibrin gels with factor XIIIa. Bioconjug Chem. 1998;10(1):75–81.

186. Sakiyama-Elbert SE, Panitch A, Hubbell JA. Development of growth factor fusion proteins for cell-triggered drug delivery. FASEB J. 2001;15(7):1300–1302.

187. Ehrbar M, Djonov VG, Schnell C, et al. Cell-demanded liberation of VEGF121 from fibrin implants induces local and controlled blood vessel growth. Circ Res. 2004;94(8):1124–1132.

188. Schmoekel HG, Weber FE, Schense JC, Gratz KW, Schawalder P, Hubbell JA. Bone repair with a form of BMP-2 engineered for incorporation into fibrin cell ingrowth matrices. Biotechnol Bioeng. 2005;89(3):253–262.

189. Lorentz KM, Yang L, Frey P, Hubbell JA. Engineered insulin-like growth factor-1 for improved smooth muscle regeneration. Biomaterials. 2012;33(2):494–503.

190. Geer DJ, Swartz DD, Andreadis ST. Biomimetic delivery of keratinocyte growth factor upon cellular demand for accelerated wound healing in vitro and in vivo. Am J Pathol. 2005;167(6):1575–1586.

191. Hu B-H, Messersmith PB. Rational design of transglutaminase substrate peptides for rapid enzymatic formation of hydrogels. J Am Chem Soc. 2003;125(47):14298–14299.

192. Ehrbar M, Rizzi SC, Schoenmakers RG, et al. Biomolecular hydrogels formed and degraded via site-specific enzymatic reactions. Biomacromolecules. 2007;8(10):3000–3007.

193. Ehrbar M, Rizzi SC, Hlushchuk R, et al. Enzymatic formation of modular cell-instructive fibrin analogs for tissue engineering. Biomaterials. 2007;28(26):3856–3866.

194. Agrawal V, Kishan KV. Promiscuous binding nature of SH3 domains to their target proteins. Protein Pept Lett. 2002;9(3):185–193.

195. Vulic K, Shoichet MS. Tunable growth factor delivery from injectable hydrogels for tissue engineering. J Am Chem Soc. 2012;134(2):882–885.

196. Beckett D, Kovaleva E, Schatz PJ. A minimal peptide substrate in biotin holoenzyme synthetase-catalyzed biotinylation. Protein Sci. 1999;8(4):921–929.

197. Chen I, Howarth M, Lin W, Ting AY. Site-specific labeling of cell surface proteins with biophysical probes using biotin ligase. Nat Methods. 2005;2(2):99–104.

198. Howarth M, Takao K, Hayashi Y, Ting AY. Targeting quantum dots to surface proteins in living cells with biotin ligase. Proc Natl Acad Sci USA. 2005;102(21):7583–7588.

199. Wylie RG, Ahsan S, Aizawa Y, Maxwell KL, Morshead CM, Shoichet MS. Spatially controlled simultaneous patterning of multiple growth factors in three-dimensional hydrogels. Nat Mater. 2011;10(10):799–806.

200. Leipzig ND, Wylie RG, Kim H, Shoichet MS. Differentiation of neural stem cells in three-dimensional growth factor-immobilized chitosan hydrogel scaffolds. Biomaterials. 2011;32(1):57–64.

201. Tam RY, Cooke MJ, Shoichet MS. A covalently modified hydrogel blend of hyaluronan–methyl cellulose with peptides and growth factors influences neural stem/progenitor cell fate. J Mater Chem. 2012;22(37):19402–19411.

202. Hartley RW. Barnase and barstar: two small proteins to fold and fit together. Trends Biochem Sci. 1989;14(11):450–454.

203. Schreiber G, Fersht AR. Interaction of barnase with its polypeptide inhibitor barstar studied by protein engineering. Biochemistry. 1993;32(19):5145–5150.

204. Ni X, Castanares M, Mukherjee A, Lupold SE. Nucleic acid aptamers: clinical applications and promising new horizons. Curr Med Chem. 2011;18(27):4206–4214.

205. Jayasena SD. Aptamers: an emerging class of molecules that rival antibodies in diagnostics. Clin Chem. 1999;45(9):1628–1650.

206. Ogawa A, Tomita N, Kikuchi N, Sando S, Aoyama Y. Aptamer selection for the inhibition of cell adhesion with fibronectin as target. Bioorg Med Chem Lett. 2004;14(15):4001–4004.

207. Daniels DA, Chen H, Hicke BJ, Swiderek KM, Gold L. A tenascin-C aptamer identified by tumor cell SELEX: systematic evolution of ligands by exponential enrichment. Proc Natl Acad Sci USA. 2003;100(26):15416–15421.

208. Hicke BJ, Marion C, Chang YF, Gould T, Lynott CK, Parma D. Tenascin-C aptamers are generated using tumor cells and purified protein. J Biol Chem. 2001;276(52):48644–48654.

209. Mi Z, Guo H, Russell MB, Liu Y, Sullenger BA, Kuo PC. RNA aptamer blockade of osteopontin inhibits growth and metastasis of MDA-MB231 breast cancer cells. Mol Ther. 2009;17(1):153–161.

210. Green LS, Jellinek D, Jenison R, Östman A, Heldin C-H, Janjic N. Inhibitory DNA ligands to platelet-derived growth factor B-chain. Biochemistry. 1996;35(45):14413–14424.

211. Potty ASR, Kourentzi K, Fang H, et al. Biophysical characterization of DNA aptamer interactions with vascular endothelial growth factor. Biopolymers. 2009;91(2):145–156.

212. Jellinek D, Lynott CK, Rifkin DB, Janjić N. High-affinity RNA ligands to basic fibroblast growth factor inhibit receptor binding. Proc Natl Acad Sci USA. 1993;90(23):11227–11231.

213. Ng EW, Shima DT, Calias P, Cunningham Jr ET, Guyer DR, Adamis AP. Pegaptanib, a targeted anti-VEGF aptamer for ocular vascular disease. Nat Rev Drug Discov. 2006;5(2):123–132.

214. Pagratis NC, Bell C, Chang YF, et al. Potent 2′-amino-, and 2′-fluoro-2′-deoxyribonucleotide RNA inhibitors of keratinocyte growth factor. Nat Biotechnol. 1997;15(1):68–73.

215. Jellinek D, Green LS, Bell C, et al. Potent 2′-amino-2′-deoxypyrimidine RNA inhibitors of basic fibroblast growth factor. Biochemistry. 1995;34(36):11363–11372.

216. Green LS, Jellinek D, Bell C, et al. Nuclease-resistant nucleic acid ligands to vascular permeability factor/vascular endothelial growth factor. Chem Biol. 1995;2(10):683–695.

217. Xiao J, Carter JA, Frederick KA, McGown LB. A genome-inspired DNA ligand for the affinity capture of insulin and insulin-like growth factor-2. J Sep Sci. 2009;32(10):1654–1664.

218. Kang J, Lee MS, Copland 3rd JA, Luxon BA, Gorenstein DG. Combinatorial selection of a single stranded DNA thioaptamer targeting TGF-beta1 protein. Bioorg Med Chem Lett. 2008;18(6):1835–1839.

219. McCauley TG, Kurz JC, Merlino PG, et al. Pharmacologic and pharmacokinetic assessment of anti-TGFbeta2 aptamers in rabbit plasma and aqueous humor. Pharm Res. 2006;23(2):303–311.

220. White RR, Roy JA, Viles KD, Sullenger BA, Kontos CD. A nuclease-resistant RNA aptamer specifically inhibits angiopoietin-1-mediated Tie2 activation and function. Angiogenesis. 2008;11(4):395–401.

221. White RR, Shan S, Rusconi CP, Shetty G, Dewhirst MW, Kontos CD. Inhibition of rat corneal angiogenesis by a nuclease-resistant RNA aptamer specific for angiopoietin-2. Proc Natl Acad Sci USA. 2003;100(9):5028–5033.

222. Zhang Z, Guo L, Tang J, Guo X, Xie J. An aptameric molecular beacon-based “signal-on” approach for rapid determination of rHuEPO-α. Talanta. 2009;80(2):985–990.

223. Orava EW, Jarvik N, Shek YL, Sidhu SS, Gariepy J. A short DNA aptamer that recognizes TNFalpha and blocks its activity in vitro. ACS Chem Biol. 2013;8(1):170–178.

224. Kubik MF, Bell C, Fitzwater T, Watson SR, Tasset DM. Isolation and characterization of 2′-fluoro-, 2′-amino-, and 2′-fluoro-/amino-modified RNA ligands to human IFN-gamma that inhibit receptor binding. J Immunol. 1997;159(1):259–267.

225. Ishiguro A, Akiyama T, Adachi H, Inoue J, Nakamura Y. Therapeutic potential of anti-interleukin-17A aptamer: suppression of interleukin-17A signaling and attenuation of autoimmunity in two mouse models. Arthritis Rheum. 2011;63(2):455–466.

226. Soontornworajit B, Zhou J, Shaw MT, Fan T-H, Wang Y. Hydrogel functionalization with DNA aptamers for sustained PDGF-BB release. Chem Commun. 2010;46(11):1857–1859.

227. Soontornworajit B, Zhou J, Zhang Z, Wang Y. Aptamer-functionalized in situ injectable hydrogel for controlled protein release. Biomacromolecules. 2010;11(10):2724–2730.

228. Battig MR, Soontornworajit B, Wang Y. Programmable release of multiple protein drugs from aptamer-functionalized hydrogels via nucleic acid hybridization. J Am Chem Soc. 2012;134(30):12410–12413.

229. Shangguan D, Li Y, Tang Z, et al. Aptamers evolved from live cells as effective molecular probes for cancer study. Proc Natl Acad Sci USA. 2006;103(32):11838–11843.

230. Chen N, Zhang Z, Soontornworajit B, Zhou J, Wang Y. Cell adhesion on an artificial extracellular matrix using aptamer-functionalized PEG hydrogels. Biomaterials. 2012;33(5):1353–1362.

231. Li S, Chen N, Zhang Z, Wang Y. Endonuclease-responsive aptamer-functionalized hydrogel coating for sequential catch and release of cancer cells. Biomaterials. 2013;34(2):460–469.

232. Zhang Z, Chen N, Li S, Battig MR, Wang Y. Programmable hydrogels for controlled cell catch and release using hybridized aptamers and complementary sequences. J Am Chem Soc. 2012;134(38):15716–15719.

233. Burdick JA, Murphy WL. Moving from static to dynamic complexity in hydrogel design. Nat Commun. 2012;3:1269.

234. Wilkinson AE, McCormick AM, Leipzig ND. Central nervous system tissue engineering: current considerations and strategies. Synthesis Lect Tissue Eng. 2011;3(2):1–120.

235. Katti DS, Lakshmi S, Langer R, Laurencin CT. Toxicity, biodegradation and elimination of polyanhydrides. Adv Drug Deliv Rev. 2002;54(7):933–961.

236. Tayebjee MH, Lip GY, Blann AD, Macfadyen RJ. Effects of age, gender, ethnicity, diurnal variation and exercise on circulating levels of matrix metalloproteinases (MMP)-2 and -9, and their inhibitors, tissue inhibitors of matrix metalloproteinases (TIMP)-1 and -2. Thromb Res. 2005;115(3):205–210.

237. Sahoo S, Chung C, Khetan S, Burdick JA. Hydrolytically degradable hyaluronic acid hydrogels with controlled temporal structures. Biomacromolecules. 2008;9(4):1088–1092.

238. Patterson J, Siew R, Herring SW, Lin AS, Guldberg R, Stayton PS. Hyaluronic acid hydrogels with controlled degradation properties for oriented bone regeneration. Biomaterials. 2010;31(26):6772–6781.

239. West JL, Hubbell JA. Photopolymerized hydrogel materials for drug delivery applications. React Polym. 1995;25(2–3):139–147.

240. Zhao X, Harris JMilton. Novel degradable poly(ethylene glycol) hydrogels for controlled release of protein. J Pharm Sci. 1998;87(11):1450–1458.

241. Elbert DL, Pratt AB, Lutolf MP, Halstenberg S, Hubbell JA. Protein delivery from materials formed by self-selective conjugate addition reactions. J Control Release. 2001;76(1–2):11–25.

242. Zustiak SP, Leach JB. Hydrolytically degradable poly(ethylene glycol) hydrogel scaffolds with tunable degradation and mechanical properties. Biomacromolecules. 2010;11(5):1348–1357.

243. Cho E, Kutty JK, Datar K, Soo Lee J, Vyavahare NR, Webb K. A novel synthetic route for the preparation of hydrolytically degradable synthetic hydrogels. J Biomed Mater Res A. 2009;90A(4):1073–1082.

244. Jo YS, Gantz J, Hubbell JA, Lutolf MP. Tailoring hydrogel degradation and drug release via neighboring amino acid controlled ester hydrolysis. Soft Matter. 2009;5(2):440–446.

245. Drury JL, Mooney DJ. Hydrogels for tissue engineering: scaffold design variables and applications. Biomaterials. 2003;24(24):4337–4351.

246. Sargeant TD, Desai AP, Banerjee S, Agawu A, Stopek JB. An in situ forming collagen–PEG hydrogel for tissue regeneration. Acta Biomater. 2012;8(1):124–132.

247. Ahmed TA, Griffith M, Hincke M. Characterization and inhibition of fibrin hydrogel-degrading enzymes during development of tissue engineering scaffolds. Tissue Eng. 2007;13(7):1469–1477.

248. Afify AM, Stern M, Guntenhoner M, Stern R. Purification and characterization of human serum hyaluronidase. Arch Biochem Biophys. 1993;305(2):434–441.

249. Oh EJ, Kang SW, Kim BS, Jiang G, Cho IH, Hahn SK. Control of the molecular degradation of hyaluronic acid hydrogels for tissue augmentation. J Biomed Mater Res A. 2008;86(3):685–693.

250. Nilasaroya A, Martens PJ, Whitelock JM. Enzymatic degradation of heparin-modified hydrogels and its effect on bioactivity. Biomaterials. 2012;33(22):5534–5540.

251. West JL, Hubbell JA. Polymeric biomaterials with degradation sites for proteases involved in cell migration. Macromolecules. 1998;32(1):241–244.

252. Mann BK, Gobin AS, Tsai AT, Schmedlen RH, West JL. Smooth muscle cell growth in photopolymerized hydrogels with cell adhesive and proteolytically degradable domains: synthetic ECM analogs for tissue engineering. Biomaterials. 2001;22(22):3045–3051.

253. Brandl FP, Seitz AK, Tessmar JK, Blunk T, Gopferich AM. Enzymatically degradable poly(ethylene glycol) based hydrogels for adipose tissue engineering. Biomaterials. 2010;31(14):3957–3966.

254. Kraehenbuehl TP, Zammaretti P, Van der Vlies AJ, et al. Three-dimensional extracellular matrix-directed cardioprogenitor differentiation: systematic modulation of a synthetic cell-responsive PEG-hydrogel. Biomaterials. 2008;29(18):2757–2766.

255. Ehrbar M, Sala A, Lienemann P, et al. Elucidating the role of matrix stiffness in 3D cell migration and remodeling. Biophys J. 2011;100(2):284–293.

256. Khetan S, Guvendiren M, Legant WR, Cohen DM, Chen CS, Burdick JA. Degradation-mediated cellular traction directs stem cell fate in covalently crosslinked three-dimensional hydrogels. Nat Mater. 2013;12(5):458–465.

257. Lévesque SG, Shoichet MS. Synthesis of enzyme-degradable, peptide-cross-linked dextran hydrogels. Bioconjug Chem. 2007;18(3):874–885.

258. Stephan MT, Moon JJ, Um SH, Bershteyn A, Irvine DJ. Therapeutic cell engineering with surface-conjugated synthetic nanoparticles. Nat Med. 2010;16(9):1035–1041.

259. Stephan MT, Stephan SB, Bak P, Chen J, Irvine DJ. Synapse-directed delivery of immunomodulators using T-cell-conjugated nanoparticles. Biomaterials. 2012;33(23):5776–5787.

260. Holden CA, Yuan Q, Yeudall WA, Lebman DA, Yang H. Surface engineering of macrophages with nanoparticles to generate a cell-nanoparticle hybrid vehicle for hypoxia-targeted drug delivery. Int J Nanomedicine. 2010;5:25–36.

261. Christian DA, Hunter CA. Particle-mediated delivery of cytokines for immunotherapy. Immunotherapy. 2012;4(4):425–441.

262. Morachis JM, Mahmoud EA, Almutairi A. Physical and chemical strategies for therapeutic delivery by using polymeric nanoparticles. Pharmacol Rev. 2012;64(3):505–519.

263. Serda RE. Particle platforms for cancer immunotherapy. Int J Nanomed. 2013;8:1683–1696.

264. Leong YS, Candau F. Inverse microemulsion polymerization. J Phys Chem. 1982;86(13):2269–2271.

265. Mitra S, Gaur U, Ghosh PC, Maitra AN. Tumour targeted delivery of encapsulated dextran-doxorubicin conjugate using chitosan nanoparticles as carrier. J Control Release. 2001;74(1–3):317–323.

266. Lee H, Mok H, Lee S, Oh YK, Park TG. Target-specific intracellular delivery of siRNA using degradable hyaluronic acid nanogels. J Control Release. 2007;119(2):245–252.

267. Braun O, Selb J, Candau F. Synthesis in microemulsion and characterization of stimuli-responsive polyelectrolytes and polyampholytes based on N-isopropylacrylamide. Polymer (Guildf). 2001;42(21):8499–8510.

268. Fernandez VVA, Tepale N, Sánchez-Díaz JC, Mendizábal E, Puig JE, Soltero JFA. Thermoresponsive nanostructured poly(N-isopropylacrylamide) hydrogels made via inverse microemulsion polymerization. Colloid Polym Sci. 2006;284(4):387–395.

269. Juraničová V, Kawamoto S, Fujimoto K, Kawaguchi H, Bartoň J. Inverse microemulsion polymerization of acrylamide in the presence of N,N-dimethylacrylamide. Die Angew Makromol Chem. 1998;258(1):27–31.

270. Bartoň J. Inverse microemulsion polymerization of oil-soluble monomers in the presence of hydrophilic polyacrylamide nanoparticles. Macromol Symp. 2002;179(1):189–208.

271. Kaneda I, Sogabe A, Nakajima H. Water-swellable polyelectrolyte microgels polymerized in an inverse microemulsion using a nonionic surfactant. J Colloid Interface Sci. 2004;275(2):450–457.

272. Gaur U, Sahoo SK, De TK, Ghosh PC, Maitra A, Ghosh PK. Biodistribution of fluoresceinated dextran using novel nanoparticles evading reticuloendothelial system. Int J Pharm. 2000;202(1–2):1–10.

273. Bharali DJ, Sahoo SK, Mozumdar S, Maitra A. Cross-linked polyvinylpyrrolidone nanoparticles: a potential carrier for hydrophilic drugs. J Colloid Interface Sci. 2003;258(2):415–423.

274. Missirlis D, Tirelli N, Hubbell JA. Amphiphilic hydrogel nanoparticles preparation, characterization, and preliminary assessment as new colloidal drug carriers. Langmuir. 2005;21(6):2605–2613.

275. Oh JK, Perineau F, Matyjaszewski K. Preparation of nanoparticles of well-controlled water-soluble homopolymers and block copolymers using an inverse miniemulsion ATRP. Macromolecules. 2006;39(23):8003–8010.

276. Oh JK, Tang C, Gao H, Tsarevsky NV, Matyjaszewski K. Inverse miniemulsion ATRP: a new method for synthesis and functionalization of well-defined water-soluble/cross-linked polymeric particles. J Am Chem Soc. 2006;128(16):5578–5584.

277. Genggeng Q, Christopher WJ, Schork FJ. RAFT inverse miniemulsion polymerization of acrylamide. Macromol Rapid Commun. 2007;28(9):1010–1016.

278. Oh JK, Dong H, Zhang R, Matyjaszewski K, Schlaad H. Preparation of nanoparticles of double-hydrophilic PEO-PHEMA block copolymers by AGET ATRP in inverse miniemulsion. J Polym Sci A Polym Chem. 2007;45(21):4764–4772.

279. An Z, Shi Q, Tang W, Tsung CK, Hawker CJ, Stucky GD. Facile RAFT precipitation polymerization for the microwave-assisted synthesis of well-defined, double hydrophilic block copolymers and nanostructured hydrogels. J Am Chem Soc. 2007;129(46):14493–14499.

280. An Z, Tang W, Wu M, Jiao Z, Stucky GD. Heterofunctional polymers and core-shell nanoparticles via cascade aminolysis/Michael addition and alkyne-azide click reaction of RAFT polymers. Chem Commun (Camb) 2008;(48):6501–6503.

281. Shen W, Chang Y, Liu G, Wang H, Cao A, An Z. Biocompatible, antifouling, and thermosensitive core−shell nanogels synthesized by RAFT aqueous dispersion polymerization. Macromolecules. 2011;44(8):2524–2530.

282. Rieger J, Grazon C, Charleux B, Alaimo D, Jérôme C. Pegylated thermally responsive block copolymer micelles and nanogels via in situ RAFT aqueous dispersion polymerization. J Polym Sci A Polym Chem. 2009;47(9):2373–2390.

283. Rider CC. Heparin/heparan sulphate binding in the TGF-beta cytokine superfamily. Biochem Soc Trans. 2006;34(Pt 3):458–460.

284. Park JE, Keller GA, Ferrara N. The vascular endothelial growth factor (VEGF) isoforms: differential deposition into the subepithelial extracellular matrix and bioactivity of extracellular matrix-bound VEGF. Mol Biol Cell. 1993;4(12):1317–1326.

285. Ferrara N. Binding to the extracellular matrix and proteolytic processing: two key mechanisms regulating vascular endothelial growth factor action. Mol Biol Cell. 2010;21(5):687–690.

286. Raines EW, Ross R. Compartmentalization of PDGF on extracellular binding sites dependent on exon-6-encoded sequences. J Cell Biol. 1992;116(2):533–543.

287. Rolny C, Spillmann D, Lindahl U, Claesson-Welsh L. Heparin amplifies platelet-derived growth factor (PDGF)- BB-induced PDGF alpha -receptor but not PDGF beta-receptor tyrosine phosphorylation in heparan sulfate-deficient cells Effects on signal transduction and biological responses. J Biol Chem. 2002;277(22):19315–19321.

288. Lyon M, Deakin JA, Mizuno K, Nakamura T, Gallagher JT. Interaction of hepatocyte growth factor with heparan sulfate Elucidation of the major heparan sulfate structural determinants. J Biol Chem. 1994;269(15):11216–11223.

289. Ohkawara B, Iemura S, ten Dijke P, Ueno N. Action range of BMP is defined by its N-terminal basic amino acid core. Curr Biol. 2002;12(3):205–209.

290. Ruppert R, Hoffmann E, Sebald W. Human bone morphogenetic protein 2 contains a heparin-binding site which modifies its biological activity. Eur J Biochem. 1996;237(1):295–302.

291. Irie A, Habuchi H, Kimata K, Sanai Y. Heparan sulfate is required for bone morphogenetic protein-7 signaling. Biochem Biophys Res Commun. 2003;308(4):858–865.

..................Content has been hidden....................

You can't read the all page of ebook, please click here login for view all page.
Reset
3.144.31.163