Fiber–Hydrogel Composites for Wound Healing: History
Please note this is an old version of this entry, which may differ significantly from the current revision.
Subjects: Polymer Science
Contributor:

The structural resemblance of fiber–hydrogel composites to natural tissues has been a driving force for the optimization and exploration of these systems in biomedicine. Indeed, the combination of hydrogel-forming techniques and fiber spinning approaches has been crucial in the development of scaffolding systems with improved mechanical strength and medicinal properties.

  • fiber–hydrogel composite
  • biodegradable polymers
  • skin regeneration
  • drug delivery platforms
  • controlled release

1. Introduction

Biomaterials are defined as nonviable materials, with potential for applications in medical devices, that possess the ability to interact with biological systems to evaluate, treat, replace or enhance the performance of any tissue [1]. Biomaterials are classified in different ways; the most common refers to their chemical nature and is subdivided in metallic materials (ferrous and non244-ferrous) and non-metallic materials (organic: polymers, biological materials, and carbons; and inorganic: ceramics and glasses). Composites are considered another very important class of biomaterials and result from the combination of two classes of materials that work in synergy to improve the properties of the final product above those of the individual components [1][2].

The continued research in this field has raised the specificity level of the biomaterials developed and, therefore, has increased its impact in the healthcare global market [1]. Polymers represent a large portion of all biomaterials used in the biomedical field (about 45%) [2], and their application appears to have no end. They can be processed in the form of particles, foams, films, membranes, hydrogels and fibers, and combinations of these 3D structures can then be made to generated intricate, target-direct, specialized biomedical systems. Biomedicine has resorted to these constructs to understand specific biological processes and to engineer high-performance therapies to treat a variety of diseases. The need to match the desired functions/characteristics of a given tissue or cell has driven the combination of different classes of biomaterials in complex constructs (e.g., fiber–hydrogel composite) that can effectively respond to the local demands and provide the necessary tools to reach the desired goals. In recent years, fiber–hydrogel composites have been disclosed as one of those systems that combine different structures to improve individual features and enhance inherent advantages to achieve successful outcomes. In biomedical engineering, the importance of these constructs is particularly noticeable in wound healing and drug delivery. In both areas, fiber–hydrogel composites can be a good alternative to the use of antibiotics and/or their controlled administration.

2. Applications of Fiber–Hydrogel Composites

Recently, research on fiber–hydrogel composites has increased significantly. These scaffolding systems have been investigated for a range of applications, including wound healing [3], regeneration of corneal stroma [4], nucleus pulposus regeneration [5], bone tissue engineering [6], antibiotic delivery [7] and heart valve tissue engineering [8]. Scaffolds with an architecture that mimics native ECM and allows cell infiltration and differentiation has emerged as a prospective solution for the treatment of various health complications. For instance, the fibrous structure in fiber–hydrogel composites is considered of enormous importance for a greater efficiency of the scaffold. This is because tissues have biological fibers with specific composition and architecture that contribute to the normal function of the tissues. Thus, with this, it is possible to simulate biological fibers, approximating the foreign scaffold to living tissue and, thus, enhance cellular growth and maturation (e.g., cell differentiation) [4]. In the following sections, we will contextualize and list in more detail some recent examples of fiber–hydrogel composites applied in wound healing and drug delivery.

2.1. Wound Healing

Wound healing is a complex physiological response that involves a cascade of cells, matrix components and other biological factors [9]. In healthy people, wound healing includes four important phases: hemostasis, inflammation, proliferation, and remodeling. This complex process allows skin functions to be restored. Wounds that fail the normal healing process in a predictable amount of time are considered chronic wounds (CW) [10][11]. Currently, wound care is based on the application of a wide variety of wound dressings (gauzes, absorbent cotton and bandages), debridement, vacuum assisted closure and grafts. Even though they are considered the therapy of choice, wound dressing have some limitations, they are incapable of maintaining the moist environment necessary for wound healing and tend to adhere to the wound, which may cause discomfort to the patient when the dressing is removed [12]. CW treatments are often associated to high economic costs, an increase in surgical procedures and the greater susceptibility of the patient to infection. Microorganisms such as Acinetobacter baumannii, Enterococcus faecalis, Pseudomonas aeruginosa and Staphylococcus aureus have the ability to colonize and infect wounds, which complicate the healing process [13][10]. In the most severe cases, patients with infected wounds, such as diabetic foot infections, include mainly antibiotics in their therapy [14]. The impact of excess and inappropriate use of antibiotics has been explored in relation to the various adverse effects, such as bacterial resistance, which has been highlighted as a serious global concern [15]. Several alternatives have been developed for a more efficient wound healing in order to prevent infection to evolve and, in the case of CW, to try and shorten the treatment period [12][16][17][18][19]. There are some properties that ideally a modern wound dressing should have, specifically, the capacity for mechanical protection and adaptation to the shape of the wound, without adhering to wound tissue per se, so as not to cause pain to the patient when removed. Absorption capacity, cytocompatibility, flexibility, ability to ensure a balanced moist environment, induce wound healing, facilitate ECM regeneration, protect the wound from external contaminants and promote debridement are also important features in the development of an effective wound dressing [13][20][21][12]. Wound dressings can be classified based on the affinity of the dressing with the wound into four distinct groups: passive, interactive, advanced and smart dressings [11]. Modern dressings take the most varied forms, including hydrogels, films, sponges, foams, nanofiber mats and, more recently, fiber–hydrogel composites [20][3]. The hydrogel has the ability to absorb exudates and maintain a balance of moisture at the wounded site. In turn, the fiber mimics the fibrous structure of ECM. Since both structures present limitations, the fibers do not facilitate cell migration and hydrogels have low mechanical stability, scaffolds combining both have been the research target of many investigations in order to uncover alternatives for the treatment of wounds [3][19][22]. The combination of the two structure in one scaffold is expected to facilitate healing by generating an environment conducive with cell recognition and attachment (ECM mimicking) with a moist and breathable atmosphere required for a healthy tissue formation. It is known that a large part of mammalian ECM has an aqueous matrix (gel) containing diverse fibrous proteins, essentially collagen, elastin and fibronectin. These proteins surround and guide cells in vivo and act as an anchoring matrix [23][24]. In humans, fibrillar collagen provides tensile strength for ECM, which limits tissue/organ distensibility as is the case of the skin [25]. The ECM is mainly responsible for cell adhesion, migration, proliferation, and regulation of their action. For a complete and effective skin regeneration, it is important that a scaffold is created that mimics the structure and normal skin conditions. Studies have shown that the reinforcement of hydrogels with fibers improves cell function, differentiation and proliferation, as well as structural stability [26][27][28]. Indeed, Schulte et al. described the manufacture of an artificial ECM scaffold consisting of biofunctionalized fibers incorporated in a semi-synthetic hydrogel of HA that allowed the control of cell adhesion [24].

There are several polymers used in fiber–hydrogel composites, namely gelatin [3][19][29]. The combination of two separate scaffolds (bilayer scaffold) was studied by Franco et al. for a possible application in skin regeneration. The formulation consisted of a first layer based on a PCL/PLGA membrane (80:20) formed by electrospinning and a second layer of CS/gelatin hydrogel (50:50) crosslinked with glutaraldehyde. The first layer showed excellent mechanical properties and biocompatibility. In the case of the second layer, they obtained a porous structure, capable of swelling more than 500% of its dry size (excellent absorbent properties). The junction of the fibrous membranes provided better mechanical support to the scaffold and, at the same time, reduced the rate of degradation of the layer formed by the hydrogel [29]. In the same light, Zhao et al. through a chemical reaction of the methacrylamide groups with gelatin formed a prepolymer to produce fibers by electrospinning (GelMA). The electrospun GelMA nanofibers were crosslinked by photo-crosslinking, with UV radiation. By manipulating the degree of modification of the gelatin with the methacrylamide groups and the photo-crosslinking time, it is possible to adjust the physical and biological properties. Characteristics such as water vapor permeability, water retention, mechanical resistance and kinetic degradation can be adapted by adjusting the time of UV light radiation. These GelMA scaffolds, which mimic the structure of the native ECM, demonstrated a better orientation of the cellular processes (e.g., cell migration of fibroblasts) and biocompatibility compared to the controls (gelatin and PLGA). The in vivo tests reinforce the potential of this scaffold since it was visible that they accelerated wound repair [19]. Sun et al. went a step further and reported the ability of the GelMA to improve the elastic biodegradable mechanical properties of the construct and its ability to improve cell adhesion, proliferation and vascularization [30]. In turn, Li et al. reports the use of gelatin for the development of a hydrogel fibers. Initially the gelatin-based compound hydrogel fibers were prepared by gel-spinning with PEG6000. Subsequently, the crosslinking agent dialdehyde carboxymethyl cellulose (DCMC) was incorporated in order to improve the thermal and mechanical properties of the hydrogel fibers composed of gelatin-PEG. This scaffold showed a strong capacity to absorb free water due to its 3D structure and porous network. The higher the DCMC content in hydrogel fibers, the more slowly they degrade. In addition, DCMC increased the compatibility of the hydrogel fibers with blood [3]. HA nanofibers are reported to promote wound healing. Due to their high solubility in water, crosslinking is required to increase their water stability. Chen et al. developed an electrospun a mixture of maleicated hyaluronate/poly(vinyl alcohol) methacrylate (MHA/MaPVA) that allowed the formation of mats with the capacity to swell and form fibrous hydrogels. The weight ratio of the nanofiber components influenced the morphology and diameter of the nanofibers. This structure was cytocompatible, promoted cell fixation and displayed high water absorption capacities [22]. PVA has also been combined with PCL to form double layer structures resultant from the combination of PCL nanofibers (hydrophobic) and PVA hydrogel (hydrophilic). After exposure to water, the PVA fiber layer was completely dissolved, and a hydrogel-like structure was formed. Despite this change, the defined shape of the scaffold was maintained due to the stability of the PCL layer in water-based environments. Several aspects were tested in this scaffold, namely, its morphology, wettability, and adhesion and proliferation of mouse fibroblasts. Here, it was seen that fibroblasts exhibited greater proliferative activity on the PCL side of the double layer. In the case of the PVA layer, the same was not seen, which may be a consequence of the greater hydrophilicity of the layer. Based on the behavior and characteristics of the double layer scaffold, the authors concluded that the scaffold had the potential to be used as a dressing or in the prevention of abdominal adhesions [31].

The rapid dissolution of fibers in an aqueous medium becomes a limitation for their application in active wound dressings. In the case of PVP fibers, their rapid solubility remains a problem despite their self-adhesive properties and their ability to incorporate molecules. Recently, to overcome this limitation Contardi et al. proposed to develop PVP-based fiber hydrogels containing hydroxycinnamic acid derivatives. A controlled release of p-cumaric and ferulic acids (derived from hydroxycinnamic acid) from the fibers was observed due to the incorporation of these in the hydrogel. The author also observed in burned skin a reduction in the levels of enzymes known to be positively regulated by reactive oxidative species in burned skin [32]. By electrospinning/electrospraying methods, Azarniya et al. reported the production of a hybrid fiber–hydrogel by combining fibrous mats and hydrogel particles. Through electrospinning, keratin/bacterial cellulose (BC) fibers were produced and simultaneously sprayed with thermosensitive hydrogel particles. The chemically crosslinked hydrogel was composed of non-ionic triblock copolymers (PEO99-PPO65-PEO99; Pluronic F127) conjugated with Tragacanto gum (TG). Due to the low spinning power of keratin, poly(oxide of ethylene) (PEO) was added to the formulation forming the keratin/BC/PEO fibers. Reductions in the diameter of keratin/PEO fibers from 243 ± 57 nm to 150 ± 43 nm and hydrophobicity were observed with the addition of 1% or more of BC. However, despite the reduction of pores, TG and BC modified mats promoted cell fixation and proliferation in fibrous structures. It was seen that the hydrogel particles were uniformly incorporated into the junction of the fibrous network. This modification improved several features of the scaffolds, including hydrophilicity, modulus of elasticity (31%), tensile strength (35%) and ductility (23%) [33]. More recently, Loo et al. developed “intelligent” peptide hydrogels, in which the short aliphatic peptides had the tendency to self-assemble into helical fibers, forming nanofiber hydrogels. These nanofibrous hydrogels were found to possess regenerative properties and to display potential to accelerate the healing of burn wounds [34].

2.2. Drug Delivery

In conventional therapies, rapid degradation and excretion of drugs during the circulation process in the body is frequently detected. Consequently, only a small amount of medication will have therapeutic effects in places of interest [35]. Several research groups have focused on the development of new controlled drug delivery systems to allow an effective distribution of drugs in the intended locations at a controlled release rate [36][37]. A drug delivery system is used to transport therapeutic substances in the body more effectively and safely, having the ability to control the amount, the time and the targeted place for drug release [38]. Several scaffolds have been used to encapsulate and deliver therapeutic drugs, namely, fibers and hydrogels [39][40][41][42].

Electrospinning systems allow drugs to be incorporated into the fibers, giving them a high drug loading capacity, increased initial burst, sustained release, and prolonged circulation. Methods of incorporation include blend (or co-, the drug is mixed in the polymer solution), side-by-side (vehicle/polymer solution and the biomolecules are loaded in a separate spinneret), multi-jet (use of multiple nozzles with one or more jets, or a nozzle with different jets), co-axial (two concentric aligned capillaries connected to a high voltage source) and emulsion electrospinning (the drug is encapsulated in an appropriate solvent to be protected from the fiber/solvent system) [43]. Just as there are different ways to incorporate drugs into fibers, drugs may also be released via three distinct mechanisms: desorption of the fiber surface, diffusion in the solid state through the fibers, and fiber degradation [44]. The fiber morphology and its high therapeutic load capacity are beneficial properties that make them potential candidates for drug delivery systems. Electrospun fibers have several advantages especially due to their large surface area and their absorption/release properties [45]. However, large-burst drug release, uncontrolled duration of drug release, and incomplete drug release are recurring problems. The possible agglomeration of bioactive agents on the surface of the fibers becomes a disadvantage of the electrospinning method since it can trigger an initial burst release, which may cause toxicity of the release site [43][32][46]. Such limitations may have implications in the scaffold biomedical goals.

To incorporate drugs into hydrogels, they can be loaded into the precursor solutions before crosslinking or can be absorbed after gelation [47]. Regarding drug release, swelling is an important property in some stimulus-sensitive drug delivery system. Certain changes in the environment may trigger swelling that allows the release of the drug due to the alterations in mesh size of the polymeric network [48]. Features like hydrophilicity, biocompatibility and tunable mechanical properties are the reason why hydrogels have been used extensively for the controlled release of drugs [36]. Although hydrogels are widely used in controlled release systems, there are some limitations that must be overcome. These scaffolds suffer from low mechanical resistance, which may be responsible for inhomogeneous release [47]. In most hydrogels, their ability to absorb large amounts of water and the presence of large pore sizes may trigger a rapid drug release [7]. In accordance, some investigations have developed/obtained better kinetic release profiles when there is a combination of hydrogels with other structures, namely fibers [36]. The effectiveness of fiber–hydrogel composites for drug administration has been demonstrated [7][35][37]. Nanofiber–hydrogel scaffolds as biofunctionalized platforms appear as attractive alternatives to the ineffective treatments related to direct drug administration.

Persistent neurological dysfunctions are usually triggered by spinal cord injuries due to failure in axon regeneration. Nguyen et al. synthesized lined mats of poly(ε-caprolactone-co-ethyl ethylene phosphate) (PCLEEP) by electrospinning and distributed them in a collagen hydrogel matrix. Both the fibers and the hydrogel contained neurotrophin-3 (model protein) known for promoting neuronal survival, axonal sprouting and regeneration. Additionally, the hydrogel contained miR-222 (model microRNA) known to contribute to the control of local protein synthesis at distal axons. Overtime, it was seen that degradation occurred within the collagen hydrogel, but the PCLEEP fibers maintained their morphology and alignment after 3 months. The composite framework allowed localized and sustained drug/gene delivery, while aligned nanofibers acted to direct remyelination of the injured area. Furthermore, they observed the regeneration of the animal model axon [35]. In a similar study, small fragmented nanofibers of poly(3-caprolactone-co-D,L-lactide) (PCL:DLLA) and collagen were individually dispersed in a hyaluronane-methylcellulose hydrogel (HAMC). These fiber–hydrogel composites were used as a cell-transport system multipotent neural/progenitor stem cells (NSPCs) for the treatment of spinal cord injuries. The results showed that the incorporation of fibers in the HAMC hydrogel influenced the behavior of the NSPC cells, highlighting a better neuronal and oligodendrocytic differentiation in the scaffold PCL:DLLA/HAMC compared to collagen/HAMC [49]. In both studies, the complex generated from the combination of fibers and hydrogels allowed for a faster cell development and consequent regeneration.

A laminated fiber–hydrogel composite based on PCL electrospun fiber mats coupled with poly(ethylene glycol)-poly(ε-caprolactone) diacrylate (PEGPCL) hydrogels processed by UV polymerization was developed to control the release of a model hydrophilic protein (e.g., bovine albumin serum, BSA). To study the release of the hydrophilic protein, BSA was added to the system before crosslinking. The results reported by Han et al. suggested the relevant role of PLC fibers (diameter of approximately 0.45 μm) in the release of the drug in a uniform and delayed manner, by reducing swelling of hydrogels and water penetration rates and by increasing the length of the diffusion path and the diffusivity of the drug. In addition, the bioactivity of proteins after release was proven since extension of PC12 cell neuritis was detected. In general, the PCL fibers in the PEGPCL hydrogel demonstrated an important role in three main areas: control of the release kinetics of the hydrophilic protein, reduction of burst release (initial) and increased duration of drug release (more than two months) [36].

Osteomyelitis is a bone disease caused mainly by methicillin-resistant Staphylococcus aureus (MRSA). Various antibiotics are administered to reduce this infection, namely the glycopeptide vancomycin hydrochloride (vanco-HCl). The bacterial plaque that forms around the infected area limits treatment by preventing the diffusion of the antibiotic vanco-HCL to the infected site, which then requires the administration of high doses. This overuse of antibiotics in addition to their impropriate function can lead to systemic toxicity. To try and solve this problem, Ahadi et al. developed a scaffold made of poly(L-lactide) (PLLA) fibers produced by electrospinning followed by aminolysed, encased in a hydrogel of silk fibroin/oxidized pectin. PLLA fibers were loaded with vanco-HCl to promote a more sustainable release of the antibiotic at the affected site, resulting in a 61% reduction in drug release. This scaffold revealed better mechanical properties compared to the single hydrogel (without fibers), namely, a higher crosslinking density (52%), a higher compression module (30%) and a lower expansion rate (15%). Biologically, the fiber–hydrogel composite was seen to have activity against MRSA and to be cytocompatible with cells, largely due to the presence of fibers aminolized with drugs [7]. Ekaputra et al. developed by electrospinning/electrospraying a hybrid mesh of PCL/collagen and HA hydrogel, HeprasilTM, loaded with vascular endothelial growth factors (VEGF) and platelet-derived growth factors (PDGF). It was seen that the fiber–hydrogel composite PCL/collagen-Heprasil was successful in allowing a double simultaneous loading of the growth factors VEGF165 and PDGF-BB and to promote their controlled release over a period of five weeks, in vitro [50]. Recently, biocompatible vehicles for the release of the crystal violet drug (CV) have also been described, in which polydopamine microfibers (PDA) were incorporated in a pullulan (PHG) hydrogel crosslinked by poly(ethylene glycol) diglicidyl ether (chemical crosslinker). PDA fibers attributed the pH-responsive drug release behavior to the PHG hydrogel. This happens in response to the acidic conditions, which increase the electrostatic repulsion force between the PDA (protonated and positively charged) and the drug CV (positive charge). This repulsion promotes the release of the drug, with a detectable a cumulative release of 60.3% (pH 7.4), which increased to 87% with a decrease in pH to 5. In addition, the incorporation of PDA fibers and the adjustment of their content allowed to regulate several properties of the composite PHG-PDAs, namely, its viscoelastic characteristics, mechanical performance, mesh size and swelling/disintegration properties of the PHG hydrogel. The developed scaffold proved to have great potential to be used in drug delivery systems, given its good cytocompatibility, non-toxicity and easily adjustable properties for a controlled release of CV [51]. Overall, data demonstrated the ability of the engineered systems to promote a controlled drug delivery, in which the fibrous mesh guaranteed the mechanical stability of the construct while the hydrogel released the loaded active compounds.

A new physical approach based on hydrogel and nanofibers (or NEEDs) for cell encapsulation has been described in the work of An et al. Here, tubular constructs with different compartments were developed, consisting of Nylon 6,6 nanofibers, manufactured by electrospinning, being subsequently impregnated in different hydrogel precursor solutions (alginate, chitosan or collagen) and crosslinked. Fibers had an average diameter of 200 nm with 1 μm interconnected pores. Compartmentation proved to be an asset for co-encapsulation, co-culture and co-distribution of different individual cells and cellular aggregates (islets), with cell viability being observed. Finally, the potential application of NEEDs for cell therapies using a type 1 diabetic model was tested, and the disease was corrected (in 8 weeks), which proved the therapeutic potential of NEEDs within primary rat islets (without the disease) [37].

In wound healing, it is important to pay attention to the biomaterials used to produce wound dressing. To achieve the desired objectives, the properties of each biomaterial are optimally combined. Studies have shown that local administration of therapeutic agents through wound dressings can improve the wound healing process [9]. In fact, a bioactive dressing of fibers of silk fibroin (SF) produced via electrospinning was developed and then combined with the alginate hydrogel (ALG) capable of supplying amniotic fluid (AF). This dressing had the ability to release AF, highly enriched with various therapeutic agents, at the wound site. The AF release profile was related to the concentration of ALG (greater release of AF in lower amounts of ALG). The increase in cell proliferation and collagen dissemination and secretion due to AF in fibroblast cultures strengthens the potential of the SF/ALG fiber–hydrogel composite to accelerate the healing process in severe wounds [52]. In a similar study, a bi-layer dressing of gelatin nanofiber mats loaded with epigallocatechin gallate (EGCG)/PVA hydrogel was produced for the treatment of acute wounds. The hydrogel was used as a protective and hydrating outer layer of the bi-layer dressing. Jaiswal el al. observed that the decrease in crosslinking time led to a slower EGCG release profile. This increased in the 2-4 days release period demonstrating the ability of this scaffold to guarantee a gradual drug release. Faster wound contraction, improvement in angiogenesis, reepithelization and less inflammatory response compared to control were also observed [53]. More recently, Chen et al. developed CS/gelatin hydrogels with polydopamine-intercalated silicate nanoflakes (PDA-Silicate). These were electrospun in the form of nanofibers loaded with the antibiotic tetracycline hydrochloride (TH). In this sandwich-like nanofiber/hydrogel composite (NF-HG) the incorporation of the fibers in the hydrogel resulted in a restriction in the release of antibiotic TH. However, it allowed a sustained release rate of TH in NF-HG for long-term protection. In addition, this structure reduced the toxicity of the drug associated to the rapid release. Furthermore, the excellent adhesiveness and anti-infectious properties demonstrated by the NF-HG, turned this formulation particularly attractive to be used as a wound dressing [54].

This entry is adapted from the peer-reviewed paper 10.3390/antibiotics10030248

References

  1. Ratner, B.D.; Hoffman, A.S.; Schoen, F.J.; Lemons, J.E.; Wagner, W.R.; Sakiyama-Elbert, S.E.; Zhang, G.; Yaszemski, M.J. Introduction to Biomaterials Science: An Evolving, Multidisciplinary Endeavor. In Biomaterials Science: An Introduction to Materials in Medicine; Wagner, W., Sakiyama-Elbert, S., Zhang, G., Yaszemski, M., Eds.; Academic Press: Cambridge, MA, USA, 2020; pp. 3–19.
  2. Mariani, E.; Lisignoli, G.; Borzì, R.M.; Pulsatelli, L. Biomaterials: Foreign Bodies or Tuners for the Immune Response? Int. J. Mol. Sci. 2019, 20, 636.
  3. Li, D.; Ye, Y.; Li, D.; Li, X.; Mu, C. Biological properties of dialdehyde carboxymethyl cellulose crosslinked gelatin-PEG composite hydrogel fibers for wound dressings. Carbohydr. Polym. 2015, 137, 508–514.
  4. Kong, B.; Chen, Y.; Liu, R.; Liu, X.; Liu, C.; Shao, Z.; Xiong, L.; Liu, X.; Sun, W.; Mi, S. Fiber reinforced GelMA hydrogel to induce the regeneration of corneal stroma. Nat. Commun. 2020, 11, 1435.
  5. Thorvaldsson, A.; Silva-Correia, J.; Oliveira, J.M.; Reis, R.L.; Gatenholm, P.; Walkenström, P. Development of Nanofiber-Reinforced Hydrogel Scaffolds for Nucleus Pulposus Regeneration by a Combination of Electrospinning and Spraying Technique. J. Appl. Polym. Sci. 2013, 128, 1158–1163.
  6. Patel, M.; Koh, W.-G. Composite Hydrogel of Methacrylated Hyaluronic Acid and Fragmented Polycaprolactone Nanofiber for Osteogenic Differentiation of Adipose-Derived Stem Cells. Pharmaceutics 2020, 12, 902.
  7. Ahadi, F.; Khorshidi, S.; Karkhaneh, A. A hydrogel/fiber scaffold based on silk fibroin/oxidized pectin with sustainable release of vancomycin hydrochloride. Eur. Polym. J. 2019, 118, 265–274.
  8. Eslami, M.; Vrana, N.E.; Zorlutuna, P.; Sant, S.; Jung, S.; Masoumi, N.; Ramazan Ali, Khavari-Nejad; Javadi, G.; Khademhosseini, A. Fiber-reinforced hydrogel scaffolds for heart valve tissue engineering. J. Biomater. Appl. 2014, 29, 399–410.
  9. Negut, I.; Dorcioman, G.; Grumezescu, V. Scaffolds for Wound Healing Applications. Polymers 2020, 12, 2010.
  10. Goldberg, S.R.; Diegelmann, R.F. What Makes Wounds Chronic. Surg. Clin. N. Am. 2020, 100, 681–693.
  11. Ambekar, R.S.; Kandasubramanian, B. Advancements in nanofibers for wound dressing: A review. Eur. Polym. J. 2019, 117, 304–336.
  12. Radhakumary, C.; Antonty, M.; Sreenivasan, K. Drug loaded thermoresponsive and cytocompatible chitosan based hydrogel as a potential wound dressing. Carbohydr. Polym. 2011, 83, 705–713.
  13. Ribeiro, D.M.L.; Júnior, A.R.C.; de Macedo, G.H.R.V.; Chagas, V.L.; Silva, L.D.S.; da Silva Cutrim, B.; Santos, D.M.; Soares, B.L.L.; Zagmignan, A.; de Càssia Mendonça de Miranda , R.; et al. Polysaccharide-Based Formulations for Healing of Skin-Related Wound Infections: Lessons from Animal Models and Clinical Trials. Biomolecules 2020, 10, 1–16.
  14. Abbas, M.; Uçkay, I.; Lipsky, B.A. In diabetic foot infections antibiotics are to treat infection, not to heal wounds. Expert Opin. Pharmacother. 2015, 16, 821–832.
  15. Tavares, T.D.; Antunes, J.C.; Ferreira, F.; Felgueiras, H.P. Biofunctionalization of natural Fiber-Reinforced Biocomposites for Biomedical Applications. Biomolecules 2020, 10, 148.
  16. Rubio-Elizalde, I.; Bernáldez-Sarabia, J.; Moreno-Ulloa, A.; Vilanova, C.; Juárez, P.; Licea-Navarro, A.; Castro-Cesenã, A.B. Scaffolds based on alginate-PEG methyl ether methacrylate-Moringa oleifera-Aloe vera for wound healing applications. Carbohydr. Polym. 2018, 206, 455–467.
  17. Joseph, B.; Augustine, R.; Kalarikkal, N.; Thomas, S.; Seantier, B.; Grohens, Y. Recent advances in electrospun polycaprolactone based scaffolds for wound healing and skin bioengineering applications. Mater. Today Commun. 2019, 19, 319–335.
  18. Felgueiras, H.P.; Teixeira, M.A.; Tavares, T.D.; Homem, N.C.; Zille, A.; Amorim, M.T.P. Antimicrobial action and clotting time of thin, hydrated poly(vinyl alcohol )/cellulose acetate fi lms functionalized with LL37 for prospective wound-healing applications. J. Appl. Polym. Sci. 2019, 138, 48626.
  19. Zhao, X.; Sun, X.; Yildirimer, L.; Lang, Q.; Lin, Z.Y.; Zheng, R.; Zhang, Y.; Cui, W.; Annabi, N.; Khademhosseini, A. Cell infiltrative hydrogel fibrous scaffolds for accelerated wound healing. Acta Biomater. 2016, 49, 66–77.
  20. Teixeira, M.A.; Paiva, M.C.; Amorim, M.T.P.; Felgueiras, H.P. Electrospun Nanocomposites Containing Cellulose and Its Derivatives Modified with Specialized Biomolecules for an Enhanced Wound Healing. Nanomaterials 2020, 10, 557.
  21. Felgueiras, H.P.; Amorim, M.T.P. Functionalization of electrospun polymeric wound dressings with antimicrobial peptides. Colloids Surfaces B Biointerfaces 2017, 156, 133–148.
  22. Chen, X.; Lu, B.; Zhou, D.; Shao, M.; Xu, W.; Zhou, Y. Photocrosslinking maleilated hyaluronate/methacrylated poly(vinyl alcohol) nanofibrous mats for hydrogel wound dressings. Int. J. Biol. Macromol. 2019, 155, 903–910.
  23. Lutolf, M.P.; Hubbell, J.A. Synthetic biomaterials as instructive extracellular microenvironments for morphogenesis in tissue engineering. Nat. Biotechonol. 2005, 23, 47–55.
  24. Schulte, V.A.; Hahn, K.; Dhanasingh, A.; Heffels, K.-H.; Groll, J. Hydrogel–fibre composites with independent control over cell adhesion to gel and fibres as an integral approach towards a biomimetic artificial ECM. Biofabrication 2014, 6, 024106.
  25. Bonnans, C.; Chou, J.; Werb, Z. Remodelling the extracellular matrix in development and disease. Nat. Rev. Mol. Cell Biol. 2014, 15, 786–801.
  26. Kim, J.H.; Choi, Y.-J.; Yi, H.-G.; Wang, J.H.; Cho, D.-W.; Jeong, Y.H. A cell-laden hybrid fiber/hydrogel composite for ligament regeneration with improved cell delivery and infiltration. Biomed. Mater. 2017, 12, 055010.
  27. Jordan, A.M.; Kim, S.-E.; Van de Voorde, K.; Pokorski, J.K.; Korley, L.T.J. In Situ Fabrication of Fiber Reinforced Three-Dimensional Hydrogel Tissue Engineering Scaffolds. ACS Biomater. Sci. Eng. 2017, 3, 1869–1879.
  28. Tonsomboon, K.; Butcher, A.L.; Oyen, M.L. Strong and tough nanofibrous hydrogel composites based on biomimetic principles. Mater. Sci. Eng. C 2016, 72, 220–227.
  29. Franco, R.A.; Nguyen, T.H.; Lee, B.-T. Preparation and characterization of electrospun PCL/PLGA membranes and chitosan/gelatin hydrogels for skin bioengineering applications. J. Mater. Sci. Mater. Med. 2011, 22, 2207–2218.
  30. Sun, X.; Lang, Q.; Zhang, H.; Cheng, L.; Zhang, Y.; Pan, G. Electrospun Photocrosslinkable Hydrogel Fibrous Scaffolds for Rapid In Vivo Vascularized Skin Flap Regeneration. Adv. Funct. Mater. 2016, 27, 1604617.
  31. Klicova, M.; Klapstova, A.; Chvojka, J.; Koprivova, B.; Jencova, V.; Horakova, J. Novel double-layered planar scaffold combining electrospun PCL fibers and PVA hydrogels with high shape integrity and water stability. Mater. Lett. 2020, 263, 127281.
  32. Contardi, M.; Kossyvaki, D.; Picone, P.; Summa, M.; Guo, X.; Heredia-Guerrero, J.A.; Giacomazza, D.; Carzino, R.; Goldoni, L.; Scoponi, G.; et al. Electrospun Polyvinylpyrrolidone (PVP) hydrogels containing hydroxycinnamic acid derivatives as potential wound dressings. Chem. Eng. J. 2020, 409, 128144.
  33. Azarniya, A.; Tamjid, E.; Eslahi, N.; Simchi, A. Modification of bacterial cellulose/keratin nanofibrous mats by a tragacanth gum-conjugated hydrogel for wound healing. Int. J. Biol. Macromol. 2019, 134, 280–289.
  34. Loo, Y.; Wong, Y.-C.; Cai, E.Z.; Ang, C.-H.; Raju, A.; Lakshmanan, A.; Koh, A.G.; Zhou, H.J.; Lim, T.-C.; Moochhala, S.M.; et al. Ultrashort peptide nanofibrous hydrogels for the acceleration of healing of burn wounds. Biomaterials 2014, 35, 4805–4814.
  35. Nguyen, L.H.; Gao, M.; Lin, J.; Wu, W.; Wang, J.; Chew, S.Y. Three-dimensional aligned nanofibers-hydrogel scaffold for controlled non-viral drug/gene delivery to direct axon regeneration in spinal cord injury treatment. Sci. Rep. 2017, 7, 42212.
  36. Han, N.; Johnson, J.; Lannutti, J.J.; Winter, J.O. Hydrogel–electrospun fiber composite materials for hydrophilic protein release. J. Control. Release 2012, 158, 165–170.
  37. An, D.; Ji, Y.; Chiu, A.; Lu, Y.-C.; Song, W.; Zhai, L.; Qi, L.; Luo, D.; Ma, M. Developing robust, hydrogel-based, nano fiber-enabled encapsulation devices (NEEDs ) for cell therapies. Biomaterials 2015, 37, 40–48.
  38. Jain, K.K. An Overview of Drug Delivery Systems. In Methods in Molecular Biology; Jain, K.K., Ed.; Humana Press: New York, NY, USA, 2020; Volume 2059, pp. 1–54.
  39. Figueroa-Pizano, M.D.; Vélaz, I.; Peñas, F.J.; Zavala-Rivera, P.; Rosas-Durazo, A.J.; Maldonado-Arce, A.D.; Martínez-Barbosa, M.E. Effect of freeze-thawing conditions for preparation of chitosan-poly (vinyl alcohol) hydrogels and drug release studies. Carbohydr. Polym. 2018, 195, 476–485.
  40. Norouzi, M.; Shabani, I.; Ahvaz, H.H.; Soleimani, M. PLGA/gelatin hybrid nanofibrous scaffolds encapsulating EGF for skin regeneration. J. Biomed. Mater. Res. Part A 2015, 103, 2225–2235.
  41. Sun, Y.; Cheng, S.; Lu, W.; Wang, Y.; Zhang, P.; Yao, Q. Electrospun fibers and their application in drug controlled release, biological dressings, tissue repair, and enzyme immobilization. RSC Adv. 2019, 9, 25712–25729.
  42. Gao, Q.; Liu, L.; Lu, X.; Zhou, H. In situ forming hydrogels based on chitosan for drug delivery and tissue regeneration. Asian J. Pharm. Sci. 2016, 11, 673–683.
  43. Teixeira, M.A.; Amorim, M.T.P.; Felgueiras, H.P. Poly(Vinyl Alcohol)-Based Nanofibrous Electrospun Scaffolds for Tissue Engineering Applications. Polymers 2020, 12, 7.
  44. Sill, T.J.; von Recum, H.A. Electrospinning: Applications in drug delivery and tissue engineering. Biomaterials 2008, 29, 1989–2006.
  45. Torres-Martinez, E.J.; Bravo, J.M.C.; Medina, A.S.; González, G.L.P.; Gómez, L.J.V. A Summary of Electrospun Nanofibers as Drug Delivery System: Drugs Loaded and Biopolymers Used as Matrices. Curr. Drug Deliv. 2018, 15, 1360–1374.
  46. Chou, S.-F.; Carson, D.; Woodrow, K.A. Current strategies for sustaining drug release from electrospun nanofibers. J. Control. Release 2015, 220, 584–591.
  47. Bas, O.; De-Juan-Pardo, E.M.; Catelas, I.; Hutmacher, D.W. The quest for mechanically and biologically functional soft biomaterials via soft network composites. Adv. Drug Deliv. Rev. 2018, 132, 214–234.
  48. Lee, K.Y.; Rowley, J.A.; Eiselt, P.; Moy, E.M.; Bouhadir, K.H.; Mooney, D.J. Controlling mechanical and Swelling Properties of Alginate Hydrogels Independently by Cross-Linker Type and Cross-Linking Density. Macromolecules 2000, 33, 4291–4294.
  49. Hsieh, A.; Zahir, T.; Lapitsky, Y.; Amsden, B.; Wan, W.; Shoichet, M.S. Hydrogel/electrospun fiber composites influence neural stem/progenitor cell fate. Soft Matter 2010, 6, 2227–2237.
  50. Ekaputra, A.K.; Prestwich, G.D.; Cool, S.M.; Hutmacher, D.W. The three-dimensional vascularization of growth factor-releasing hybrid scaffold of poly (ε-caprolactone )/collagen fibers and hyaluronic acid hydrogel. Biomaterials 2011, 32, 8108–8117.
  51. Su, T.; Zhao, W.; Wu, L.; Dong, W.; Qi, X. Facile fabrication of functional hydrogels consisting of pullulan and polydopamine fibers for drug delivery. Int. J. Biol. Macromol. 2020, 163, 366–374.
  52. Ghalei, S.; Nourmohammadi, J.; Solouk, A.; Mirzadeh, H. Enhanced Cellular Response Elicited by Addition of Amniotic fluid to Alginate Hydrogel-Electrospun Silk Fibroin Fibers for Potential Wound Dressing Application. Colloids Surfaces B Biointerfaces 2018, 172, 82–89.
  53. Jaiswal, M.; Gupta, A.; Agrawal, A.K.; Jassal, M.; Dinda, A.K.; Koul, V. Bi-Layer Composite Dressing of Gelatin Nanofibrous Mat and Poly Vinyl Alcohol Hydrogel for Drug Delivery and Wound Healing Application: In-Vitro and In-Vivo Studies. J. Biomed. Nanotechnol. 2013, 9, 1495–1508.
  54. Chen, Y.; Qiu, Y.; Wang, Q.; Li, D.; Hussain, T.; Ke, H.; Wei, Q. Mussel-inspired sandwich-like nanofibers/hydrogel composite with super adhesive, sustained drug release and anti-infection capacity. Chem. Eng. J. 2020, 399, 125668.
More
This entry is offline, you can click here to edit this entry!
Video Production Service