nanotechnology in biomaterials: nanofibers in tissue...

32
195 10 Nanotechnology in Biomaterials: Nanofibers in Tissue Engineering Deniz Yucel, Halime Kenar, Albana Ndreu, Tugba Endogan, Nesrin Hasirci, and Vasif Hasirci Departments of Biological Sciences and Chemistry, Middle East Technical University, Ankara, Turkey 10.1 Biomaterials Devices made of synthetic or natural materials have been used in the medical area and introduced into the human body to improve human health since ancient times. Romans, Chinese, and Aztecs used gold wires in dentistry more than 2,000 years ago. Ancient Egyptians and Greeks sutured wounds with plant bers and animal-derived materials and used wood for prosthetic limbs. Since then, scientists have continued their research to nd bioactive agents to cure illnesses and to improve the quality of human life by using articial prostheses. There are various denitions of biomaterials expressed in different ways given in the literature, but more or less they have the same meaning. One of the very early deni- tions came from the Clemson University Advisory Board for Biomaterials (1976), which described a biomaterial as “a systemically and pharmacologically inert substance designed for implantation within or incorporation with living systems (Park, 1981).” Another def- inition of biomaterials is “materials of synthetic as well as of natural origin in contact with tissue, blood, and biological uids, and intended for use for prosthetic, diagnostic, therapeutic, and storage applications without adversely affecting the living organism and its components” (Bruck, 1980). Biomaterials can be dened as materials to be used to CONTENTS 10.1 Biomaterials ...................................................................................................................... 185 10.2 Tissue Engineering .......................................................................................................... 186 10.3 Importance of Nano/Microbrous Scaffolds............................................................... 189 10.4 Fabrication of Nano/Microbrous Scaffolds ............................................................... 189 10.5 Nano/Microbers in Tissue Engineering Applications ............................................. 191 10.5.1 Bone Tissue Engineering .................................................................................. 191 10.5.2 Nerve Tissue Engineering ................................................................................ 197 10.5.3 Cardiac Muscle Tissue Engineering ............................................................... 204 References..................................................................................................................................... 208 K10254_C010.indd 195 5/12/11 7:18:06 PM

Upload: others

Post on 11-Jul-2020

3 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Nanotechnology in Biomaterials: Nanofibers in Tissue ...biomed.metu.edu.tr/sites/biomat.metu.edu.tr/files/Yucel et al, 2011.pdf · membranes (wound dressings, drug delivery systems,

195

10Nanotechnology in Biomaterials: Nanofibers in Tissue Engineering

Deniz Yucel, Halime Kenar, Albana Ndreu, Tugba Endogan, Nesrin Hasirci, and Vasif HasirciDepartments of Biological Sciences and Chemistry, Middle East Technical University, Ankara, Turkey

10.1 Biomaterials

Devices made of synthetic or natural materials have been used in the medical area and introduced into the human body to improve human health since ancient times. Romans, Chinese, and Aztecs used gold wires in dentistry more than 2,000 years ago. Ancient Egyptians and Greeks sutured wounds with plant !bers and animal-derived materials and used wood for prosthetic limbs. Since then, scientists have continued their research to !nd bioactive agents to cure illnesses and to improve the quality of human life by using arti!cial prostheses.

There are various de!nitions of biomaterials expressed in different ways given in the literature, but more or less they have the same meaning. One of the very early de!ni-tions came from the Clemson University Advisory Board for Biomaterials (1976), which described a biomaterial as “a systemically and pharmacologically inert substance designed for implantation within or incorporation with living systems (Park, 1981).” Another def-inition of biomaterials is “materials of synthetic as well as of natural origin in contact with tissue, blood, and biological "uids, and intended for use for prosthetic, diagnostic, therapeutic, and storage applications without adversely affecting the living organism and its components” (Bruck, 1980). Biomaterials can be de!ned as materials to be used to

CONTENTS10.1 Biomaterials ...................................................................................................................... 18510.2 Tissue Engineering .......................................................................................................... 18610.3 Importance of Nano/Micro!brous Scaffolds............................................................... 18910.4 Fabrication of Nano/Micro!brous Scaffolds ............................................................... 18910.5 Nano/Micro!bers in Tissue Engineering Applications ............................................. 191

10.5.1 Bone Tissue Engineering .................................................................................. 19110.5.2 Nerve Tissue Engineering ................................................................................ 19710.5.3 Cardiac Muscle Tissue Engineering ............................................................... 204

References ..................................................................................................................................... 208

K10254_C010.indd 195 5/12/11 7:18:06 PM

Page 2: Nanotechnology in Biomaterials: Nanofibers in Tissue ...biomed.metu.edu.tr/sites/biomat.metu.edu.tr/files/Yucel et al, 2011.pdf · membranes (wound dressings, drug delivery systems,

196 Bionanotechnology II: Global Prospects

substitute a tissue or organ of a living system that is not fully functioning, or to support the biological system in its function while in intimate contact with the tissues in a safe, reli-able, economic, and physiologically acceptable manner (Hench and Erthridge, 1982). Black (1992) de!ned biomaterial as “a nonviable material used in a medical device, intended to interact with biological systems.” In addition, during the Second Consensus Conference on De!nitions in Biomaterials Science, a biomaterial was de!ned as “a material intended to interface with biological systems to evaluate, treat, augment, or replace any tissue, organ or function of the body” (Williams, 1992). The same de!nition is found in the Williams Dictionary of Biomaterials (Liverpool University Press, 1999).

Biomaterials are used in many application areas, such as orthopedic (total joint replace-ments, bone plates and screws, etc.), dental (tooth !llings, crowns, implants, etc.), cardio-vascular (cardiac pacemaker, arti!cial heart valve, blood vessels, etc.), ophthalmic (contact and intraocular lenses), wound healing (sutures, wound dressing, tissue adhesive, ligating clips, staples, etc.), and drug delivery (oral, inhalation injectable delivery systems, micro- and nanodevices, sheets, !bers, and sponges, etc.) systems. Biomaterials can be found in different forms, such as tubes (arti!cial veins, arti!cial trachea), !bers (sutures), !lms and membranes (wound dressings, drug delivery systems, arti!cial kidney), gels (burn covers, topical drug delivery systems), and porous scaffolds (tissue engineering), depending on the site and type of use. Metals (titanium, stainless steel, cobalt-chromium alloys, gold, silver, platinum), polymers (nylon, silicones, Te"on, dacron, polylactides), ceramics (aluminum oxide, carbon, hydroxyapatite), natural materials (collagen, hyaluronic acid, reconstituted tissue such as porcine heart valves), and composites (carbon or ceramic wires, !ber-rein-forced polymers) have been used as biomaterials in the design and production of biomedi-cal devices. Material to be used as a biomaterial must ful!ll some requirements. First, it should be compatible with the biological environment and should not cause any adverse carcinogenic or allergic tissue reaction; i.e., it should be biocompatible. Degradation prod-ucts of a biomaterial must also be nontoxic and noncarcinogenic, and have the desired bioactivity toward the cells and tissue. Physical, chemical, and mechanical properties of the biomaterial should match those of the target tissue.

10.2 Tissue Engineering

Tissue engineering was once considered a sub!eld of biomaterials, but since it has gained too much importance in the last decades, it is now considered a !eld in its own right. Tissue engineering is the process of creating living three-dimensional (3D) tissues and organs by using engineering and materials methods, and speci!c combinations of cells and cell signals, both chemical and mechanical (Grif!th, 2002). This approach aims to reproduce both the form and function of the tissue. Different from nonliving biomaterials, tissue engineering is based on the formation and regeneration of a new functional tissue by com-bining living components (cells, tissue fragments) and carrier scaffolds in a construct to be implanted into the body (Kneser et al., 2002; Guelcher et al., 2006).

There are three approaches in repairing tissues. The !rst one is the injection of tissue-speci!c viable cells directly into the damaged site of the tissue. The second approach is the delivery of tissue-inducing substances, such as growth and differentiation factors, to

K10254_C010.indd 196 5/12/11 7:18:06 PM

Page 3: Nanotechnology in Biomaterials: Nanofibers in Tissue ...biomed.metu.edu.tr/sites/biomat.metu.edu.tr/files/Yucel et al, 2011.pdf · membranes (wound dressings, drug delivery systems,

Nanotechnology in Biomaterials: Nanofibers in Tissue Engineering 197

target locations. The third one is the tissue engineering approach, in which cells are grown in 3D scaffolds. The use of cell suspensions or tissue-inducing substances is considered when the defects are small, well contained, or not suitable for implanting a 3D structure. To engineer tissues with practical size and predetermined shapes, the !rst two approaches are not appropriate. The main principle of the tissue engineering strategy is to seed a porous, biodegradable 3D cell carrier (scaffold, matrix) with cells, add signaling molecules like growth factors, culture the construct in a medium for growing and proliferation of cells, and then implant it into the defect area to grow a new tissue. It is expected that the cells of the natural tissue would also attach to the scaffold, proliferate, differentiate, and get organized, forming a normal, healthy tissue while the scaffold itself degrades. At the end, healthy tissue !lls the defect site and no arti!cial matrix remains. Scaffolds, cells, and signaling biomolecules are the main components of tissue engineering devices. All of these components play very important roles during tissue regeneration. The extracel-lular matrix (ECM) is a microenvironment composed of parenchymal cells (functional cells), mesenchymal cells (support cells), and structural materials. Every tissue and organ in our body is formed in these microenvironments. The body serves as a bioreactor, which applies biomechanical forces and provides biochemical signals to the cells and the ECM to develop and maintain tissue and organs (Barnes et al., 2007). In the tissue engineering approach, the biological way of tissue formation in the body is mimicked by taking place in a porous matrix.

A scaffold is one of the essential components of tissue engineering since it provides 3D support for the cells, as ECM does. In vitro, cells do not grow in a way to !ll a volume if there is no 3D support. Porous scaffolds provide some place for cell attachment and viability through transport of nutrients and metabolites, and allow cell expansion and tis-sue organization. There are some requirements for proper scaffolds. First, they should be biocompatible, and therefore should not cause any adverse responses, such as allergic and carcinogenic reactions in the body. They must have an interconnected, highly porous struc-ture with a high surface area for cell adhesion and reorganization, for transport of nutri-ents and removal of metabolic waste, and for homogeneous cell distribution throughout the scaffold. They should have adequate mechanical strength to withstand the pressures and mechanical forces applied by the biological system. The degradation rate of the scaf-fold should be in accordance with the tissue growth; when the tissue is totally regenerated, the scaffold should be almost fully degraded. A suitable surface chemistry is extremely important for cell attachment, proliferation, and differentiation Ma, 2004a; Stevens et al., 2008. The material of the scaffold should be easily processed to achieve the desired shapes and sizes. Finally, the scaffold should have some speci!c properties for speci!c tissue engi-neering applications, such as osteoconductivity for bone, electrical conductivity for nerve, and contraction for myocardial tissue engineering.

Various materials have been studied as potential scaffolds. Some metals are good choices for load-bearing implants since they have superior mechanical properties (Catledge et al., 2004). Recently, porous titanium (Ti) and titanium alloys were used as scaffold biomaterials for bone tissue engineering, but they are not degradable in the body. Inorganic materials are also used in bone tissue engineering studies (Hench and Polak, 2002). Ceramics such as hydroxyapatite (HAp), Bioglass, A-W glass ceramics, and

-tricalcium phosphate ( -TCP) closely mimic bone tissues. They are osteoconductive or osteoinductive, and enhance the absorption of bone ECM proteins, resulting in improved osteoblast adhesion (Ducheyne and Qiu, 1999; Dieudonne et al., 2002; Smith et al., 2004).

K10254_C010.indd 197 5/12/11 7:18:06 PM

Page 4: Nanotechnology in Biomaterials: Nanofibers in Tissue ...biomed.metu.edu.tr/sites/biomat.metu.edu.tr/files/Yucel et al, 2011.pdf · membranes (wound dressings, drug delivery systems,

198 Bionanotechnology II: Global Prospects

However, ceramics have some drawbacks, like fragility, brittleness, low tensile strength, and being mostly not biodegradable. Biological and synthetic polymeric materials have been widely used in various tissue engineering applications due to their degradability, biocompatibility, mechanical properties suitable for many applications, and ease of pro-cessability. Some examples of natural biodegradable polymers are collagen, !brinogen, gelatin, chitosan, cellulose, alginate, starch, silk, and hyaluronic acid. The main advan-tages of these materials are their low immunogenicity, bioactivity, capability of inter-acting with the host tissue, chemical versatility, and availability from various sources, as in starch and chitosan. Of these, collagen, !brin, hyaluronic acid, agarose, chitosan, and alginate are materials especially used in the production of scaffolds for engineering of bone and cartilage (Sabir et al., 2009). The most widely used synthetic polymers are a family of linear aliphatic polyesters, poly( -hydroxy acid) (PHA), such as poly(lactic acid) (PLA), poly(glycolic acid) (PGA), and their copolymer poly(lactic acid-co-glycolic acid) (PLGA) (Mooney et al., 1996; Seal et al., 2001). For example, PGA has been used in the engineering of cartilage, tendon, ureter, intestine, blood vessel, heart valve, and other tissues. Poly( -caprolactone) (PCL), poly(propylene fumarate) (PPF), poly(carbonate), poly(phosphazenes), poly(phosphoesters) and poly(anhydrides), tyrosine-derived poly-mers, and biodegradable urethane-based polymers are other important synthetic, bio-degradable polymers used in tissue engineering applications. A problem with synthetic polymers is their degradation products, which may reduce the local pH, induce an in"ammatory response, or create some adverse reactions and being nonbiocompatible.

In the past few years, polymer-ceramic composites have gained increased importance in the engineering of several types of tissues, such as bone, cartilage, tendon, and ligament (Marra et al., 1999; Zhang and Ma, 1999; Ma et al., 2001; Zhao et al., 2002; Liu and Ma, 2004). The composite is expected to have improved compressive mechanical properties compared to the polymer, and better structural integrity and "exibility than ceramics, and thus the combination of ceramic and polymer created reinforced porous scaffolds with enhanced bioactivity and controlled resorption rates (Zhang and Ma, 1999; Ma et al., 2001).

During the selection of a scaffold material, an important step is to decide on the fab-rication technique. The processing technique is expected not to change certain material properties, like biocompatibility and surface chemistry. The technique should be repro-ducible in terms of scaffold porosity, pore size, pore distribution, and interconnectivity (Leong et al., 2003). There are many techniques that can be used to process materials into scaffolds, such as solvent casting, phase inversion, !ber bonding, melt-based technologies, high-pressure-based methods, freeze drying, and rapid prototyping (Ma and Langer, 1999; Kinikoglu et al., 2009; Sangsanoh et al., 2009; Yilgor et al., 2008; Vrana et al., 2007; Zorlutuna et al., 2007; Mooney 1996; Thomson et al., 1995; Mikos et al., 1993). Each production tech-nique has some disadvantages and advantages over the others and may not be applicable to some polymers. Therefore, appropriate processing techniques should be chosen to pro-duce a scaffold with the desired properties for the speci!c application.

To date, scaffolds have been produced in our group in various forms, like foam (Zorlutuna et al., 2008; Kinikoglu et al., 2009, Dogan et al., 2009, Ulubayram et al., 2001), !lm (Vrana et al., 2007; Zorlutuna et al., 2007; Kenar et al., 2008; Zorlutuna et al., 2009), and !bers (Ndreu et al., 2008; Kenar et al., 2009; Yucel et al., 2010), each having different chemistries, and a number of studies have been carried out with them either in vitro or in vivo. In the last decades, foam and !lm-based scaffolds were popular for 3D and 2D applications; however, in recent years there has been an increase in the use of !brous scaffolds, especially made by electrospinning.

K10254_C010.indd 198 5/12/11 7:18:06 PM

Page 5: Nanotechnology in Biomaterials: Nanofibers in Tissue ...biomed.metu.edu.tr/sites/biomat.metu.edu.tr/files/Yucel et al, 2011.pdf · membranes (wound dressings, drug delivery systems,

Nanotechnology in Biomaterials: Nanofibers in Tissue Engineering 199

10.3 Importance of Nano/Microfibrous Scaffolds

Many extracellular proteins have a !brous structure with diameters on the low or submi-crometer scales. For example, collagen is the most abundant ECM protein in the human body, and it has a !brous structure with !ber bundle diameters varying from 50 to 500 nm, depending on the site and species (Hay, 1991). In the native tissues, the structural ECM proteins are one to two orders of magnitude smaller than the cells, and therefore, the cells are in direct contact with many ECM !bers. It is now well known that many biologi-cally active molecules, extracellular matrix components, and cells interact with each other using functional groups on the nanoscale. It is therefore a useful approach to mimic the !brous nature of the extracellular matrix when fabricating scaffolds (Barnes et al., 2007). Nano!bers have a high surface-to-volume ratio, which enhances the number of cells that adhere to the surface (Ma and Langer, 1999). Cell migration, proliferation, and differentia-tion follow adhesion (Palecek et al., 1997), and it is expected that these reactions will be much higher on nano!brous scaffolds than on foams or !lms. As a result, nano!bers have attracted the attention of many researchers who considered them as potential scaffolds for applications such as tissue engineered vasculature (Xu et al., 2004; Lee et al., 2007), bone (Sui et al., 2007; Cui et al., 2007), nerve (Yang et al., 2005; Schnell et al., 2007), and tendon and ligament (Lee et al., 2005; Sahoo et al., 2006).

10.4 Fabrication of Nano/Microfibrous Scaffolds

Since !brous scaffolds with nanometric !ber diameters have been found to be satisfac-tory for various tissue engineering applications, extensive research toward developing processes for the fabrications of these !brous structures is being conducted. The main methods used in the fabrication of !brous scaffolds are self-assembly, template synthe-sis, drawing and extrusion, phase separation, wet spinning, and electrospinning. Each of these procedures leads to !bers with different dimensions and organizations.

In the case of self-assembly, atoms and molecules arrange themselves through weak and noncovalent interactions, and it is known as a bottom-up method since the built-up nanoscale !bers are formed starting from smaller molecules. It yields !bers with small dimensions (less than 100 nm wide and up to a few micrometers long), and it offers novel properties and functionalities that cannot be achieved by conventional organic synthesis (Hasseinkhan et al., 2006). The main disadvantage of this method is its complexity, in addition to being a long and extremely elaborate process with low productivity (Ma et al., 2005).

Another fabrication method is template synthesis. As the name implies, a nanoporous membrane is used as a mold or template in order to obtain the material of interest in the form of solid nano!brous or hollow-shaped tubules. This method uses a polymer solution to be extruded through a porous membrane by application of pressure. Fibers are formed when the extruded polymer comes into contact with a precipitating solution (Feng et al., 2002). Fiber diameter depends on the pore size of the template used, and it varies from a few to a hundred nanometers. Many materials, like metals, polymers, and carbon, can be utilized as templates for !ber fabrication; however, this method is limited in that the length of nano!bers can only be a few micrometers.

K10254_C010.indd 199 5/12/11 7:18:06 PM

Page 6: Nanotechnology in Biomaterials: Nanofibers in Tissue ...biomed.metu.edu.tr/sites/biomat.metu.edu.tr/files/Yucel et al, 2011.pdf · membranes (wound dressings, drug delivery systems,

200 Bionanotechnology II: Global Prospects

Drawing is a method to produce single nano!bers. It requires a minimum amount of equipment and is a discontinuous process. A micropipette is dipped into a droplet near the solution-solid surface contact line via a micromanipulator, and then the micropipette is withdrawn from the liquid at a certain speed, yielding a nano!ber. These steps are repeated many times, each time with a different droplet. The solution viscosity, however, increases with solvent evaporation, and some !ber breaking occurs due to instabilities that occur during the process (Ondarcuhu and Joachim, 1998). The main limitation of this method is that it can only use solutions of viscoelastic materials, which can undergo strong deformations that are cohesive enough to withstand the stresses developed during the pull. Again, the !ber diameter is dependent on the ori!ce size used. It is dif!cult to obtain !ber diameters of less than 100 nm.

Phase separation is another process utilized in nano!ber production. The main principle of this method is that a polymer dissolved in an appropriate solvent is stirred at a certain gelation temperature for a period of time until a homogeneous solution is obtained. Then, the formed gel is immersed in water several times for solvent exchange to occur. The !nal gel is lyophilized. Phase separation occurs due to chemical incompatibility and results in nano!bers, but the problem is that a long time is needed to convert the polymer into a nanoporous foam. The !ber diameter ranges from 50 to 500 nm with a length of a few micrometers (Zhang et al., 2005), which is a disadvantage for some applications. Moreover, only polymers that have gelation capability can be used to obtain nano!brous structures.

Wet spinning is a process similar to drawing, with the difference being that a polymeric solution is precipitated or coagulated by dilution, adding into a nonsolvent or a chemical reaction. Fibers produced by this method are continuous and in microscale (from 10 to 100 µm). Polymer solution concentration and the spinneret diameter are the two crucial parameters that in"uence the resulting !ber diameter.

The last, and one of the most preferred, methods used to prepare !bers in nano- and low microscale is the electrospinning process. High electrostatic forces are applied to draw continuous !bers from a polymeric solution through a syringe. Basically, four components are required to accomplish the process: a capillary tube ending in a needle of small diam-eter, a high-voltage supplier, a syringe pump, and a metallic component. One of the elec-trodes is connected to the needle of the syringe containing the polymer, and the other one is connected to the collector. Leakage of the polymer solution is prevented by surface tension; however, as the applied potential increases, the surface tension contribution starts to decrease as a result of charge repulsion, and the "uid shape at the needle tip becomes like a cone, what is called the Taylor cone. The voltage is increased until the repulsive elec-trostatic forces overcome the in"uence of solution surface tension. Meanwhile, the solvent evaporates and the charged jet of the solution is ejected from the needle tip in the shape of a long, continuous, thin !ber. Therefore, !bers with various diameters are collected on either a stationary or rotating collector. Rotating collectors are used to produce aligned nano!bers, which are more preferable in certain applications. A variety of polymers have been used until now to produce !bers from tens of nanometers to a few micrometers (Jin et al., 2004; Li et al., 2005; Ma et al., 2005; Ndreu et al., 2008; Erisken et al., 2008; Jeong et al., 2009; Kenar et al., 2009; Yucel et al., 2010).

Certain properties are expected from a polymer that is proven to be spinnable. The !bers should be consistent, defect-free, and have controllable diameters. The main parameters to control are process conditions such as polymer type, its concentration, solvents, applied potential, distance between the needle tip and the collector, and the collector type. These parameters are modi!ed to control !ber thickness, porosity and pore size (or rather the spacing between the !bers or !ber density), surface chemistry, and surface topography

K10254_C010.indd 200 5/12/11 7:18:06 PM

Page 7: Nanotechnology in Biomaterials: Nanofibers in Tissue ...biomed.metu.edu.tr/sites/biomat.metu.edu.tr/files/Yucel et al, 2011.pdf · membranes (wound dressings, drug delivery systems,

Nanotechnology in Biomaterials: Nanofibers in Tissue Engineering 201

(Deitzel et al., 2001; Huang et al., 2003; Katti et al., 2004; Barhate et al., 2006; Ndreu et al., 2008; Wang J et al., 2009). Therefore, optimization of the process conditions in order to get the best results is challenging with so many parameters.

Finally, it can be said that compared to the other nano/micro!ber-production methods, electrospinning is more economical, simpler, yields continuous !bers, and is versatile enough to be used for the production of nano!bers from a variety of materials. Therefore, electrospinning represents an attractive technique to be used for producing nano!ber-based scaffolds for tissue engineering.

10.5 Nano/Microfibers in Tissue Engineering Applications

The use of polymeric nano/micro!bers in tissue engineering applications is rapidly grow-ing. The main areas include the engineering of skin (Venugopal and Ramakrishna, 2005; Noh et al., 2006; Yang et al., 2009; Jeong et al., 2009; Torres et al., 2009), ligament and tendon (Petrigliano et al., 2006; Teh et al., 2007, 2008; Sahoo et al., 2009; Fan et al., 2009; Yin et al., 2009), skeletal muscle (Riboldi et al., 2005; Choi et al., 2008), cartilage (Li WJ et al., 2005; Li et al., 2006; Janjanin et al., 2008; Lu et al., 2008; Hu et al., 2009), bone (Santos et al., 2008; Zhang et al., 2008; Reed et al., 2009; Wang J et al., 2011; Prabhakaran et al., 2009), cardio-vascular (Kenar et al., 2009; Rockwood et al., 2008; Li WJ et al., 2006), and nerve (Yucel et al., 2010; Neal et al., 2009; Nisbet et al., 2008; Corey et al., 2007) tissues. The increase in the number of review articles (Ashammakhi et al., 2007a, 2007b, 2008; Agarwal et al., 2009; Jang et al., 2009) and publications related to the application of nano/micro!bers in these !elds shows the interest in electrospinning for tissue engineering. In this review we will concentrate more on the studies carried out by our research group, and also by some of the leading researchers in the !elds of bone, cardiovascular, and nerve tissue engineering.

10.5.1 Bone Tissue Engineering

Prior to designing a bone substitute or a tissue-engineered bone, it is important to know well the structure and function of the natural bone so that the material type chosen is the most suitable to regenerate the function of the damaged bone. Bone is a complex, physically hard, rigid, and highly organized tissue. This tissue is composed of an intracellular matrix in a !brous form made mainly of collagen types I and V, and some inorganic materials. Apart from the major collagen matrix, some other noncollageneous proteins, like osteo-calcin and osteopontin, are also present. There are three cell types present in the bone structure: osteoblasts, osteocytes, and osteoclasts. Osteoblasts, derived from mesenchymal precursor cells in the marrow, are the bone cells responsible for formation and structural organization of the bone matrix, and also bone mineralization. When osteoblasts become trapped in the matrix, they secrete and become osteocytes. Osteocytes are the most abun-dant cells found in compact bone. They are networked to each other via long cytoplasmic extensions that occupy tiny canals called canaliculi, which are used for exchange of nutri-ents and waste. The space that an osteocyte occupies is called a lacuna. Unlike osteoblasts, these cells have reduced synthetic activity and are not capable of mitotic division. They are actively involved in the turnover of bony matrix, through various mechanosensory mech-anisms. They destroy bone through a mechanism called osteocytic osteolysis. The third bone cell type, osteoclasts, are polarized cells and have a crucial role in bone resorption,

K10254_C010.indd 201 5/12/11 7:18:06 PM

Page 8: Nanotechnology in Biomaterials: Nanofibers in Tissue ...biomed.metu.edu.tr/sites/biomat.metu.edu.tr/files/Yucel et al, 2011.pdf · membranes (wound dressings, drug delivery systems,

202 Bionanotechnology II: Global Prospects

which includes an initial mineral dissolution and then an organic phase degradation (Jang et al., 2009).

As mentioned above, the main approach in the tissue engineering process is mimicking the ECM. Therefore, for bone tissue engineering it is crucial to make a design whose prop-erties would mimic best the structure and function of bone ECM. It is important to choose a material that interacts well with bone cells in the bone formation processes. Moreover, it is favorable that materials with good mechanical properties and can withstand various external stresses applied on bone are incorporated into the material design. Since the orga-nization of all bone compartments is in nanoscale or at the molecular level, a nanoscale-based design would be optimal to mimic the natural bone. Various strategies have been tried in designing a tissue engineered bone.

Both synthetic and natural materials have been utilized in this !eld. Among all poly-meric materials, a group of polyesters, such as PLA, PGA, PCL, poly(3-hydroxybutyric acid) (PHB), and their copolymers, has been the most extensively studied polymers to produce the nano!brous system for the regeneration of bone tissues (Burg et al., 2000). A summary of nano/micro!brous scaffolds used in bone tissue engineering studies is given in Table 10.1. Yoshimoto et al. (2003) used electrospun PCL nano!bers, and microscopic analysis of mes-enchymal stem cell-seeded scaffolds revealed a good cell penetration in addition to a high level of mineralization and collagen type I production after 4 weeks of culture (Yoshimoto et al., 2003). Sombatmankhong et al. produced mats from PHB, poly(3-hydroxybutyrate-co-3-hydroxyvalerate) (PHBV) and their 50/50 w/w blends and studied the biocompatibility of these mats by a human osteosarcoma cell line (Saos-2) and mouse !broblasts (L929). Both cells adhered well on all types of !brous scaffolds; however, the highest alkaline phos-phatase (ALP) activity was seen on the PHB/PHBV blend (Sombatmankhong et al., 2007).

In another study, PLA and poly(ethylene glycol) (PEG)-PLA diblock polymers with con-trolled topographies and chemistries were prepared by electrospinning in order to study the in"uence of surface topography and !ber diameter on cell behavior. In the presence of osteogenic factors the cell numbers on !brous scaffolds were reported to be equal to or higher than those on smooth surfaces. Furthermore, the authors indicated an increase in cell density with an increase in !ber diameter and no in"uence of surface substratum on ALP activity (Badami et al., 2006). The in"uence of !ber diameter on cell behavior was also studied by other researchers. Ndreu et al. (2008) studied the in"uence of different types of nano/micro!bers (diameter range: 300 nm–1.5 µm) on Saos-2 cells. Good cell adherence, proliferation, and in!ltration were observed on all types of scaffolds (PHBV, PHBV-PLGA, PHBV-P(L,DL)LA, and PHBV-PLLA); however, the highest number of cell attachments was observed on the !bers with the lowest diameter (PHBV-PLLA !ber diameter: ~350 nm) when the scaffolds were examined with a scanning electron microscope (SEM) and a con-focal laser scanning microscope (CLSM) (Figure 10.1).

Reed and his co-workers (2009) carried out a study on the reconstruction of a compound tissue based on the fact that healing does not occur as a single tissue, but as a compound one, such as bone-periosteum-skin. In order to perform this study, they seeded human foreskin !broblasts, murine keratinocytes, and periosteal cells on PCL nano!bers and showed that these scaffolds supported the growth of all these cell types. Even though keratinocytes grew randomly throughout the scaffolds, they reported improved longevity of the cocultured cells and a strong degree of osteoinduction. As a consequence, construc-tion of a composite tissue has been reported by the authors (Reed et al., 2009).

Compared to the synthetic materials, natural materials have better biocompatibility, and more importantly, they possess the main cues that are necessary for a cell to attach and grow. Natural polymers are also advantageous in that they can be degraded by naturally

K10254_C010.indd 202 5/12/11 7:18:06 PM

Page 9: Nanotechnology in Biomaterials: Nanofibers in Tissue ...biomed.metu.edu.tr/sites/biomat.metu.edu.tr/files/Yucel et al, 2011.pdf · membranes (wound dressings, drug delivery systems,

Nanotechnology in Biomaterials: Nanofibers in Tissue Engineering 203

TAB

LE 1

0.1

Fibr

ous

Scaf

fold

s Fa

bric

ated

by

Elec

tros

pinn

ing

for

Bone

Tis

sue

Engi

neer

ing

Poly

mer

Typ

ePo

lym

er C

once

ntra

tion

/Com

posi

tion

/Sol

vent

Fibe

r D

iam

eter

In V

itro

Stu

dies

: C

ell T

ype

Ref

eren

ce

PLA

/BG

nan

o!lle

r3.

5%, w

/v; 9

0:10

, w:w

CH

LH

undr

eds

of n

anom

eter

sM

C3T

3-E1

Noh

et a

l., 2

010

Gel

atin

/silo

xane

15–2

5% w

/wG

elat

in:G

PMS:

CaN

O3 fo

rmic

aci

dN

anom

eter

sca

leBM

SCs

Ren

et a

l., 2

010

PVA

/CO

L/n-

HA

p7%

w/w

PVA

:CO

L (5

5:45

)n-

HA

p: 5

–10%

w/w

of P

VA d

eion

ized

wat

er, H

Ac

~320

nm

—A

sran

et a

l., 2

010

PAN

/-T

CP

10%

w/w

DM

F~3

00 n

mM

G-6

3Li

u et

al.,

201

0

CH

I7%

w/w

TFA

/DC

M 7

0:30

v/v

126

20

nmM

C3T

3-E1

Sang

sano

h et

al.,

200

9

PCL/

nano

-apa

tite

(nA

p)80

:20%

w/w

80%

TFE

in H

2O32

0–43

0 nm

RBM

Yang

F. e

t al.,

200

9

CO

L-H

Ap

80:2

0% w

/wSo

l con

cent

ratio

n (0

.023

–0.0

40 g

/ml)

HFP

75–1

60 n

mM

C3T

3-E1

Song

et a

l., 2

008

CH

I/PV

A a

nd C

ECS/

PVA

CH

I (7%

w/w

): PV

A (1

0% w

/w) (

80:2

0 v/

v)A

q. H

Ac

100–

700

nmL9

29Ya

ng e

t al.,

200

8

CH

I-H

Ap

HA

p/C

HI (

30:7

0)

10%

w/w

UH

MW

PEO

HA

c:D

MSO

(10:

1, w

/w)

214

25

nmhF

OB

Zha

ng e

t al.,

200

8

PCL-

-TC

P12

% w

/wD

CM

0–15

% w

/w

-TC

P

200–

2,00

0 nm

MC

3T3-

E1Er

iske

n et

al.,

200

8

PLG

A/C

OL

8 g/

ml (

50:5

0 (w

:w))

HFP

382

125

nm

BMH

SCs

Ma

et a

l., 2

008

PLA

/HA

p6%

w/w

DC

M a

nd 1

,4-d

ioxa

ne31

3 nm

MG

63Su

i et a

l., 2

007 (con

tinue

d )

K10254_C010.indd 203 5/12/11 7:18:07 PM

Page 10: Nanotechnology in Biomaterials: Nanofibers in Tissue ...biomed.metu.edu.tr/sites/biomat.metu.edu.tr/files/Yucel et al, 2011.pdf · membranes (wound dressings, drug delivery systems,

204 Bionanotechnology II: Global ProspectsTA

BLE

10.

1 (C

ON

TIN

UED

)

Fibr

ous

Scaf

fold

s Fa

bric

ated

by

Elec

tros

pinn

ing

for

Bone

Tis

sue

Engi

neer

ing

Poly

mer

Typ

ePo

lym

er C

once

ntra

tion

/Com

posi

tion

/Sol

vent

Fibe

r D

iam

eter

In V

itro

Stu

dies

: C

ell T

ype

Ref

eren

ce

PHB,

PH

BV, P

HB

- PH

BV

(50:

50 w

/w)

14%

w/v

CH

L2,

300–

4,00

0 nm

SaO

S-2

and

L929

Som

batm

ankh

ong

et a

l., 2

007

Silk

!br

oin

(Nan

g-La

i and

D

OA

E-7

type

)10

–40%

(w/v

)85

% F

A21

7–61

0 nm

(Nan

g-La

i)18

3–81

0 nm

(DO

AE-

7)M

C3T

3-E1

Mee

chai

sue

et a

l., 2

007

PLA

(L- a

nd D

L-ty

pe)/

PEG

8.2%

w/w

(L-t

ype)

, 5%

w/w

(DL-

type

), 26

% w

/w (P

EG-P

LLA

), 26

% w

/w (P

EG-P

DLL

A)

HFP

246

79

nm (L

-typ

e)14

1 3

9 nm

(DL-

type

)17

1 6

6 nm

(PEG

-PLL

A)

889

446

nm

(PEG

-PD

LLA

)

MC

3T3-

E1Ba

dam

i et a

l., 2

006

Silk

/PEO

/nH

Ap/

BMP-

27.

86%

(w/v

),si

lk:P

EO (8

2:18

w/w

)5

g nH

AP/

100

g si

lk3

mg

BMP-

2/m

g si

lk !

broi

n

520

55

nmhM

SCs

Li e

t al.,

200

6

Gel

atin

/HA

p80

:20

(% w

/w)

HFP

200–

400

nmM

G63

Kim

et a

l., 2

005

PCL-

CaC

O3

PCL:

CaC

O3

75:

25 a

nd 2

5:75

% w

/wC

HL:

MeO

H (7

5:25

% w

/w)

760–

900

nmhF

OB

Fujih

ara

et a

l., 2

005

Silk

!br

oin/

PEO

7.5%

w/w

(80/

20 w

/w) i

n H

2O70

0 5

0 nm

Bone

mar

row

str

omal

ce

lls (B

MSC

s)Jin

et a

l., 2

004

PCL,

gel

atin

, PC

L/ge

latin

Gel

atin

: 2.5

–12.

5% w

/vPC

L: 1

0% w

/v50

:50

blen

d: 1

0% w

/vTF

E

Tens

of n

m–1

#m

BMSC

Zha

ng e

t al.,

200

5

PCL

10%

w/w

CH

L40

0 2

00 n

mBM

SCYo

shim

oto

et a

l., 2

003

Abb

revi

atio

ns: B

G, b

ioac

tive

glas

s; B

MSC

s, b

one

mar

row

-der

ived

mes

ench

ymal

ste

m c

ells

; CEC

S, N

-car

boxy

ethy

l chi

tosa

n; C

HI,

chito

san;

CH

L, c

hlor

ofor

m; C

OL,

col

-la

gen;

DC

M, d

ichl

orom

etha

ne; D

MF,

N,N

-dim

ethy

l fo

rmam

ide;

DM

SO, d

imet

hyl

sulfo

xide

; FA

, for

mic

aci

d; G

PMS,

3-(

glyc

idox

ypro

pyl)

trim

etho

xysi

lane

; H

Ac,

ace

tic a

cid;

HA

p, h

ydro

xyap

atite

; hFO

B, h

uman

feta

l ost

eobl

ast;

HFP

, 1,1

,1,3

,3,3

-hex

a"uo

ro-2

-pro

pano

l; hM

SC, h

uman

bon

e m

arro

w-d

eriv

ed m

esen

chy-

mal

ste

m c

ell;

L929

, mou

se !

brob

last

s; M

eOH

, met

hano

l; M

C3T

3-E1

, mou

se c

alva

ria-

deri

ved

mur

ine

preo

steo

blas

t cel

ls; M

G63

, hum

an o

steo

blas

ts c

ells

; PA

N,

poly

(acr

ylon

itrile

); PC

L, p

oly(

-cap

rola

cton

e); P

EG, p

oly(

ethy

lene

gly

col);

PEO

, pol

y(et

hyle

ne o

xide

); PH

B, p

oly(

hydr

oxyb

utyr

ic a

cid)

; PH

BV, p

oly(

3-hy

drox

y-bu

tyri

c-co

-3-h

ydro

xyva

leri

c ac

id);

PLA

, pol

y(la

ctic

aci

d); P

LGA

, pol

y(la

ctic

aci

d-co

-gly

colic

aci

d); R

BM, r

at b

one

mar

row

cel

ls; T

FA, t

ri"u

oroa

cetic

aci

d; T

FE,

2,2,

2-tr

i"uo

roet

hano

l; -T

CP,

-t

rica

lciu

m p

hosp

hate

.

K10254_C010.indd 204 5/12/11 7:18:07 PM

Page 11: Nanotechnology in Biomaterials: Nanofibers in Tissue ...biomed.metu.edu.tr/sites/biomat.metu.edu.tr/files/Yucel et al, 2011.pdf · membranes (wound dressings, drug delivery systems,

Nanotechnology in Biomaterials: Nanofibers in Tissue Engineering 205

occurring enzymes, and therefore, they can be used as biodegradable scaffolds. The most commonly used natural polymers are proteins (collagen, gelatin, !brinogen, silk, casein, etc.) and polysaccharides (chitosan, cellulose, hyaluronic acid, etc.). Sangsanoh et al. (2009) studied biological evaluation of electrospun chitosan nano!bers and !lms in vitro with four different cell types: Schwann cells, osteoblast-like cells, keratinocytes, and !broblasts. Both substrate forms were seen to support cell attachment and especially promoted pro-liferation of keratinocytes. While Schwann cells attached well on nano!brous structures, they did not adhere on !lms. On the other hand, even though osteoblast-like cells and !bro-blasts showed good attachment on both nano!brous and !lm surfaces they did not prolifer-ate well (Sangsanoh et al., 2009). Jin et al. (2004) produced silk !broin/poly(ethylene oxide) (PEO) nano!brous matrices (700–750 nm), examined the behavior of human bone marrow stromal cells (BMSCs), and reported that the matrices supported BMSC attachment and proliferation during 14 days of incubation with extensive matrix coverage (Jin et al., 2004).

A blend of natural and synthetic polymers is advantageous in that they combine the better mechanical properties of synthetic polymers with the biofunctionality of natural ones. In this approach the properties of the scaffold can be changed easily and can be arranged as desired by changing the composition and the ratio of the blend. Ngiam et al. (2009) prepared nano-hydroxyapatite (n-HAp) containing electrospun PLGA and PLGA/collagen (COL) blend nano!bers. Biomineralization of n-HAp on electrospun nano!bers was achieved by using an alternate calcium and phosphate solution dipping method. n-HAp crystallite deposition was predominant on PLGA/COL nano!bers compared to on PLGA, which shows that collagen is a good template for n-HAp nucleation. It was shown that the amount of n-HAp present had a greater in"uence on early cell behavior, such as attachment and proliferation, than in the intermediate stages because their ALP and total protein expression results for mineralized PLGA and PLGA/COL were comparable dur-ing the culture period (Ngiam et al., 2009).

An important requirement in bone tissue engineering is that the scaffold should be highly porous so that cells can be homogeneously distributed throughout the structure. Poor cell in!ltration into the structure is reported when electrospun scaffolds are used; the cells are observed more on the surface than the inside of the scaffold. Therefore, it is of great importance to !nd an approach to produce nano!brous scaffolds with a higher degree of cell penetration. Ekaputra et al. (2008) used three strategies to solve this problem: selective leaching of a water-soluble !ber phase (PEO or gelatin), the use of micron-sized

A B C

D

FIGURE 10.1SEM micrographs of (A) unseeded and (B) Saos-2 cell-seeded PHBV-P(L,DL-LA) (70:30) scaffolds 14 days after incubation. (C) CLSM images of Saos-2-seeded PHBV10 scaffolds 7 days after incubation (top view showing cell-matrix interaction). (D) CLSM of cell in!ltration within the scaffold as viewed in cross section (Z-axis direction). (From Ndreu et al., Nanomed, 3, 45–60, 2008.)

K10254_C010.indd 205 5/12/11 7:18:07 PM

Page 12: Nanotechnology in Biomaterials: Nanofibers in Tissue ...biomed.metu.edu.tr/sites/biomat.metu.edu.tr/files/Yucel et al, 2011.pdf · membranes (wound dressings, drug delivery systems,

206 Bionanotechnology II: Global Prospects

!bers as the scaffold, and a combination of micron-sized !bers with codeposition of a hyaluronic acid (HA)-derived hydrogel, Heprasil. In the !rst procedure, PCL/COL solu-tion was electrospun in the presence of PEO, which was then removed selectively in order to increase the scaffold porosity. In the second approach, PCL/COL was electrospun in micrometer-sized PCL/COL (µmPCL/COL). The third method involved the cospraying of µmPCL/COL matrix with Heprasil. All the scaffold types supported attachment and pro-liferation of human fetal osteoblasts (hFOB). While selective leaching improved cellular in!ltration to a low degree compared to meshes obtained by conventional electrospinning, µmPCL/COL micro!bers allowed better penetration. This effect was more pronounced when Heprasil regions were present in the structure.

A fabrication method that can be used to solve the low penetration problem is the use of electrospun nano/micro!bers in combination with 3D macro!ber deposition. In this design, macro!bers are expected to play a role in the structural integrity and mechani-cal properties of the scaffolds, whereas the nano/micro!bers can act as a cell entrapment system and a cell adhesion promoter. The in"uence of this nano/micro!brous network on the behavior of endothelial cells (EC) seeded onto a scaffold based on a starch and PCL blend was studied by Santos et al. (2008). They have shown that the nano!bers in"uenced cell morphology and the maintenance of the physiological expression pattern. They also showed these EC-seeded scaffolds to be sensitive to pro-in"ammatory stimulus; that is, the cells could migrate and form capillary-like structures within a 3D gel of collagen in response to some angiogenesis-stimulating factors (Santos et al., 2008).

A procedure that has been recently reported is on-site layer-by-layer (LBL) tissue regen-eration, which involves electrospinning one layer of the polymeric solution, then seeding the cells onto this layer and repeating these steps to create a 3D scaffold with the cells already inside. This method is advantageous in that a highly speci!c 3D environment can be created by varying the composition of the !brous layers, !ber thickness, diameter, and orientation as desired, and the neotissue formation can be unambiguously reduced (Agarwal et al., 2009).

Another way to improve cell carrier properties is surface modi!cation of the !bers. A higher degree of spreading and growth of Saos-2 on PHBV and some of its blends has been shown by our research group by treating with oxygen plasma in order to increase hydro-philicity by adding some functional groups on the scaffold surface (Ndreu et al., 2008).

An appropriate biomechanical property is another challenge in bone tissue engineer-ing, since the engineered tissue must be able to retain its shape and withstand the forces applied after implantation. It is well known that a signi!cant part of the bone is constituted of calcium phosphate mineral phase. The presence of bone-inducing inorganic components within bone substitutes enhances calcium phosphate deposition after the osteogenic dif-ferentiation process. The apatite minerals and proteins/peptides, as well as drugs encap-sulated within the nano!bers, are a promising strategy used for nano!brous materials to achieve adequate mechanical properties and bone-speci!c bioactivity (Jang et al., 2009).

HAp plays a crucial role in the biomechanics of bone tissue, especially in increasing the compressive strength. Therefore, an increase in the number of papers reporting the addi-tion of this material to nano!brous scaffolds is being seen. Gelatin-HAp electrospun nano-!brous scaffolds were produced by Kim et al. (2005) to mimic the human bone matrix, and higher cellular activity was reported with these scaffolds, compared to those prepared with only gelatin. Zhang et al. (2008) have reported the use of electrospinning in combina-tion with an in situ coprecipitation synthesis in the preparation of nanocomposite !bers of HAp/chitosan (CHI). Production of continuous nano!bers was possible after addition of a !ber-forming polymer, ultrahigh molecular weight PEO. Continuous HAp/CHI nano!bers

K10254_C010.indd 206 5/12/11 7:18:07 PM

Page 13: Nanotechnology in Biomaterials: Nanofibers in Tissue ...biomed.metu.edu.tr/sites/biomat.metu.edu.tr/files/Yucel et al, 2011.pdf · membranes (wound dressings, drug delivery systems,

Nanotechnology in Biomaterials: Nanofibers in Tissue Engineering 207

with a diameter of 214 25 nm were produced successfully, and the HAp nanoparticles were incorporated into the electrospun nano!bers. In vitro cell culture with hFOB showed that the incorporation of HAp nanoparticles into CHI nano!brous scaffolds led to signi!-cantly more bone formation than that with electrospun pure CHI scaffolds (Zhang et al., 2008). In another study, COL and HAp were combined in PLLA, forming three different scaffold types: PLLA, PLLA/HA, and PLLA/COL/HAp. In these scaffolds COL and HAp were expected to provide cell recognition sites and introduce osteoconductivity, an impor-tant issue in bone mineralization, respectively. Fiber diameter was found to decrease upon the addition of HAp and COL to PLLA. Human fetal osteoblasts were found to adhere and grow actively on PLLA/COL/HAp nano!bers, with mineral deposition increased by 57% in comparison to PLLA/HAp nano!bers (Prabhakaran et al., 2009).

Some other ingredients introduced to polymers to improve the biomechanical properties and material-cell interaction for bone tissue engineering applications are calcium carbon-ate (CaCO3) and -TCP nanoparticles. Fujihara and his co-workers (2005) used a guided bone regeneration (GBR) approach with the aim of producing a calcium-rich and mechani-cally stable membrane. This was achieved by the preparation of PCL nano!bers contain-ing CaCO3 nanoparticles. A PCL-based membrane was compared with that based on PCL/CaCO3, and a higher cell adherence was observed in the latter. The tensile strength of PCL nano!brous membranes decreased when the amount of calcium carbonate nanoparticles was increased; that is, an increase in the amount of CaCO3 resulted in more brittle mem-branes. However, it was stated that these membranes have suf!cient tensile strength for guided bone regeneration (Fujihara et al., 2005).

Functionally graded, nonwoven mats of PCL with -TCP nanoparticles were produced by using a hybrid, twin-screw extrusion/electrospinning (TSEE) process (Erisken et al., 2008). This procedure allows feeding of various liquid and solid materials, achieving melt-ing, dispersion, pressurization, and electrospinning within a single process, with the com-position changing in a time-dependent manner. The potential of these scaffolds for tissue engineering and bone-cartilage interface formation was demonstrated by using mouse calvaria-derived murine preosteoblast (MC3T3-E1) cells. The ability to control the incorpo-ration of -TCP resulted in a better mimicking of the compositional and structural charac-teristics of bone tissue (Erisken et al., 2008).

10.5.2 Nerve Tissue Engineering

The structure of the nervous system is important for its functionality; therefore, the archi-tecture of the nerve tissue should be well known in order to be able to design the most suit-able nerve substitute for nerve regeneration. In nervous tissue the axons are bundled into cables that provide long-distance communication between the brain and spinal cord and the rest of the body. The nerves in the peripheral nervous system (PNS) are composed of bundled motor and sensory axons in parallel arrays in connective tissue and are called the nerve trunk (Schmidt and Leach, 2003). The individual axons and their Schwann cell sheaths are surrounded by endoneurium, which is composed predominantly of oriented collagen !bers. Groups of axons are wrapped by the perinerium, which is composed of many layers of "attened cells (i.e., !broblasts) and collagen to form fascicles. An outer sheath of loose !brocollagenous tissue, the epineurium, assembles individual nerve fas-cicles into a nerve trunk. The vascularization in these nerves is provided by capillaries within the support tissue of the nerve trunk, or by vessels penetrating into the nerve from surrounding arteries and veins. Within the central nervous system (CNS), the brain and spinal cord, such bundled together axons are called nerve tracts. Typically, the neuron

K10254_C010.indd 207 5/12/11 7:18:07 PM

Page 14: Nanotechnology in Biomaterials: Nanofibers in Tissue ...biomed.metu.edu.tr/sites/biomat.metu.edu.tr/files/Yucel et al, 2011.pdf · membranes (wound dressings, drug delivery systems,

208 Bionanotechnology II: Global Prospects

consists of dendrites, an axon, and a cell body (Schmidt and Leach, 2003). A gray matter localized in the center of the spinal cord is composed of the cell bodies of excitatory neu-rons, glial cells, and blood vessels. The gray matter is surrounded by white matter, which provides protection and insulation of the spinal cord. White matter consists of axons and glial cells, such as oligodendrocytes, astrocytes, and microglia. Oligodendrocytes myeli-nate the axons in the CNS, while astrocytes are active in the blood-nerve barrier, separat-ing the CNS from blood proteins and cells. Fascicles, axons in bundles, come out from the white matter and then exit the encasing bone of the spinal column. Following travel through the PNS-CNS transition zone, nerves enter the PNS.

Nervous system injuries may result in permanent functional disabilities, and thus have a signi!cant impact on the patient’s quality of life. In severe damage of the peripheral nervous system, self-regeneration of the tissue is dif!cult, and it is almost impossible in the central nervous system (Meek et al., 2002; Schmidt and Leach, 2003). For that reason, the repair of the damaged tissue in the nervous system is complicated. Surgical inter-ventions (end-to-end anastomosis) are used for small nerve gaps (Sanghvi et al., 2004); however, for long gaps (longer than 5 mm) the common treatment is to use autografts. The inherent drawbacks of autografts, such as limited supply, permanent loss of the nerve function at the donor site, and the requirement of a second surgery, are great challenges for researchers and compel them to investigate alternative treatments (Millesi, 1991; Terzis et al., 1997).

The rapidly developing alternative, nerve tissue engineering, aims to restore the function of damaged neural tissue through use of native cells and cell carriers. The cells commonly used in nerve tissue engineering are mature neurons, stem cells, and supportive cells, such as Schwann cells, astrocytes, and oligodendrocytes. The construct developed by nerve tis-sue engineering should imitate the anatomic structure of the nervous system for proper performance. When the native architecture of the nerve tissue is considered, it becomes apparent that a directional cell growth is a prerequisite for functional nerve regeneration. The desired physical properties of a cell carrier are biodegradability, high porosity, and also a cell guiding tool, such as an internal oriented matrix with appropriate chemical structure (ECM proteins, especially laminin, for nerve tissue). Topographical cues for cell migration could serve to mimic the native nerve fascicles (Hudson et al., 1999). The guid-ance needed for the tissue-engineered nerve construct may be achieved by the use of !lms with patterned surfaces, or longitudinally oriented microchannels and !brous scaffolds.

Fibrous scaffolds to be used in nerve regeneration could be produced by electrospinning and self-assembly (Table 10.2). Since they are inherently porous and have a high surface area-to-volume ratio, these !brous scaffolds promote attachment, survival, and growth of cells (Chiu et al., 2005). An architecture similar to ECM makes !brous scaffolds a promis-ing tool for nerve tissue engineering. The ideal scaffold should be biocompatible to avoid in"ammatory response, be biodegradable, and have appropriate mechanical properties to provide the support required for proper healing (Hudson et al., 1999). The material of the cell carrier is a very important determinant in achieving a scaffold suitable for nerve tissue engineering. The degradation rate and mechanical properties of synthetic poly-mers such PLLA, PLGA, PHBV, and PCL can be controlled by chosing the appropriate compound, and as such, they are very suitable for tissue engineering. Natural materi-als such as PHBV, collagen, gelatin, chitosan, and especially laminin have speci!c physi-cal properties and biomolecular recognition sites as in native tissue and, therefore, they have been increasingly investigated as !brous scaffolds for nerve tissue regeneration (Cao et al., 2009). Blends of polymers have also been extensively used in nerve tissue engineer-ing to achieve the desired scaffold properties. For instance, natural polymers lack proper

K10254_C010.indd 208 5/12/11 7:18:07 PM

Page 15: Nanotechnology in Biomaterials: Nanofibers in Tissue ...biomed.metu.edu.tr/sites/biomat.metu.edu.tr/files/Yucel et al, 2011.pdf · membranes (wound dressings, drug delivery systems,

Nanotechnology in Biomaterials: Nanofibers in Tissue Engineering 209TA

BLE

10.

2N

ano/

Mic

ro!b

rous

Tex

ture

s U

sed

for

Ner

ve T

issu

e En

gine

erin

g

Poly

mer

Typ

e

Poly

mer

C

once

ntra

tion

/C

ompo

siti

on/S

olve

ntFi

ber

Dia

met

erIn

Vit

ro S

tudi

es:

Cel

l Typ

eIn

Viv

o St

udie

s:N

erve

Inj

ury

Mod

elR

efer

ence

Fibe

rs F

abri

cate

d by

El

ectr

ospi

nnin

g

Lam

inin

3–8%

w/v

HFP

90–3

00 n

m (R

F)H

uman

adi

pose

ste

m

cells

and

PC

12 c

ells

—N

eal e

t al.,

200

9

Lam

inin

-PLL

A b

lend

10%

w/v

; 1:2

50, w

:wH

FP10

0–50

0 nm

(RF)

Rat

PC

12 c

ells

—K

oh e

t al.,

200

8

PCL

10%

w/v

CH

L:M

eOH

3:1,

v:v

750

100

nm

(RF)

Brai

n-de

rive

d ne

ural

st

em c

ells

—N

isbe

t et a

l., 2

008

PCL-

pp*

PCL-

PPPC

L-PP

16%

w/w

DC

M:M

eOH

8:2

, v:v

2.26

0

.08

µm (R

F)1.

03

0.0

3 µm

(AF)

Hum

an S

chw

ann

cells

—C

hew

et a

l., 2

008

CH

L:M

eOH

3:1,

v:v

100–

500

nm (R

F)M

ouse

(129

str

ain)

embr

yoni

c st

em c

ells

—Th

ouas

et a

l., 2

008

20%

w/v

DC

M: D

MF

80:2

0, v

:v25

0 nm

(RF/

AF)

Mou

se e

mbr

yoni

c st

em

cells

CE3

and

RW

4—

Xie

et a

l., 2

009b

10–2

0% w

/vD

CM

:DM

F22

0 3

6 nm

(50

3 n

m s

hell

thic

knes

s)C

hick

em

bryo

nic

(E8)

do

rsal

root

gan

glia

ex

plan

t

—X

ie e

t al.,

200

9a

PCL/

GEL

10%

w/v

; 1:1

, w:w

TFE

232

194

nm

(RF)

160

86

nm (A

F)R

at S

chw

ann

cell

line

(RT4

-D6P

2T)

—G

upta

et a

l., 2

009

PCL/

CH

I6%

w/w

; 70:

30, w

:wH

FP18

9 5

6 nm

(RF/

AF)

Neo

nata

l mou

se

cere

bellu

m C

17.2

ste

m

cells

—G

hase

mi-M

obar

akeh

et

al.,

200

8

PCL:

CH

I, 75

:25,

w:w

PCL

in H

FIP,

CH

I in

TFA

/DC

M

190

26

nm (R

F)R

at S

chw

ann

cell

line

RT4-

D6P

2T—

Prab

haka

ran

et a

l.,

2008

* PC

L-PP

: PC

L co

ated

with

pol

ypyr

role

(con

tinue

d )

K10254_C010.indd 209 5/12/11 7:18:07 PM

Page 16: Nanotechnology in Biomaterials: Nanofibers in Tissue ...biomed.metu.edu.tr/sites/biomat.metu.edu.tr/files/Yucel et al, 2011.pdf · membranes (wound dressings, drug delivery systems,

210 Bionanotechnology II: Global ProspectsTA

BLE

10.

2 (C

ON

TIN

UED

)

Nan

o/M

icro

!bro

us T

extu

res

Use

d fo

r N

erve

Tis

sue

Engi

neer

ing

Poly

mer

Typ

e

Poly

mer

C

once

ntra

tion

/C

ompo

siti

on/S

olve

ntFi

ber

Dia

met

erIn

Vit

ro S

tudi

es:

Cel

l Typ

eIn

Viv

o St

udie

s:N

erve

Inj

ury

Mod

elR

efer

ence

PLLA

PLLA

3 w

/w%

CH

L52

4 3

05 n

m (R

F/A

F)D

orsa

l roo

t gan

glia

ex

plan

t—

Cor

ey e

t al.,

200

7

1% w

/w3%

w/w

2% w

/w5%

w/w

DC

M:D

MF

70:3

0, v

:v

250

nm (R

F)1.

25 µ

m (R

F)30

0 nm

(AF)

1.5

µm (A

F)

Neo

nata

l mou

se

cere

bellu

m C

17.2

ste

m

cells

—Ya

ng e

t al.,

200

5

PLLA

8% w

/wC

HL

1.2–

1.6

#m (A

F)Em

bryo

nic

(E9)

chi

ck

dors

al ro

ot g

angl

ia

expl

ants

and

rat

Sc

hwan

n ce

lls

—W

ang

et a

l., 2

009

PLLA

/ PL

GA

10%

w/v

THF

and

DM

F51

0 3

80 n

m (P

LLA

, RF)

760

300

nm

(PLG

A, R

F)M

ouse

em

bryo

nic

cort

ical

neu

rons

—N

isbe

t et a

l., 2

007

PLG

A, 7

5/25

(coa

ted

with

pol

ypyr

role

)6.

5% w

/w (R

F)7.

0% w

/w (A

F)H

FIP

520

150

nm

(RF)

430

180

nm

(AF)

(85

41

nm s

hell

thic

knes

s)

PC12

cel

ls a

nd r

at

embr

yoni

c hi

ppoc

ampa

l ne

uron

s

—Le

e et

al.,

200

9

Poly

(-c

apro

lact

one-

co-e

thyl

eth

ylen

e ph

osph

ate)

(PC

LEEP

)

12%

w/w

DC

M5.

08

0.0

5 #m

(AF)

—15

mm

ner

ve le

sion

gap

in

rat s

ciat

ic n

erve

(cel

l-fre

e)C

hew

et a

l., 2

007

Blen

d of

PLG

A 7

5:25

/PC

L5.

5% w

/w P

CL:

4% w

/w P

LGA

CH

L:M

eOH

3:1

279

87

nm (R

F)—

10 m

m n

erve

gap

in r

at

scia

tic n

erve

(cel

l-fre

e)Pa

nser

i et a

l., 2

008

Fibe

rs F

abri

cate

d by

Se

lf-A

ssem

bly

RA

D16

-I*

(Arg

-Ala

-A

sp) a

nd R

AD

16-I

I**

Cel

l cul

ture

med

ia10

–20

nmR

at P

C12

cel

ls

Mou

se h

ippo

cam

pal a

nd

cere

bella

r gr

anul

e ne

uron

sR

at h

ippo

cam

pal

neur

ons

Inje

ctio

n of

sel

f-as

sem

bled

(2

0 µl

) in

rat r

ight

leg

(cel

l-fre

e)

Hol

mes

et a

l., 2

000

K10254_C010.indd 210 5/12/11 7:18:07 PM

Page 17: Nanotechnology in Biomaterials: Nanofibers in Tissue ...biomed.metu.edu.tr/sites/biomat.metu.edu.tr/files/Yucel et al, 2011.pdf · membranes (wound dressings, drug delivery systems,

Nanotechnology in Biomaterials: Nanofibers in Tissue Engineering 211IK

VAV

(I

le–L

ys–V

al–A

la–V

al)

Cel

l cul

ture

med

ia5–

8 nm

Mou

se (E

13) n

eura

l pr

ogen

itor

cells

Spin

al c

ord

inje

ctio

n (T

10)

at a

dep

th o

f 1.5

mm

in

rats

(cel

l fre

e)

Silv

a et

al.,

200

4

RA

DA

16-

I (A

rg–A

la–A

sp–A

la)

Mill

i-Q w

ater

10 n

m—

1.5–

2 m

m d

eep

cut w

ound

in

the

optic

trac

t, su

peri

or

colli

culu

s in

ham

ster

bra

in

(cel

l fre

e)

Ellis

-Beh

nke

et a

l.,

2006

RA

DA

16-I

pep

tide

Cel

l cul

ture

med

ia10

nm

Rat

neu

ral p

roge

nito

r ce

lls a

nd S

chw

ann

cells

1 m

m ti

ssue

rem

oval

by

a tr

anse

ctio

n in

spi

nal c

ord

dors

al c

olum

n (b

etw

een

C6

and

C7)

in r

ats

Guo

et a

l., 2

007

Abb

revi

atio

ns: R

F, r

ando

m !

ber;

AF,

alig

ned

!ber

; HFP

, 1,1

,1,3

,3,3

-hex

a"uo

ro-2

-pro

pano

l; PC

L, p

oly(

-cap

rola

cton

e); C

HL,

chl

orof

orm

; MeO

H, m

etha

nol;

DC

M,

dich

loro

met

hane

; DM

F, d

imet

hylf

orm

amid

e; G

EL, g

elat

in; T

FE, 2

,2,2

-tri

"uor

oeth

anol

; CH

I, ch

itos

an; H

FIP,

hex

a"uo

ro-2

-pro

pano

l; TH

F, te

trah

ydro

fura

n;

TFA

, tri

"uor

oace

tic

acid

; PLL

A, p

oly(

L-la

ctic

aci

d); P

LGA

, pol

y(la

ctic

aci

d-co

-gly

colic

aci

d).

K10254_C010.indd 211 5/12/11 7:18:08 PM

Page 18: Nanotechnology in Biomaterials: Nanofibers in Tissue ...biomed.metu.edu.tr/sites/biomat.metu.edu.tr/files/Yucel et al, 2011.pdf · membranes (wound dressings, drug delivery systems,

212 Bionanotechnology II: Global Prospects

mechanical properties and also lose mechanical strength very quickly due to degradation, while synthetic polymers alone are generally too hydrophobic and lack binding sites for cell adhesion (Gupta et al., 2009). Consequently, a blend or a graft of natural and synthetic polymers is an effective way to obtain scaffolds with desirable properties. A summary of nano/micro!brous scaffolds used in vitro and in vivo in nerve regeneration studies is given in Table 10.2.

Electrospinning is a straightforward technique to obtain random and aligned nano/micro!bers. Even though the randomly oriented !bers are easier to obtain and are more frequently used in nerve tissue engineering, the use of the aligned !bers is a challenging but rewarding approach in mimicking the unidirectional orientation to maintain func-tion of the nerve tissue (Table 10.2). Fibers aligned longitudinally (parallel to nerve axis) may provide a suitable environment that encourages contact guidance and the directional neuronal/axonal growth desired in nerve tissue engineering. Yang et al. (2005) observed the positive effect of contact guidance by aligned !brous scaffolds (diameters of aligned micro- and nano!bers: 1.5 µm and 300 nm, respectively) on neural stem cells that had been induced to elongate neurites parallel to the !ber axis. The cell elongation and the neurite outgrowth were random on the randomly oriented mats (diameters of random micro- and nano!bers: 1.25 µm and 250 nm, respectively), proving the signi!cance of guidance. This guidance was observed on aligned nano/micro!bers regardless of !ber diameter. The effect of !ber arrangement was also observed in our studies carried out with neural stem cells seeded on !brous mats. Electrospun PHBV-PLGA !bers were produced under opti-mized conditions (PHBV-PLGA solution: 1:1 w/w, 15% w/v, in chloroform: dimethylforma-mide (CHL:DMF)) with a diameter of ca. 1.5 µm (Yucel et al., 2010). The stem cells seeded on the aligned !bers were well oriented along the axis of the !bers, while the cells on the random !bers appeared to be in clusters distributed in all directions (Figure 10.2). Similar results were also obtained with dorsal root ganglion (DRG) explants cultured on PLLA nano!bers (Patel et al., 2007). Signi!cant extension of neurites was observed on aligned !bers, while there was no neurite outgrowth on randomly oriented !bers. Like other cells, human Schwann cells responded to the !ber orientation with elongation along the !bers

A B

20 µmCentral Laboratory

20 µmM

etu Central Laboratory

FIGURE 10.2CLSM images of neural stem cells on !bronectin-coated PHBV-PLGA scaffolds. (A) Randomly oriented and (B) aligned !bers (100 ; scale bar, 250 µm; two-sided arrow, direction of !ber axis; Acridine orange stain). Insets show the SEM images of the !brous mats.

K10254_C010.indd 212 5/12/11 7:18:08 PM

Page 19: Nanotechnology in Biomaterials: Nanofibers in Tissue ...biomed.metu.edu.tr/sites/biomat.metu.edu.tr/files/Yucel et al, 2011.pdf · membranes (wound dressings, drug delivery systems,

Nanotechnology in Biomaterials: Nanofibers in Tissue Engineering 213

(Chew et al., 2008). In addition, aligned electrospun !bers appeared to enhance Schwann cell maturation more than the randomly oriented !bers did, as was shown by the upregu-lation of the myelin-speci!c gene. In vitro results were con!rmed by the in vivo studies in which anatomical and functional measurements showed that constructs with aligned polymer !bers signi!cantly enhanced nerve regeneration in 17 mm peripheral nerve gaps in rats over those of randomly oriented !bers (Kim et al., 2008).

Micro- and nanoscale topography was shown to play an important role in neural stem cell differentiation. Yang et al. (2005) observed that the mouse neural stem cell differentia-tion rate might depend on the !ber dimension. The differentiation rate on nano!bers 300 nm in diameter was twice as high as on micro!bers with a 1.5 µm diameter. However, these results on the effect of !ber diameter on neural cell differentiation are controversial. Christopherson and his colleagues (2009) found that under differentiation conditions, rat neural stem cells stretched multidirectionally and preferentially differentiated into oligo-dendrocytes on !bers with a diameter of 283 nm, but they elongated along a single !ber axis and preferentially differentiated into neuronal lineage on !bers 749 nm and 1,452 nm thick. Despite these con"icting results, topographical and biological cues seem to be important in the control of stem cell behavior.

Electrospinning approaches involving blending, coaxial extrusion, and emulsion spin-ning are used in the incorporation of bioactive agents like proteins and growth factors to enhance cell attachment, growth, differentiation, and survival of cells in nerve tissue engineering applications. Incorporation of laminin, a neurite-promoting basement mem-brane protein, to a synthetic polymer by blending was reported as a more ef!cient method to produce biomimetic scaffolds than covalent immobilization and physical adsorption of the bioactive agents (Koh et al., 2008). In a recent study, nerve growth factor (NGF), a common neurotrophic factor involved in peripheral nerve regeneration, was incorporated in poly(L-lactide-co- -caprolactone) !bers by emulsion electrospinning (Li et al., 2010). Pheochromocytoma 12 (PC12) cells, which differentiate into a neuronal phenotype in the presence of NGF, were used in a study where NGF was released from !brous mats for a sustained period, revealing its bioactivity. A glial cell-derived neurotrophic factor (GDNF), important in motor neuron survival, was entrapped in aligned biodegradable polymeric !bers (a copolymer of -caprolactone and ethyl ethylene phosphate (PCLEEP)) (Chew et al., 2007). An in vitro release pro!le of GDNF showed that after an initial burst release of about 30%, the remaining GDNF was released in a fairly sustained manner for almost 2 months.

Electrical stimulation is another tool that affects neurite extension. Electrical stimula-tion using an external electrical !eld and an electroconductive polypyrrole (PPy) !lm was shown to enhance neurite extension in vitro (Schmidt et al., 1997). Xie et al. (2009a) cul-tured DRG explants on conductive (PPy) core-sheath nano!bers electrospun with PCL, and results showed that upon electrical stimulation, the rate of neurite extension was enhanced on both random and aligned nano!bers without any change in the direction of the neurites’ extension. In another study, PPy was deposited on electrospun PLGA !bers to investigate the response of PC12 cells (Lee et al., 2009). Upon application of 10 mV/cm on PPy-PLGA scaffolds, PC12 cells exhibited 40–50% longer neurites and 40–90% more neurite formation than unstimulated cells on the same scaffolds.

In conclusion, the topography of nano/micro!brous scaffolds closely resembles that of the natural ECM, and this is an important factor in tissue engineering. Considering the architecture of longitudinally oriented nerve tissue, uniaxially aligned !bers could create an appropriate environment for guided nerve regeneration. The use of the aforementioned tools with aligned !brous scaffolds could be a promising approach in the design of a bio-mimetic substitute for nerve regeneration.

K10254_C010.indd 213 5/12/11 7:18:08 PM

Page 20: Nanotechnology in Biomaterials: Nanofibers in Tissue ...biomed.metu.edu.tr/sites/biomat.metu.edu.tr/files/Yucel et al, 2011.pdf · membranes (wound dressings, drug delivery systems,

214 Bionanotechnology II: Global Prospects

10.5.3 Cardiac Muscle Tissue Engineering

The heart is the center of the circulation system and distributes blood throughout the body. The function of the heart is vital to supply oxygen and nutrients to, and remove waste products from, the body via the blood in order to maintain the balance that is nec-essary to sustain life (Vander et al., 1994). Strong muscular contractions in the ventricles pump blood out of the heart and into the circulatory system. These muscular contrac-tions are produced by the muscle tissue that makes up the walls of the ventricles. Healthy heart muscle wall is composed of three layers: a muscular sheet, the myocardium, lined on either side by two collagenous membranes (containing type I and type III collagen and elastin); the endocardium, which is populated by endothelial cells; and the epicardium, also called visceral pericardium (van de Graaff, 1998). The myocardium is the layer of functional beating muscle that consists of !broblasts and highly oriented cardiomyocytes (muscle !bers) in a matrix of collagen. The cardiomyocytes are connected end to end in the longitudinal direction and side by side in the transverse direction (Parker and Ingber, 2007). Collagen types I and III are the predominant interstitial collagens in the myocar-dium that generate structural integrity for the adjoining cardiomyocytes, providing the means by which myocyte shortening is translated into overall ventricular pump function. Basement membrane components include laminin, entactin, !bronectin, collagen type IV, and !brillin, and proteoglycans include chondroitin sulfate, dermatan sulfate, and hepa-ran sulfate (Kassiri and Khokha, 2005). The cardiomyocytes facilitate conduction of the electrical signals needed to initiate contractile movement in order to pump blood out of the ventricles (Vander et al., 1994). The elementary myocardial functional units are not lone cardiomyocytes, but rather are multicellular assemblies of these highly oriented cells. The cardiomyocytes are connected with intercalated discs that integrate individual electrical activation and contraction into a pumping action. Gap junctions in these intercalated discs allow the action potential to travel through the membranes of the myocytes, thus facilitat-ing signal propagation and a synchronized contractile pulse (Vander et al., 1994).

Heart failure, stemming from cardiovascular diseases, is the number one cause of death in industrialized countries. About 5 million people in the United States have heart failure, and the number is growing; heart failure contributes to or causes about 300,000 deaths each year. The inability of the heart to deliver suf!cient blood to meet the body’s meta-bolic requirements leads to heart failure. The most common causes are coronary artery disease and hypertension (high blood pressure), which damage the myocardium. Damage to any part of the intricate structure of the heart, though, can impair cardiac performance and result in heart failure; examples include diseases of the heart valves, the electrical conduction system, or external pressure around the heart, due to constriction of the peri-cardial sac in which the heart is located (Jawad et al., 2007). Congenital heart disease is also a considerable problem worldwide, affecting approximately 1% of infants (Wu et al., 2006). Congenital heart defects such as atrial septal defect, ventricular septal defect, double outlet ventricles, and hypoplastic left heart syndrome are associated with aplastic, defec-tive, or necrotic myocardial structures, where patch closure, correction of the defect, or revascularization is usually required (Ko!dis et al., 2002). Although the currently available prostheses are adequate for restoring ventricular geometry and maintaining ventricular pressure, and thus may be life saving, they do not actively adapt to the physiological envi-ronment and mechanical demands, as they are nonliving materials. When implanted into the immature heart of a child, these materials do not grow with the pediatric patient. Therefore, these prostheses have limited durability and are prone to infection, immuno-logic reactivity, and thrombosis, which often requires repeated operations (Mirensky and

K10254_C010.indd 214 5/12/11 7:18:08 PM

Page 21: Nanotechnology in Biomaterials: Nanofibers in Tissue ...biomed.metu.edu.tr/sites/biomat.metu.edu.tr/files/Yucel et al, 2011.pdf · membranes (wound dressings, drug delivery systems,

Nanotechnology in Biomaterials: Nanofibers in Tissue Engineering 215

Breuer, 2008). These constructs are also subject to obstructive tissue ingrowths and !brotic responses with shrinkage and calci!cation leading to graft failure (Endo et al., 2001). At that point, tissue engineering provides a new experimental approach aimed at vital, semi-autologous replacement structures with the capacity of growth and adaptability.

The negligible ability of adult cardiomyocytes to proliferate has triggered an intense search for progenitor cells that can be used in repairing damaged myocardium. It is clear that natural cardiomyocyte regeneration, including differentiation of progenitors residing in the myocardium or the recruitment of stem cells from outside (e.g., from endothelial cells or from the bone marrow), is insuf!cient to overcome cardiomyocyte death in the acutely or chronically damaged heart (Nakamura and Schneider, 2003). The potential of cardiac stem cells resident in the heart, embryonic stem cells, bone marrow stem cells, and fetal stem cells (from tissues of umbilical cord) to differentiate into cardiomyogenic lineage either in vivo or in vitro in the presence of differentiation factors, conditioned media, or cocultures has been investigated, but there is still a need for elaboration and improvement in the procedures to be able to obtain high numbers of fully differentiated cardiomyocytes. Since the main concern in myocardial tissue engineering is the functionality of the !nal construct, the majority of in vitro studies use rat neonatal cardiomyocytes or cardiomyo-cytes derived from embryonic stem cells that are already able to contract.

Fibrous scaffolds whose !ber diameter ranges from several microns to submicron sizes (Table 10.3) are increasingly gaining importance over spongy scaffolds with random pores used to obtain tissue-engineered cardiac muscle. The main obstacle in overcoming the issues involved in the engineering of cardiovascular grafts is production of substitutes that can withstand the pulsation (i.e., have adequate shape recovery), the high pressure, and the "ow rate of the blood (Nisbet et al., 2009). An additional challenge in cardiovascu-lar tissue engineering is the necessity for mechanical and biological compliance. Polymers naturally found in the cardiac tissue, like collagen and elastin, are good choices as scaf-fold materials, since they carry attachment sites for cells, and especially elastin has elastic properties, but they have poor mechanical properties. On the other hand, synthetic poly-mers have higher mechanical strength, but have problems, such as being too hydrophobic and rigid, lacking cell binding functional groups, or having inaproppriate degradation rates (rapidly degrading or nondegrading). Thus, as mentioned earlier, blends or copoly-mers of synthetic or natural polymers are employed often in the production of scaffolds with the desired properties (Zong et al., 2005, Li et al., 2006a, 2006b). Currently cell survival in 3D cardiac grafts of spongy design is a critical issue; generally the cells are concentrated in the outer regions of the grafts and are scarce in the inside due to restricted transporta-tion of nutrients and waste in and out of the scaffold, resulting in an undesired core effect (Bursac et al., 1999). Electrospinning enables us to fabricate highly porous scaffolds with extensively interconnected pores that allow cell-to-cell contact and migration in all direc-tions, facilitate the transportation of nutrients and metabolites, and encourage blood vessel formation. All of these combined help cell survival throughout the scaffold, including the central portion. Electrospun mats of PCL with random, unaligned !bers were seeded with cardiomyocytes by Ishii et al. (2005), and the mats were stacked to generate a 3D cardiac graft. Five layers of these mats were successfully stacked without any indication of core ischemia. The individual layers adhered intimately, morphological and electrical commu-nication between the layers was established, and synchronized beating was also observed. This study demonstrated the feasibility of obtaining a functional 3D cardiac graft from cell sheets supported by nano!bers mimicking the native structure of cardiac ECM. Fiber density on the electrospun mats could be tailored depending on the intended use. For example, electrospun mats with tight !ber spacing may act as a sieve to prevent blood cell

K10254_C010.indd 215 5/12/11 7:18:08 PM

Page 22: Nanotechnology in Biomaterials: Nanofibers in Tissue ...biomed.metu.edu.tr/sites/biomat.metu.edu.tr/files/Yucel et al, 2011.pdf · membranes (wound dressings, drug delivery systems,

216 Bionanotechnology II: Global ProspectsTA

BLE

10.

3

Nan

o/M

icro

!bro

us T

extu

res

Use

d in

Car

diac

Mus

cle

Tiss

ue E

ngin

eeri

ng

Poly

mer

Typ

ePo

lym

er C

once

ntra

tion

/C

ompo

siti

on/S

olve

ntFi

ber

Dia

met

erIn

Vit

ro S

tudi

es: C

ell T

ype

Ref

eren

ce

Extr

usio

nPG

A—

13 #

mN

eona

tal r

at c

ardi

omyo

cyte

sBu

rsac

et a

l., 1

999

Papa

daki

et a

l., 2

001

Col

lage

n ty

pe I

25 m

g/m

lD

I wat

er-H

Cl

(pH

2.5

–3.0

)

~ 25

0 nm

(AF)

Rat

em

bryo

nic

card

iac

myo

cyte

sEv

ans

et a

l., 2

003

Yost

et a

l., 2

004

Elec

tros

pinn

ing

PCL

10%

(w/w

)C

HL:

MeO

H (1

:1)

100

nm–5

#m

(RF)

Neo

nata

l rat

car

diom

yocy

tes

Shin

et a

l., 2

004

Ishi

i et a

l., 2

005

PLLA

Blen

d of

PG

LA (9

0:10

)/PL

LABl

end

of P

LGA

(75:

25)/

PEG

-PD

LLA

30–5

0% (w

/w)

DM

F0.

9–1

#m (R

F/A

F)N

eona

tal r

at c

ardi

omyo

cyte

sZ

ong

et a

l., 2

005

Blen

d of

PLG

A/g

elat

in/

-ela

stin

10%

PLG

A:8

% g

elat

in: 2

0%

elas

tin (w

/v) H

FP38

0 8

0 nm

(RF)

H9c

2 ra

t car

diac

myo

blas

tsN

eona

tal r

at b

one

mar

row

str

omal

cel

lsLi

et a

l., 2

006a

Blen

ds o

f PA

Ni/

gela

tin3%

(w/v

) CPS

A-P

AN

i8%

(w/v

) gel

atin

H

FP

From

803

1

21 n

m to

61

1

3nm

(RF)

H9c

2 ra

t car

diac

myo

blas

tsLi

et a

l., 2

006b

Biod

egra

dabl

e po

lyur

etha

ne15

% (w

/v)

DC

M2–

10 #

m (R

F)M

ouse

ESC

-der

ived

car

diom

yocy

tes

C2C

12 s

kele

tal m

yobl

ast l

ine

HL-

1 ca

rdio

myo

cyte

cel

l lin

e

From

stei

n et

al.,

200

8

Biod

egra

dabl

e po

lyur

etha

ne18

% (w

/v)

DC

M1.

8 1

.26

#m (A

F)2.

0 1

.92

#m (R

F)N

eona

tal r

at c

ardi

omyo

cyte

sR

ockw

ood

et a

l., 2

008

Blen

d of

PH

BV/P

(L-D

,L)L

A/P

GS

5% (w

/v)

PHBV

:P(L

-D,L

)LA

:PG

S 49

:49:

2 (w

/w)

CH

L:D

MF

(10:

1)

1.10

–1.2

5 #m

(AF)

Hum

an W

hart

on’s

jelly

MSC

sK

enar

et a

l., 2

009

Abb

revi

atio

ns: P

GA

, pol

y(gl

ycol

ic a

cid)

; PC

L, p

oly(

-cap

rola

cton

e); P

LLA

, pol

y(L-

lact

ide)

; PG

LA, p

oly(

glyc

olid

e-co

-lac

tide

); PL

GA

, pol

y(la

ctid

e-co

-gly

colid

e);

PEG

, pol

y(et

hyle

ne g

lyco

l); P

DLL

A, p

oly(

D,L

-lac

tide

); PH

BV, p

oly(

3-hy

drox

ybut

yrat

e-co

-3-h

ydro

xyva

lera

te);

P(L-

D,L

)LA

, pol

y(L-

D,L

-lac

tic

acid

); PG

S,

poly

(gly

cero

l se

baca

te);

CH

L, c

hlor

ofor

m;

MeO

H,

met

hano

l; D

MF,

N,N

-dim

ethy

l fo

rmam

ide;

HFP

, 1,

1,1,

3,3,

3-he

xa"u

oro-

2-pr

opan

ol;

DC

M,

dich

lo-

rom

etha

ne; P

AN

i, po

lyan

iline

(con

duct

ive

poly

mer

).

K10254_C010.indd 216 5/12/11 7:18:08 PM

Page 23: Nanotechnology in Biomaterials: Nanofibers in Tissue ...biomed.metu.edu.tr/sites/biomat.metu.edu.tr/files/Yucel et al, 2011.pdf · membranes (wound dressings, drug delivery systems,

Nanotechnology in Biomaterials: Nanofibers in Tissue Engineering 217

leakage or provide a homogeneous surface to obtain intact endothelial cell sheets, or act as a supporting outer surface in cardiovascular scaffolds (Hong et al., 2009; Soletti et al., 2010). Conversely, !ber spacing and density may also be adjusted and loosened to promote cell in!ltration in the myocardial patches.

Anisotropy in the architecture of the constructs plays an important role in achieving compliance and functionality in cardiovascular tissue engineering. Parallel submicron !bers may be produced to have circumferential orientations similar to those of the cells and !brils of the medial layer of the native artery (Xu et al., 2004). It was demonstrated that anisotropic poly(ester urethane)urea scaffolds exhibit mechanical properties closely resem-bling those of the native pulmonary heart valve lea"ets (Courtney et al., 2006). Cardiac muscle contracts like a syncytium owing to the multicellular assembly of myo!bers ori-ented parallel to each other, connected end to end via intercalated discs in the longitudinal direction and connected side to side in the transverse direction. It is this cellular arrange-ment that integrates individual contraction into a synchronous pumping action, whereas myo!ber disarrays are known to cause arrhythmia. It was shown by Black et al. (2009) that cardiomyocyte alignment augments the twitch force. Prior to emergence of techniques to produce aligned nano/micro!brous scaffolds, anisotropy had been introduced to the car-diomyocytes on the polymer scaffolds in three different ways: (1) by micropatterning (e.g., using biodegradable, elastomeric polyurethane !lms patterned by microcontact printing of laminin (McDevitt et al., 2003)), (2) by cyclic stretching (e.g., stretching of cardiomyo-cyte-seeded scaffolds (Akhyari et al., 2002; Zimmermann et al., 2002, 2004; Zimmermann and Eschenhagen, 2003; Fedak et al., 2003)), and (3) by applying electrical signals designed to mimic those in the native heart to the tissue engineered constructs (e.g., rat cardiomyo-ctes resuspended in Matrigel were seeded onto collagen sponges (Radisic et al., 2004)). Electrospinning, on the other hand, offers a much simpler way to achieve the anisotropy and allows greater control over composition, mechanical properties, and structure of a graft, thus making it easier to match the properties of the scaffold and the native tissue. It was shown by Zong et al. (2005) that cardiomyocytes can align parallel to micron-size !bers. Their electrospun mats with oriented !bers of PLLA and PLGA were used to assess the in"uence of scaffolds on primary cardiomyocyte attachment, structure, and function. The primary cardiomyocytes showed a preference for relatively hydrophobic surfaces (PLLA), where they aligned along the direction of !bers and developed mature contractile machinery (sarcomers). The only drawback of these aligned !ber scaffolds was the inabil-ity of the cardiomyocytes to penetrate into the scaffold due to the inadequate space among the !bers. A very similar phenomenon was reported by Evans et al. (2003), who observed that rat embryonic cardiac myocytes were unable to penetrate the 3D aligned collagen !ber scaffold with a pore size of 1–10 microns on the outer surface of a tubular scaffold. Aligned !ber scaffolds with a !ber density suitable for cell penetration were produced by our group by using a frame collector to obtain aligned !bers (Kenar et al., 2010). At least eight or nine Wharton’s jelly (WJ) MSC layers could be obtained in a single mat of average thickness of 12 3 #m. Both cytoskeletal and nuclear alignments of the cells were observed (Figure 10.3). A wrapping method was used to obtain a thicker homogeneous construct, which maintained its original anisotropic property. Another technique, microintegra-tion, was developed by Stankus et al. (2006), where simultaneous electrospraying of cells and electrospinning of poly(ester urethane)urea (PEUU) was utilized in order to fabricate thicker constructs with more uniform cell incorporation for use in cardiovascular tissue engineering applications. Smooth muscle cell-microintegrated PEUU was strong, "exible, and anisotropic, and Trypan blue staining revealed no signi!cant decrease in cell viability as a result of the fabrication process.

K10254_C010.indd 217 5/12/11 7:18:09 PM

Page 24: Nanotechnology in Biomaterials: Nanofibers in Tissue ...biomed.metu.edu.tr/sites/biomat.metu.edu.tr/files/Yucel et al, 2011.pdf · membranes (wound dressings, drug delivery systems,

218 Bionanotechnology II: Global Prospects

Studies carried out with aligned !ber mats with !ber diameters ranging from 0.25 to 3.00 #m show that cells grown on them align equally well along the direction of the !bers (Evans et al., 2003; Yost et al., 2004; Zong et al., 2005; Rockwood et al., 2008; Kenar et al., 2010). The single study carried out on cardiomyocyte function on electrospun random and aligned polymer !bers showed that cellular alignment enhances cardiomyocyte matura-tion; cells grown on aligned electrospun polyurethanes had signi!cantly lower steady-state levels of atrial natriuretic peptide (ANP) and, as a result, released less ANP over time than cardiomyocytes on random !bers (Rockwood et al., 2008).

In conclusion, nano!bers are novel scaffold materials for tissue engineering, and the accumulating data show them to be superior to most other designs due to the versatility of the approach. They are especially suited to the highly organized nature of biological tissues. With developments in the processing systems, they will become the major scaffold type.

ReferencesAgarwal S, Wendorff JH, Greiner A. 2009. Progress in the !eld of electrospinning for tissue engineer-

ing applications. Adv Mater 21: 3343–3351.Akhyari P, Fedak PW, Weisel RD, Lee TY, Verma S, Mickle DA, Li RK. 2002. Mechanical stretch

regimen enhances the formation of bioengineered autologous cardiac muscle grafts. Circulation 106: I137–I142.

Ashammakhi N, Ndreu A, Nikkola L, Wimpenny I, Yang Y. 2008. Advancing tissue engineering by using electrospun nano!bers. Regen Med 3: 547–574.

(A) (B)

FIGURE 10.3(See color insert.) Cross-sectional (X-axis ~ 60 tilted) CLSM images of FITC-Phalloidin and propidium iodide-stained WJ MSCs on a single, electrospun, aligned !ber mat of PHBV:P(L-D,L)LA:PGS (poly(glycerol sebacate)) (100 ) showing (A) F-actin and (B) nuclear alignment (two-sided arrow, direction of aligned !ber axis). Top images are the magni!ed versions of insets.

K10254_C010.indd 218 5/12/11 7:18:09 PM

Page 25: Nanotechnology in Biomaterials: Nanofibers in Tissue ...biomed.metu.edu.tr/sites/biomat.metu.edu.tr/files/Yucel et al, 2011.pdf · membranes (wound dressings, drug delivery systems,

Nanotechnology in Biomaterials: Nanofibers in Tissue Engineering 219

Ashammakhi N, Ndreu A, Piras AM, Nikkola L, Sindelar T, Ylikauppila H, Harlin A, Gomes ME, Neves NM, Chiellini E, Chiellini F, Hasirci V, Redl H, Reis RL. 2007a. Biodegradable nanomats produced by electrospinning: expanding multifunctionality and potential for tissue engineer-ing. J Nanosci Nanotechnol 7: 862–882.

Ashammakhi N, Ndreu A, Yang Y, Ylikauppila H, Nikkola L, Hasirci V. 2007b. Tissue engineering: a new take-off using nano!ber-based scaffolds. J Craniofac Surg 18: 3–17.

Asran AS, Henning S, Michler GH. 2010. Polyvinyl alcohol-collagen-hydroxyapatite biocomposite nano!brous scaffold: mimicking the key features of natural bone at the nanoscale level. Polymer 51: 868–876.

Badami AS, Kreke MR, Thompson MS, Rif"e JS, Goldstein AS. 2006. Effect of !ber diameter on spreading, proliferation, and differentiation of osteoblastic cells on electrospun poly(lactic acid) substrates. Biomaterials 27: 596–606.

Barhate RS, Loong CK, Ramakrishna S. 2006. Preparation and characterization of nano!brous !lter-ing media. J Membr Sci 283: 209–218.

Barnes CP, Sell SA, Boland ED, Simpson DG, Bowlin GL. 2007. Nano!ber technology: designing the next generation of tissue engineering scaffold. Adv Drug Delivery Rev 59: 1413–1433.

Black J. 1999. Biological performance of materials, 3rd ed. M. Dekker, New York, P 6.Black LD, Meyers JD, Weinbaum JS, Shvelidze YA, Tranquillo RT. 2009. Cell-induced alignment aug-

ments twitch force in !brin gel-based engineered myocardium via gap junction modi!cation. Tissue Eng A 15(10): 3099–3108.

Bruck SD. 1980. Properties of biomaterials in the physiological environment. CRC Press, Boca Raton, FL.Burg KJL, Porter S, Kellam JF. 2000. Biomaterial developments for bone tissue engineering. Biomaterials

21: 2347–2359.Bursac N, Papadaki M, Cohen RJ, Schoen FJ, Eisenberg SR, Carrier RL, Vunjak-Novakovic G, Freed

LE. 1999. Cardiac muscle tissue engineering: toward an in vitro model for electrophysiological studies. Am J Physiol Heart Circ Physiol 277: H433–H444.

Cao H, Liu T, Chew SY. 2009. The application of nano!brous scaffolds in neural tissue engineering. Adv Drug Deliv Rev 61(12): 1055–1064.

Catledge SA, Vohra YK, Bellis SL, Sawyer AA. 2004. Mesenchymal stem cell adhesion and spreading on nanostructured biomaterials. J Nanosci Nanotechnol 4: 986–989.

Chew SY, Mi R, Hoke A, Leong KW. 2007. Aligned protein-polymer composite !bers enhance nerve regeneration: a potential tissue-engineering platform. Adv Funct Mater 17(8): 1288–1296.

Chew SY, Mi R, Hoke A, Leong KW. 2008. The effect of the alignment of electrospun !brous scaffolds on Schwann cell maturation. Biomaterials 29(6): 653–661.

Chiu JB, Luu YK, Fang D, Hsiao B, Chu B, Hadjiargyrou M. 2005. Electrospun nano!brous scaffolds for biomedical applications. J Biomed Nanotechnol 1: 115–132.

Choi JS, Lee SJ, Christ GJ, Atala A, Yoo JJ. 2008. The in"uence of electrospun aligned poly( capro-lactone)/collagen nano!ber meshes on the formation of self-aligned skeletal muscle myotubes. Biomaterials 29: 2899–2906.

Christopherson GT, Song H, Mao HQ. 2009. The in"uence of !ber diameter of electrospun substrates on neural stem cell differentiation and proliferation. Biomaterials 30(4): 556–564.

Corey JM, Lin DY, Mycek KB, Chen Q, Samuel S, Feldman EL, Martin DC. 2007. Aligned electrospun nano!bers specify the direction of dorsal root ganglia neurite growth. J Biomed Mater Res A 83(3): 636–645.

Courtney T, Sacks MS, Stankus J, Guan J, Wagner WR. 2006. Design and analysis of tissue engineer-ing scaffolds that mimic soft tissue mechanical anisotropy. Biomaterials 27: 3631–3638.

Cui W, Li X, Zhou S, Weng J. 2007. In situ growth of hydroxyapatite within electrospun poly(DL-lactide) !bers. J Biomed Mater Res A 82(4): 831–841.

Deitzel JM, Kleinmeyer J, Harris D, Tan NCB. 2001. The effect of processing variables on the mor-phology of electrospun nano!bres and textiles. Polymer 42: 261–272.

Dieudonné SC, van den Dolder J, de Ruijter JE, Paldan H, Peltola T, van’t Hof MA, Happonen RP, Jansen JA. 2002. Osteoblast differentiation of bone marrow stromal cells cultured on silica gel and sol–gel-derived titania. Biomaterials 23: 3041–3051.

K10254_C010.indd 219 5/12/11 7:18:09 PM

Page 26: Nanotechnology in Biomaterials: Nanofibers in Tissue ...biomed.metu.edu.tr/sites/biomat.metu.edu.tr/files/Yucel et al, 2011.pdf · membranes (wound dressings, drug delivery systems,

220 Bionanotechnology II: Global Prospects

Dogan S, Demirer S, Kepenekci I, Erkek B, Kiziltay A, Hasirci N, Müftüo ̌glu S, Nazikoglu A, Renda N, Dincer UD, Elhan A, Kuterdem E. 2009. Epidermal growth factor-containing wound closure enhances wound healing in non-diabetic and diabetic rats. Int Wound J 6: 107–115.

Ducheyne P, Qiu Q. 1999. Bioactive ceramics: the effect of surface reactivity on bone formation and bone cell function. Biomaterials 20: 2287–2303.

Ekaputra AK, Prestwich GD, Cool SM, Hutmacher DW. 2008. Combining electrospun scaffolds with electrosprayed hydrogels leads to three-dimensional cellularization of hybrid constructs. Biomacromolecules 9: 2097–2103.

Ellis-Behnke RG, Liang YX, You SW, Tay DK, Zhang S, So KF, Schneider GE. 2006. Nano neuro knit-ting: peptide nano!ber scaffold for brain repair and axon regeneration with functional return of vision. Proc Natl Acad Sci USA 103(13): 5054–5059.

Endo S, Saito N, Misawa Y, Sohara Y. 2001. Late pericarditis secondary to pericardial patch implanta-tion 25 years prior. Eur J Cardiothorac Surg 20: 1059–1060.

Erisken C, Kalyon DM, Wang H. 2008. Functionally graded electrospun polycaprolactone and -tricalcium phosphate nanocomposites for tissue engineering applications. Biomaterials 29:

4065–4073.Evans HJ, Sweet JK, Price RL, Yost M, Goodwin RL. 2003. Novel 3D culture system for study of car-

diac myocyte development. Am J Physiol Heart Circ Physiol 285: H570–H578.Fan H, Liu H, Toh SL, Goh JCH. 2009. Anterior cruciate ligament regeneration using mesenchymal

stem cells and silk scaffold in large animal model. Biomaterials 30: 4967–4977.Fedak PWM, Weisel RD, Verma S, Mickle DAG, Li R-K. 2003. Restoration and regeneration of fail-

ing myocardium with cell transplantation and tissue engineering. Semin Thorac Cardiovasc Surg 15(3): 277–286.

Feng L, Li S, Li K, Zhai J, Song Y, Yiang L. 2002. Superhydrophobic surface of aligned polyacrylo-nitrile nano!bers. Angew Chem Int Ed 41: 1221–1223.

Fromstein JD, Zandstra PW, Alperin C, Rockwood D, Rabolt JF, Woodhouse KA. 2008. Seeding biore-actor-produced embryonic stem cell-derived cardiomyocytes on different porous, degradable, polyurethane scaffolds reveals the effect of scaffold architecture on cell morphology. Tissue Eng A 14(3): 369–378.

Fujihara K, Kotaki M, Ramakrishna S. 2005. Guided bone regeneration membrane made of poly-caprolactone/calcium carbonate composite nano-!bers, Biomaterials 26: 4139–4147.

Ghasemi-Mobarakeh L, Prabhakaran MP, Morshed M, Nasr-Esfahani MH, Ramakrishna S. 2008. Electrospun poly(epsilon-caprolactone)/gelatin nano!brous scaffolds for nerve tissue engi-neering. Biomaterials 29(34): 4532–4539.

Grif!th LG. 2002. Emerging design principles in biomaterials and scaffolds for tissue engineering. Ann NY Acad Sci USA 961: 83–95.

Guelcher SA, Hollinger JO. 2006. An introduction to biomaterials. Taylor & Francis Group, CRC Press, Boca Raton, FL.

Guo J, Su H, Zeng Y, Liang YX, Wong WM, Ellis-Behnke RG, So KF, Wu W. 2007. Reknitting the injured spinal cord by self-assembling peptide nano!ber scaffold. Nanomedicine 3(4): 311–321.

Gupta D, Venugopal J, Prabhakaran MP, Dev VR, Low S, Choon AT, Ramakrishna S. 2009. Aligned and random nano!brous substrate for the in vitro culture of Schwann cells for neural tissue engineering. Acta Biomater 5(7): 2560–2569.

Hay ED. 1991. Cell biology of extracellular matrix, 2nd ed. Plenum Press, New York.Hench LL, Erthridge EC. 1982. Biomaterials—an interfacial approach, Vol. 4, A. Noordergraaf, ed.

Academic Press, New York.Hench LL, Polak JM. 2002. Third-generation biomedical materials. Science 295: 1014–1017.Holmes TC, de Lacalle S, Su X, Liu G, Rich A, Zhang S. 2000. Extensive neurite outgrowth and

active synapse formation on self-assembling peptide scaffolds. Proc Natl Acad Sci USA 97(12): 6728–6733.

Hong Y, Ye S-H, Nieponice A, Soletti L, Vorp DA, Wagner WR. 2009. A small diameter, !brous vas-cular conduit generated from a poly(ester urethane)urea and phospholipid polymer blend. Biomaterials 30: 2457–2467.

K10254_C010.indd 220 5/12/11 7:18:09 PM

Page 27: Nanotechnology in Biomaterials: Nanofibers in Tissue ...biomed.metu.edu.tr/sites/biomat.metu.edu.tr/files/Yucel et al, 2011.pdf · membranes (wound dressings, drug delivery systems,

Nanotechnology in Biomaterials: Nanofibers in Tissue Engineering 221

Hosseinkhani H, Hosseinkhani M, Tian F, Kobayashi H, Tabata Y. 2006. Ectopic bone formation in collagen sponge self-assembled peptide-amphiphile nano!bers hybrid scaffold in a perfusion culture bioreactor. Biomaterials 27: 5089–5098.

Hu J, Feng K, Liu X, Ma PX. 2009. Chondrogenic and osteogenic differentiations of human bone marrow-derived mesenchymal stem cells on a nano!brous scaffold with designed pore net-work. Biomaterials 30: 5061–5067.

Huang ZM, Zhang YZ, Kotaki M, Ramakrishna S. 2003. A review on polymer nano!bers by electro-spinning and their application in nanocomposites. Compos Sci Technol 63: 2223–2253.

Hudson TW, Evans GR, Schmidt CE. 1999. Engineering strategies for peripheral nerve repair. Clin Plast Surg 26: 617–628.

Ishii O, Shin M, Sueda T, Vacanti JP. 2005. In vitro tissue engineering of a cardiac graft using a degradable scaffold with an extracellular matrix–like topography. J Thorac Cardiovasc Surg 130: 1358–1363.

Jang JH, Castano O, Kim HW. 2009. Electrospun materials as potential platforms for bone tissue engineering. Adv Drug Deliv Rev 61: 1065–1083.

Janjanin S, Li WJ, Morgan MT, Shanti RM, Tuan RS. 2008. Mold-shaped, nano!ber scaffold-based car-tilage engineering using human mesenchymal stem cells and bioreactor. J Surg Res 149: 47–56.

Jawad H, Ali NN, Lyon AR, Chen QZ, Harding SE, Boccaccini AR. 2007. Myocardial tissue engineer-ing: a review. J Tissue Eng Regen Med 1(5): 327–342.

Jeong L, Yeo IS, Kim HN, Yoon YI, Jang DH, Yung SY, Min BM, Park WH. 2009. Plasma-treated silk !broin nano!bers for skin regeneration. Int J Biol Macromol 44: 222–228.

Jin HJ, Chen J, Karageorgiou V, Altman G.H, Kaplan DL. 2004. Human bone marrow stromal cell responses on electrospun silk !broin mats. Biomaterials 25(6): 1039–1047.

Kassiri Z, Khokha R. 2005. Myocardial extra-cellular matrix and its regulation by metalloproteinases and their inhibitors. Thromb Haemost 93: 212–219.

Katti DS, Robinson KW, Ko FK, Laurencin CT. 2004. Bioresorbable nano!ber-based systems for wound healing and drug delivery: optimization of fabrication parameters. J Biomed Mater Sci 70B: 286–296.

Kenar H, Kocabas A, Aydinli A, Hasirci V. 2008. Chemical and topographical modi!cation of PHBV surface to promote osteoblast alignment and con!nement. J Biomed Mater Res A 85: 1001–1010.

Kenar H, Kose GT, Hasirci V. 2009. Design of a 3D aligned myocardial tissue construct from biode-gradable polyesters. J Mater Sci Mater Med 21(3): 989–997.

Kim HW, Song JH, Kim HE. 2005. Nano!ber generation of gelatin-hydroxyapatite biomimetics for guided tissue regeneration. Adv Funct Mater 15: 1988–1994.

Kim YT, Haftel VK, Kumar S, Bellamkonda RV. 2008. The role of aligned polymer !ber-based con-structs in the bridging of long peripheral nerve gaps. Biomaterials 29(21): 3117–3127.

Kinikoglu B, Auxenfans C, Pierrillas P, Justin V, Breton P, Burillon C, Hasirci V, Damour O. 2009. Reconstruction of a full-thickness collagen-based human oral mucosal equivalent. Biomaterials 30: 6418–6425.

Kneser U, Schaefer DJ, Munder B, Klemt C, Andree C, Stark GB. 2002. Tissue engineering of bone. Min Invas Ther Allied Technol 11: 107–116.

Ko!dis T, Akhyari P, Wachsmann B, Boublik J, Mueller-Stahl K, Leyh R, Fischer S, Haverich A. 2002. A novel bioarti!cial myocardial tissue and its prospective use in cardiac surgery. Eur J Cardiothorac Surg 22(2): 238–243.

Koh HS, Yong T, Chan CK, Ramakrishna S. 2008. Enhancement of neurite outgrowth using nano-structured scaffolds coupled with laminin. Biomaterials 29(26): 3574–3582.

Langer R, Vacanti JP. 1993. Tisue engineering. Science 260: 920–926.Lee CH, Shin HJ, Cho IH, Kang YM, Kim IA, Park KD, Shin JW. 2005. Nano!ber alignment and

direction of mechanical strain affect the ECM production of human ACL !broblast. Biomaterials 26(11): 1261–1270.

Lee JY, Bashur CA, Goldstein AS, Schmidt CE. 2009. Polypyrrole-coated electrospun PLGA nano!-bers for neural tissue applications. Biomaterials 30(26): 4325–4335.

K10254_C010.indd 221 5/12/11 7:18:09 PM

Page 28: Nanotechnology in Biomaterials: Nanofibers in Tissue ...biomed.metu.edu.tr/sites/biomat.metu.edu.tr/files/Yucel et al, 2011.pdf · membranes (wound dressings, drug delivery systems,

222 Bionanotechnology II: Global Prospects

Lee SJ, Yoo JJ, Lim GJ, Atala A, Stitzel J. 2007. In vitro evaluation of electrospun nano!ber scaffolds for vascular graft application. J Biomed Mater Res A 83(4): 999–1008.

Leong KF, Cheah CM, Chua CK. 2003. Solid free form fabrication of three dimensional scaffolds for engineering replacement tissues and organs. Biomaterials 24: 3262–3278.

Li M, Guo Y, Wei Y, MacDiarmid AG, Lelkes PI. 2006b. Electrospinning polyaniline-contained gelatin nano!bers for tissue engineering applications. Biomaterials 27: 2705–2715.

Li M, Mondrinos MJ, Chen X, Gandhi MR, Ko FK, Lelkes PI. 2006a. Co-electrospun poly(lactide-co-glycolide), gelatin, and elastin blends for tissue engineering scaffolds. J Biomed Mater Res A 79: 963–973.

Li WJ, Jiang YJ, Tuan RS. 2006. Chondrocyte phenotype in engineered !brous matrix is regulated by !ber size. Tissue Eng 12: 1775–1785.

Li WJ, Tuli R, Okafor C, Derfoul A, Danielson KG, Hall DJ, Tuan RS. 2005. A three dimensional nano!brous scaffold for cartilage tissue engineering using human mesenchymal stem cells. Biomaterials 26: 599–609.

Li X, Su Y, Liu S, Tan L, Mo X, Ramakrishna S. 2010. Encapsulation of proteins in poly(L-lactide-co-caprolactone) !bers by emulsion electrospinning. Colloids Surf B Biointerfaces 75(2): 418–424.

Liu H, Cai Q, Lian P, Fang Z, Duan S, Yang X, Deng X, Ryu S. 2010. -Tricalcium phosphate nanopar-ticles adhered carbon nano!brous membrane for human osteoblasts cell culture. Mater Lett 64: 725–728.

Liu X, Ma PX. 2004. Polymeric scaffolds for bone tissue engineering. Ann Biomed Eng 32(3): 477–486.Lu G, Sheng B, Wei Y, Wang G, Zhang L, Ao Q, Gong Y, Zhang X. 2008. Collagen nano!ber-covered

porous biodegradable carboxymethyl chitosan microcarriers for tissue engineering cartilage. Eur Polym J 44: 2820–2829.

Mak, Chan CK, Liuo S, Hwang WYK, Fery O, Ramakrishna S. 2008. Electrospan new !lter scaffolds for rapid and rich capture of bone marrow derived hematopoietic stem cells. Biomaterials. 29: 2096–2103.

Ma PX. 2004a. Scaffolds for tissue fabrication. Mater Today Rev 30–41.Ma PX. 2004b. Tissue engineering. In: Kroschwitz JI, ed., Encyclopedia of polymer science and technology,

3rd ed. John Wiley & Sons, Hobokan NJ.Ma PX, Langer R. 1999. In: Yarmush M, Morgan, J, eds., Tissue engineering methods and protocols.

Humana Press, Totowa, NJ, pp. 47–56.Ma PX, Zhang R, Xiao G, Franceschi R. 2001. Engineering new bone tissue in vitro on highly porous

poly( -hydroxyl acids)/hydroxyapatite composite scaffolds. J Biomed Mater Res 54: 284–293.Ma Z, Kotaki M, Inai R, Ramakrishna S. 2005. Potential of nano!ber matrix as tissue engineering

scaffolds. Tissue Eng 11: 101–109.Marra KG, Szem JW, Kumta PN, DiMilla PA, Weiss LE. 1999. In vitro analysis of biodegradable

polymer blend/hydroxyapatite composites for bone tissue engineering. J Biomed Mater Res 47: 324–335.

McDevitt TC, Woodhouse KA, Hauschka SD, Murry CE, Stayton PS. 2003. Spatially organized layers of cardiomyocytes on biodegradable polyurethane !lms for myocardial repair. J Biomed Mater Res A 66(3): 586–595.

Meechaisue C, Wutticharoenmongkol P, Waraput R, Huangjing T, Ketbumrung N, Pavasant P, Supaphol P. 2007. Preparation of electrospun silk !broin !ber mats as bone scaffolds: a prelimi-nary study. Biomed Mater 2: 181–188.

Meek MF, Varejao AS, Geuna S. 2002. Muscle grafts and alternatives for nerve repair. J Oral Maxillofac Surg 60: 1095–1096.

Mikos AG, Bao Y, Linda LG, Ingber DE, Vacanti JP, Langer R. 1993. Preparation of poly(glycolic acid) bonded !ber structures for cell attachment and transplantation. J Biomed Mater Res 27: 183–189.

Millesi H. 1991. Indications and techniques of nerve grafting. In: Gelbertman RH, ed., Operative nerve repair and reconstruction. Lippincott JB, Philadelphia, PA, pp. 525–544.

Mirensky TL, Breuer CK. 2008. The development of tissue-engineered grafts for reconstructive car-diothoracic surgical applications. Pediatr Res 63(5): 559–568.

K10254_C010.indd 222 5/12/11 7:18:09 PM

Page 29: Nanotechnology in Biomaterials: Nanofibers in Tissue ...biomed.metu.edu.tr/sites/biomat.metu.edu.tr/files/Yucel et al, 2011.pdf · membranes (wound dressings, drug delivery systems,

Nanotechnology in Biomaterials: Nanofibers in Tissue Engineering 223

Mooney DJ, Baldwin DF, Suh NP, Vacanti JP, Langer R. 1996. Novel approach to fabricate porous sponges of poly(D,L-lacto-co-glycolic acid) without the use of organic solvents. Biomaterials 17: 1417–1422.

Nakamura T, Schneider MD. 2003. The way to a human’s heart is through the stomach: visceral endo-derm-like cells drive human embryonic stem cells to a cardiac fate. Circulation 107: 2638–2639.

Ndreu A, Nikkola L, Ylikauppila H, Ashammakhi N, Hasirci V. 2008. Electrospun biodegradable nano!brous mats for tissue engineering. Nanomed 3: 45–60.

Neal RA, McClugage SG, Link MC, Sefcik LS, Ogle RC, Botchwey EA. 2009. Laminin nano!ber meshes that mimic morphological properties and bioactivity of basement membranes. Tissue Eng Part C Methods 15(1): 11–21.

Ngiam M, Liao S, Patil AJ, Cheng Z, Chan CK, Ramakrishna S. 2009. The fabrication of nano-hy-droxyapatite on PLGA and PLGA/collagen nano!brous composite scaffolds and their effects in osteoblastic behavior for bone tissue engineering. Bone 45: 4–16.

Nisbet DR, Forsythe JS, Shen W, Finkelstein DI, Horne MK. 2009. Review paper: a review of the cel-lular response on electrospun nano!bers for tissue engineering. J Biomater Appl 24: 7–29.

Nisbet DR, Pattanawong S, Ritchie NE, Shen W, Finkelstein DI, Horne MK, Forsythe JS. 2007. Interaction of embryonic cortical neurons on nano!brous scaffolds for neural tissue engineer-ing. J Neural Eng 4(2): 35–41.

Nisbet DR, Yu LM, Zahir T, Forsythe JS, Shoichet MS. 2008. Characterization of neural stem cells on electrospun poly(epsilon-caprolactone) submicron scaffolds: evaluating their potential in neu-ral tissue engineering. J Biomater Sci Polym Ed 19(5): 623–634.

Noh HK, Lee SW, Kim JM, Oh JE, Kim KH, Chung CP, Choi SC, Park WH, Min BM. 2006. Electrospinning of chitin nano!bers: degradation behavior and cellular response to normal human keratinocytes and !broblasts. Biomaterials 27: 3934–3944.

Noh K, Lee H, Shin U, Kim H. 2010. Composite nano!ber of bioactive glass nano!ller incorporated poly(lactic acid) for bone regeneration. Mater Lett 64: 802–805.

Ondarcuhu T, Joachim C. 1998. Drawing a single nano!ber over hundreds of microns. Europhys Lett 42: 215–220.

Palecek SP, Loftus JC, Ginsberg MH, Lauffenburger DA, Horwitz AF. 1997. Integrin-ligand bind-ing properties govern cell migration speed through cell-substratum adhesiveness. Nature 385: 537–540.

Panseri S, Cunha C, Lowery J, Del Carro U, Taraballi F, Amadio S, Vescovi A, Gelain F. 2008. Electrospun micro- and nano!ber tubes for functional nervous regeneration in sciatic nerve transections. BMC Biotechnol 11: 8–39.

Papadaki M, Bursac N, Langer R, Merok J, Vunjak-Novakovic G, Freed LE. 2001. Tissue engineer-ing of functional cardiac muscle: molecular, structural, and electrophysiological studies. Am J Physiol Heart Circ Physiol 280: H168–H178.

Park JB. 1981. Biomaterials Science and Engineering. Plenum Press New York, p1.Park J.B, Bronzino JD. 2003. Biomaterials: Principles and Applications, CRC Press, Boca Raton, FL, p1.Parker KK, Ingber DE. 2007. Extracellular matrix, mechanotransduction and structural hierarchies in

heart tissue engineering. Phil Trans R Soc B 362: 1267–1279.Patel S, Kurpinski K, Quigley R, Gao H, Hsiao BS, Poo MM, Li S. 2007. Bioactive nano!bers: synergis-

tic effects of nanotopography and chemical signaling on cell guidance. Nano Lett 7: 2122–2128.Petrigliano FA, McAllister DR, Wu BM. 2006. Tissue engineering for anterior cruciate ligament recon-

struction: a review of current strategies. Arthroscopy J Arthroscopic Related Surg 22: 441–451.Prabhakaran MP, Venugopal J, Ramakrishna S. 2009. Electrospun nanostructured scaffolds for bone

tissue engineering. Acta Biomater 5: 2884–2893.Prabhakaran MP, Venugopal JR, Chyan TT, Hai LB, Chan CK, Lim AY, Ramakrishna S. 2008.

Electrospun biocomposite nano!brous scaffolds for neural tissue engineering. Tissue Eng Part A 14(11): 1787–1797.

Radisic M, Park H, Shing H, Consi T, Schoen FJ, Langer R, Freed LE, Vunjak-Novakovic G. 2004. Functional assembly of engineered myocardium by electrical stimulation of cardiac myocytes cultured on scaffolds. Proc Natl Acad Sci USA 101(52): 18129–18134.

K10254_C010.indd 223 5/12/11 7:18:09 PM

Page 30: Nanotechnology in Biomaterials: Nanofibers in Tissue ...biomed.metu.edu.tr/sites/biomat.metu.edu.tr/files/Yucel et al, 2011.pdf · membranes (wound dressings, drug delivery systems,

224 Bionanotechnology II: Global Prospects

Reed CR, Han L, Andrady A, Caballero M, Jack MC, Collins JB, Saba SC, Loboa EG, Cairns BA, van Aalst JA. 2009. Composite tissue engineering on polycaprolactone nano!ber scaffolds. Ann Plast Surg 62: 505–512.

Ren L, Wang J, Yang FY, Wang L, Wang D, Wang TX, Tian MM. 2010. Fabrication of gelatin-siloxane !brous mats via sol-gel and electrospinning procedure and its application for bone tissue engi-neering. Mater Sci Eng C 30: 437–444.

Riboldi SA, Sampaolesi M, Neuenschwander P, Cossu G, Mantero S. 2005. Electrospun degrad-able polyesterurethane membranes: potential scaffolds for skeletal muscle tissue engineering. Biomaterials 26: 4606–4615.

Rockwood DN, Akins RE r, Parrag IC, Woodhouse KA, Rabolt JF. 2008. Culture on electrospun poly-urethane scaffolds decreases atrial natriuretic peptide expression by cardiomyocytes in vitro. Biomaterials 29: 4783–4791.

Sabir MI, Xu X, Li L. 2009. A review on biodegradable polymeric materials for bone tissue engineer-ing applications. J Mater Sci 44: 5713–5724.

Sahoo S, Ang LT, Goh JCH, Toh SL. 2009. Bioactive nano!bers for !broblastic differentiation of mesenchy-mal precursor cells for ligament/tendon tissue engineering applications. Differentiation 79: 102–110.

Sahoo S, Ouyang H, Goh JC, Tay TE, Toh SL. 2006. Characterization of a novel polymeric scaffold for potential application in tendon/ligament tissue engineering. Tissue Eng 12(1): 91–99.

Sanghvi AB, Archit B, Murray JL, Schmidt CE. 2004. Tissue engineering of peripheral nerve. Encyclopedia Biomater Biomed Eng 1(1): 1613–1621.

Sangsanoh P, Suwantong O, Neamnark A, Cheepsunthorn P, Pavasant P, Supaphol P. 2009. In vitro biocompatibility of electrospun and solvent-cast chitosan substrata towards Schwann, osteo-blast, keratinocyte and !broblast cells. Eur Polym J 46: 428–440.

Santos MI, Tuzlakoglu K, Fuchs S, Gomes ME, Peters KE, Unger RE, Piskin E, Reis RL, Kirkpatrick CJ. 2008. Endothelial cell colonization and angiogenic potential of combined nano- and micro-!brous scaffolds for bone tissue engineering. Biomaterials 29: 4306–4313.

Schmidt CE, Leach JB. 2003. Neural tissue engineering: strategies for repair and regeneration. Annu Rev Biomed Eng 5: 293–347.

Schmidt CE, Shastri VR, Vacanti JP, Langer R. 1997. Stimulation of neurite outgrowth using an elec-trically conducting polymer. Proc Natl Acad Sci USA 94(17): 8948–8953.

Schnell E, Klinkhammer K, Balzer S, Brook G, Klee D, Dalton P, Mey J. 2007. Guidance of glial cell migration and axonal growth on electrospun nano!bers of poly-epsilon-caprolactone and a collagen/poly-epsilon-caprolactone blend. Biomaterials 28(19): 3012–3025.

Seal BL, Otero TC, Panitch A. 2001. Polymeric biomaterials for tissue and organ regeneration. Mater Sci Eng 34(4–5): 147–230.

Shin M, Ishii O, Sueda T, Vacanti JP. 2004. Contractile cardiac grafts using a novel nano!brous mesh. Biomaterials 25: 3717–3723.

Silva GA, Czeisler C, Niece KL, Beniash E, Harrington DA, Kessler JA, Stupp SI. 2004. Selective dif-ferentiation of neural progenitor cells by high-epitope density nano!bers. Science 303(5662): 1352–1355.

Smith IO, Baumann MJ, McCabe LR. 2004. Electrostatic interactions as a predictor for osteoblast attachment to biomaterials. J Biomed Mater Res A 70: 436–441.

Soletti L, Hong Y, Guan J, Stankus JJ, El-Kurdi MS, Wagner WR, Vorp DA. 2010. A bilayered elas-tomeric scaffold for tissue engineering of small diameter vascular grafts. Acta Biomaterialia 6: 110–122.

Sombatmankhong K, Sanchavanakit N, Pavasant P, Supaphol P. 2007. Bone scaffolds from electro-spun !ber mats of poly(3-hydroxybutyrate), poly(3-hydroxybutyrate-co-3-hydroxyvalerate) and their blend. Polymer 48: 1419–1427.

Song JH, Kim HE, Kim HW. 2008. Electrospun !brous web of collagen–apatite precipitated nano-composite for bone regeneration. J Mater Sci Mater Med 19: 2925–2932.

Stankus JJ, Guan J, Fujimoto K, Wagner WR. 2006. Microintegrating smooth muscle cells into a bio-degradable, elastomeric !ber matrix. Biomaterials 27: 735–744.

K10254_C010.indd 224 5/12/11 7:18:09 PM

Page 31: Nanotechnology in Biomaterials: Nanofibers in Tissue ...biomed.metu.edu.tr/sites/biomat.metu.edu.tr/files/Yucel et al, 2011.pdf · membranes (wound dressings, drug delivery systems,

Nanotechnology in Biomaterials: Nanofibers in Tissue Engineering 225

Stevens B, Yang Y, Mohandas A, Stucker B, Nguyen KT. 2008. A review of materials, fabrication methods, and strategies used to enhance bone regeneration in engineered bone tissues. J Biomed Mater Res B Appl Biomater 85B: 573–582.

Sui G, Yang X, Mei F, Hu X, Chen G, Deng X, Ryu S. 2007. Poly-L-lactic acid/hydroxyapatite hybrid membrane for bone tissue regeneration. J Biomed Mater Res A 82(2): 445–454.

Teh TKH, Goh JCH, Toh SL. 2007. Advanced micro-nano !brous silk scaffold for tendon/ligament tissue engineering. J Biomech 40: 119–120.

Teh TKH, Goh JCH, Toh SL. 2008. Comparative study of random and aligned nano!brous scaffolds for tendon/ligament tissue engineering. J Biomech 41: 527.

Terzis JK, Sun DD, Thanos PK. 1997. History and basic science review: past, present, and future of nerve repair. J Reconstr Microsurg 13: 215–225.

Thomson RC, Yaszemski MJ, Powers JM, Mikes AG. 1995. Fabrication of biodegradable polymer scaffolds to engineer trabecular bone. J Biomater Sci 7: 23–38.

Thouas GA, Contreras KG, Bernard CC, Sun GZ, Tsang K, Zhou K, Nisbet DR, Forsythe JS. 2008. Biomaterials for spinal cord regeneration: outgrowth of presumptive neuronal precursors on electrospun poly(epsilon)-caprolactone scaffolds microlayered with alternating polyelectro-lytes. Conf Proc IEEE Eng Med Biol Soc 2008: 1825–1828.

Ulubayram K, Cakar AN, Korkusuz P, Ertan C, Hasirci N. 2001. EGF containing gelatin-based wound dressings. Biomaterials 22: 1345–1356.

Vargas EA, do Vale BNC, de Brito J, de Queiroz AA. 2010. Hyperbranched polyglycerol electrospun nano!bers for wound dressing applications. Acta Biomater 6: 1069–1078.

van de Graaff KM. 1998. Human anatomy. 5th ed. Weber State University, McGraw-Hill Companies.Vander AJ, Sherman JH, Luciano DS. 1994. Human physiology. 6th ed. McGraw-Hill Companies.Venugopal J, Ramakrishna S. 2005. Biocompatible nano!ber matrices for the engineering of a dermal

substitute for skin regeneration. Tissue Eng 11: 847–854.Vrana NE, Elsheikh A, Builles N, Damour O, Hasirci V. 2007. Effect of human corneal keratocytes and

retinal pigment epithelial cells on the mechanical properties of micropatterned collagen !lms. Biomaterials 28: 4303–4310.

Wang HB, Mullins ME, Cregg JM, Hurtado A, Oudega M, Trombley MT, Gilbert RJ. 2009. Creation of highly aligned electrospun poly-L-lactic acid !bers for nerve regeneration applications. Neural Eng 6(1): 016001.

Wang J, Shah A, Yu X. 2011. The in"uence of !ber thickness, wall thickness and gap distance on the spiral nano!brous scaffolds for bone tissue engineering. Mater Sci Eng C 34: 50–56.

Williams DF. 1987. De!nition in biomaterials. In: Progress in biomedical engineering. Elsevier, Amsterdam, p. 67.

Williams DF, Black J, Doherty PJ. 1992. Second consensus conference on de!nitions in biomaterials, Chester, England. In: Doherty PJ, Williams RF, Williams DF, Lee AJC, eds., Biomaterial–tissue interfaces. Advances in biomaterials, Vol. 10. Elsevier, Amsterdam.

Wu KH, Lu XD, Liu YL. 2006. Recent progress of pediatric cardiac surgery in China. Chin Med J (Engl) 119(23): 2005–2012.

Xie J, Macewan MR, Willerth SM, Li X, Moran DW, Sakiyama-Elbert SE, Xia Y. 2009a. Conductive core-sheath nano!bers and their potential application in neural tissue engineering. Adv Funct Mater 19(14): 2312–2318.

Xie J, Willerth SM, Li X, Macewan MR, Rader A, Sakiyama-Elbert SE, Xia Y. 2009b. The differentia-tion of embryonic stem cells seeded on electrospun nano!bers into neural lineages. Biomaterials 30(3): 354–362.

Xu CY, Inai R, Kotaki M, Ramakrishna S. 2004. Aligned biodegradable nano!brous structure: a poten-tial scaffold for blood vessel engineering. Biomaterials 25(5): 877–886.

Yang D, Jin Y, Zhou Y, Ma G, Chen X, Lu F, Nie J. 2008. In situ mineralization of hydroxyapatite on electrospun chitosan-based nano!brous scaffolds. Macromol Biosci 8: 239–246.

Yang F, Both SK, Yang X, Walboomers XF, Jansen JA. 2009. Development of an electrospun nano-apatite/PCL composite membrane for GTR/GBR application. Acta Biomater 5(9): 3295–3304.

K10254_C010.indd 225 5/12/11 7:18:10 PM

Page 32: Nanotechnology in Biomaterials: Nanofibers in Tissue ...biomed.metu.edu.tr/sites/biomat.metu.edu.tr/files/Yucel et al, 2011.pdf · membranes (wound dressings, drug delivery systems,

226 Bionanotechnology II: Global Prospects

Yang F, Murugan R, Wang S, Ramakrishna S. 2005. Electrospinning of nano/microscale poly(L-lactic acid) aligned !bers and their potential in neural tissue engineering. Biomaterials 26(15): 2603–2610.

Yang X, Shah JD, Wang H. 2009. Nano!ber enabled layer-by-layer approach toward three dimen-sional tissue formation. Tissue Eng A 15: 945–956.

Yilgor P, Sousa RA, Reis RL, Hasirci N, Hasrici V. 2008. 3D plotted PCL scaffolds for stem cell based bone tissue engineering. Macromol Symp 269: 92–99.

Yin Z, Chen X, Chen JL, Shen WL, Nguyen TMH, Gao L, Ouyang HW. 2010. The regulation of tendon stem cell differentiation by the alignment of nano!bers. Biomaterials 31: 2163–2175.

Yoshimoto H, Shin YM, Terai H, Vacanti JP. 2003. A biodegradable nano!ber scaffold by electrospin-ning and its potential for bone tissue engineering. Biomaterials 24: 2077–2082.

Yost MJ, Baicu CF, Stonerock CE, Goodwin RL, Price RL, Davis JM, Evans H, Watson PD, Gore CM, Sweet J, Creech L, Zile MR, Terracio L. 2004. A novel tubular scaffold for cardiovascular tissue engineering. Tissue Eng 10(1/2): 273–284.

Yucel D, Kose GT, Hasirci V. 2010. Polyester based nerve guidance conduit design. Biomaterials 31: 1596–1603.

Zhang R, Ma PX. 1999. Poly(alpha-hydroxy acids)/hydroxyapatite porous composites for bone tis-sue engineering. 1. Preparation and morphology. J Biomed Mater Res 44: 446–455.

Zhang Y, Lim CT, Ramakrishna S, Huang ZM. 2005. Recent development of polymer nano!bers for biomedical and biotechnological applications. J Mater Sci Mater Med 16: 933–946.

Zhang Y, Venugopal JR, El-Turki A, Ramakrishna S, Su B, Lim CT. 2008. Electrospun biomimetic nanocomposite nano!bers of hydroxyapatite/chitosan for bone tissue engineering. Biomaterials 29: 4314–4322.

Zhang Y, Ouyang H, Lim CT, Ramakrishna S, Huang ZM. 2005. Electrospinning of gelatin !bers and gelatin/PCL composite !brous scaffolds. J Biomed Mater Res Part B: Appl Biomater 72B: 156–165.

Zhao F, Yin YJ, Lu WW, Leong JC, Zhang WJ, Zhang JY, Zhang M. F, Yao KD. 2002. Preparation and histological evaluation of biomimetic three-dimensional hydroxyapatite/chitosan-gelatin net-work composite scaffolds. Biomaterials 23: 3227–3234.

Zimmermann WH, Eschenhagen T. 2003. Cardiac tissue engineering for replacement therapy. Heart Fail Rev 8: 259–269.

Zimmermann WH, Melnychenko I, Eschenhagen T. 2004. Engineered heart tissue for regeneration of diseased hearts. Biomaterials 25: 1639–1647.

Zimmermann WH, Schneiderbanger K, Schubert P, Didie M, Munzel F, Heubach JF, Kostin S, Neuhuber WL, Eschenhagen T. 2002. Tissue engineering of a differentiated cardiac muscle con-struct. Circ Res 90: 223–230.

Zong X, Bien H, Chung C-Y, Yin L, Fang D, Hsiao BS, Chu B, Entcheva E. 2005. Electrospun !ne-textured scaffolds for heart tissue constructs. Biomaterials 26: 5330–5338.

Zorlutuna P, Builles N, Damour O, Elsheikh A, Hasirci V. 2007. In"uence of keratocytes and reti-nal pigment epithelial cells on the mechanical properties of polyester-based tissue engineering micropatterned !lms. Biomaterials 28: 3489–3496.

Zorlutuna P, Rong Z, Vadgama P, Hasirci V. 2009. In"uence of nanopatterns on endothelial cell adhe-sion: enhanced cell retention under shear stress. Acta Biomater 5: 2451–2459.

Zorlutuna P, Tezcaner A, Hasirci V. 2008. A novel construct as a cell carrier for tissue engineering. J Biomater Sci Polym Ed 19: 399–410.

K10254_C010.indd 226 5/12/11 7:18:10 PM