1,307
Views
8
CrossRef citations to date
0
Altmetric
ORIGINAL ARTICLE

The potential of nanofibers in tissue engineering and stem cell therapy

, &
Pages 1195-1200 | Received 10 Feb 2015, Accepted 11 Mar 2015, Published online: 04 Jun 2015

Abstract

Electrospinning is a technique in which materials in solution are shaped into continuous nano- and micro-sized fibers. Combining stem cells with biomaterial scaffolds and nanofibers affords a favorable approach for bone tissue engineering, stem cell growth and transfer, ocular surface reconstruction, and treatment of congenital corneal diseases. This review seeks to describe the current examples of the use of scaffolds in stem cell therapy. Stem cells are classified as adult or embryonic stem (ES) cells, and the advantages and drawbacks of each group are detailed. The nanofibers and scaffolds are further classified in and , which describe specific examples from the literature. Finally, the current applications of biomaterial scaffolds containing stem cells for tissue engineering applications are presented. Overall, this review seeks to give an overview of the biomaterials available for use in combination with stem cells, and the application of nanofibers in stem cell therapy.

Introduction

Electrospinning, which is a technology that enables the manufacture of an ultrafine fiber, was initiated in the 1990 s. From the time the term electrostatic spinning was first used, it has gained popular attention in both the industrial and academic communities (CitationReneker and Chun 1996, CitationAnton 1934, CitationHuang et al. 2003, CitationLi and Xia 2004). Electrospinning is a technique that enables the fabrication of fibers with diameters in the nanometer range, which produces very large specific surface area, greater than that of the current microfibers produced from conventional dry/wet and melt spinning techniques. Consequently, electrospun nanofibers are very suitable for improving a diversity of structures or products whose purposes are reliant on surface area. Among the promising applications, one of the most potential uses is in increasing nanofibrous cellular scaffolds and nanofibers for tissue engineering and stem cell applications. Up to the present time, electrospun fibers have been used in a wide range of regenerative medicine and stem cell applications.

Stem cells

Adult stem cells, including bone marrow mesenchymal stem cells (MSCs), have been proved for their multipotency in leading to the formation of bone, cartilage, connective tissue, fat or muscle. Defined inductive media can be commonly used for the accurate control of the differentiation lineage in vitro. Scientists have used autologous MSCs in conventional tissue engineering ex vivo, in combination with biomaterials, to generate a cell-based scaffold with the aim of replacing or improving tissue regeneration in vivo. Furthermore, through an incompletely understood mechanism, endogenous MSCs can be located in cell-free implanted scaffolds, which is known as homing. Homing of MSCs contributes to the renewal of the wounded tissue and demonstrates their critical role in the reformative process.

On other hand, embryonic stem (ES) cells are pluripotent cells and they have the potential for constant self-renewal (CitationHan et al. 2012).

Nanofibers and scaffolds

Nanofibers have diameters ranging from tens of nanometers to microns when prepared by electrostatic spinning or electrospinning technologies which enable the production of nonwoven and ultrafine nanoscale nanofibers. This technology was established for the first time in the 1990s by Reneker and coworkers.

There are several methods to produce nanofibers, such as self-assembly (CitationWilliamson and Coombes 2004, CitationNaik et al. 2003) template synthesis (CitationReneker and Chun 1996, CitationDi et al. 2008, CitationDoshi and Reneker 1993, CitationLiang et al. 2007), phase separation (CitationMa et al. 2005a, CitationWidmer et al. 1998), drawing (CitationZhong et al. 2011, CitationGuarino et al. 2014), and electrospinning (CitationBadami et al. 2006, CitationBhattarai et al. 2005).

Phase separation is one of best methods for the production of nanoporous foams and is preferred for use in many areas; however, as this method needs a long time for the completion of the process, it is not the best (CitationEatemadi et al. 2014a, CitationEatemadi et al. 2014b, CitationDaraee et al. 2014a, CitationDaraee et al. 2014b). By using this process and by modifying thermodynamic and kinetic factors, it is easy to achieve the production and fabrication of various nanoporous foams and foam scaffolds. Using the phase separation process, this can be achieved in five basic steps—suspension of polymer, phase separation and gelation, extraction of solvent from the gel by means of water, freezing, and then freeze-drying under vacuum (CitationMa et al. 2005b).

In the self-assembly method, atoms and molecules reform and assemble themselves due to noncovalent and fragile forces, for example electrostatic interactions, hydrophobic forces, and hydrogen bonding, and finally produce a stable construction (CitationHartgerink et al. 2001, CitationZhang 2003). Different structures can be produced by the self-assembly method, such as bilayer, unilamellar and multilamellar vesicles, membranes, nanoparticles, films, fibers, tubes, capsules, and micelles (CitationVenugopal and Ramakrishna 2005). In comparison with the electrospinning method, fibers synthesized by self-assembly are much thinner, but the complexity of the procedure as well as the low productivity are the main limitations of this method (CitationMa et al. 2005a).

The electrospinning method has unique properties such as affordability, very high surface-to-volume ratio, availability of a wide range of materials that can be selected, flexibility of adoption for a broad range of sizes and shapes, simplicity, and tunable porosity (CitationMellatyar et al. 2014, CitationSeidi et al. 2014).

In this process, several biomaterials can be used to fabricate nanofibers, and it requires very low amounts of initial solutions. Fabrication of fibers using the electrospinning method achieves fibers ranging from 3 nm to several micrometers in diameter, but fibers obtained by other methods have diameters in the range of 500 nm to a few microns (CitationZhang 2003).

Generally, owing to these tremendous properties that offer considerable advantages, the electrospinning method is the most popular method for the fabrication of nanofibers. As nanofibers are very tiny in diameter (micrometers to submicrons or nanometers), they possess numerous remarkable characteristics such as flexibility in surface functionalities, superior mechanical performance (for example, stiffness and tensile strength), and very large surface area to volume ratio compared with any other known form of the material.

As discussed above, these important properties make polymer nanofibers potential and optimal candidates for several major applications.

As nanofibrous materials have considerable potential for a variety of applications and offer some benefits of nanostructured materials, they are being well studied and developed. Nanofibrous materials can be synthesized from biocompatible and biodegradable polymers that are natural or synthetic, and also from composites of the two. These biodegradable polymers have been used to fabricate nanofibers for several purposes, based on the applications. Some polymers have been produced for short-time usage, such as cell carriers, delivery agents, and scaffolds (until new tissue becomes mature and independent). In these cases, the polymer will be substituted by original tissues. Other polymers have been used for long-term usage, such as for implants in surgery (CitationHohman et al. 2001). Nanofibers have attracted considerable interest in biotechnology and medicine because of the high potential of using biomaterials in different applications ().

Table I. Example of nanofiber applications in tissue engineering.

Electrospun polymeric nanofibers and scaffolds have been fabricated using copolymers, homopolymers, polymeric blends, and organic-inorganic hybrid materials. Among accessible biopolymers, the most extensively studied nanofiber systems for the regeneration of tissues and in stem cell applications which have been studied are poly-L-lactide (PLA), poly-glycolide (PGA), poly (e-caprolactone) (PCL), and their copolymers (CitationBurg et al. 2000). Nanofibers can be used globally in stem cell applications and they have significant potential in the field of stem cell research and therapy.

Bone tissue engineering by stem cells

Numerous kinds of cells are used for nanofiber and scaffold seeding in this field, comprising bone cells and stem cells. The use of stem cells embodies a current approach in the field of bone tissue engineering, and the revolution is embodied by the assessment of proliferation, cell adhesion, and mostly differentiation on the scaffold and nanofiber, as well as motivating cell differentiation in the direction of the desired phenotype. The mesenchymal stem cells (MSCs) are the central cell lines used for bone tissue engineering applications. Adult bone marrow comprises MSCs that contribute to the regeneration of mesenchymal tissues, such as bone, muscle, cartilage, tendon, ligament, and stromal cells from adipose tissue. Actually, under suitable stimulation given by the scaffold, MSCs experience osteogenic differentiation over a definite pathway, as well as acquire osteoblastic indicators and secrete calcium crystals and extracellular matrix (CitationYoshimoto et al. 2003).

Scaffolds for stem cell growth and transfer

One of the main problems related to stem cell therapy remains the deficiency of an appropriate carrier to transfer stem cells to the exact location in tissue. Up to now, numerous materials and scaffolds have been studied for the transfer of stem cells, such as self-assembling peptide nanofibers studied for cell-based therapy on infarcted myocardium (CitationDubois et al. 2008), macroporous hydrogels used to transfer SCs for spinal cord injury repair (CitationSyková et al. 2006), polymers and collagen sponges, (CitationRama et al. 2001) and fibrin glue used for cell transfer onto the ocular surface (CitationSchwab et al. 2006). Also, stem cells for therapeutic application on the ocular surface cannot be directly injected to the damaged eye as a cell suspension. To permanently anchor transferred cells, numerous carriers have been suggested and studied. Up to now, human amniotic membrane symbolizes the gold standard. In the majority of cases studied, the amniotic membrane was applied as a cell carrier for the transfer and growth of stem cells (CitationNakamura et al. 2004, CitationJiang et al. 2010, CitationShimazaki et al. 2002). Nevertheless, amniotic membrane is a biomaterial of human origin, with initial challenges in regulating its preparation, and therefore an exploration for substitute cell carriers and modes of stem cell transplantation on the ocular surface continues. In , some of the electrospun polymeric scaffolds for bone tissue engineering based on stem cells have been summarized.

Table II. Electrospun polymeric scaffolds for bone tissue engineering.

The use of MSCs for ocular surface reconstruction

Ocular surface reconstruction, challenged by the low number of available cells and the frequent absence of autologous limbal stem cells (LSC), led to attempts to use MSCs developed in vitro to substitute LSCs for corneal reconstruction and regeneration. These attempts are based on the potential of MSCs to differentiate into other cell types, as well as their immunomodulatory properties which can be valuable for the assessments of the local inflammatory response after transplantation of stem cells.

For the first time, (CitationMa et al. 2006) showed that MSCs derived from human bone marrow and transplanted by means of an amniotic membrane onto chemically burned rat ocular surface supported corneal healing and were involved in the reconstruction of the damaged cornea. Their healing effects contributed to the inhibition of angiogenesis and inflammation.

The helpful application of MSCs was approved in another study using a model of chemically burned rat ocular surface and the subconjunctival administration of rat MSCs (CitationYao et al. 2012), the application of MSCs with a special hollow plastic tube (CitationOh et al. 2008), or the transplantation of rat MSCs onto the ocular surface using amniotic membrane (CitationJiang et al. 2010). In another study, scientists (CitationMartínez-Conesa et al. 2012) injected a suspension of human MSCs into a rabbit model of corneal stromal defect and observed the restoration of the stromal organization, the maintenance of transparency, and the acquisition of vision. These studies were developed by a model of LSC deficiency in a murine model of a mechanically damaged ocular surface and transplantation of syngeneic MSCs seeded on nanofiber scaffolds (CitationZajicova et al. 2010), or in rabbits injected with autologous MSCs under an amniotic membrane (CitationReinshagen et al. 2011). All of these experimental studies have shown that MSCs significantly attenuate inflammation, increase corneal wound healing, and reduce corneal neovascularization.

The use of MSCs to treat congenital corneal diseases

CitationLiu et al. (2010) launched an experimental model to study the potential of overcoming the lack of corneas for keratoplasty to heal congenital corneal diseases of genetic mutation and heal the cloudy and thin corneas of lumican-null mice with human umbilical MSCs. Corneal stromal thickness and corneal transparency were significantly improved by transplanted MSCs. This study suggests that MSCs may also be a competent key for the cure of congenital corneal diseases, including keratocyte dysfunction.

Conclusion

Electrospinning has gained a reputation within the tissue engineering and stem cell research community as a promising means of fabricating scaffolds and nanofibers. This technology of constructing nanofibers enables the assessment of fiber diameter, geometry, fiber alignment, biomaterial composition, and drug/protein combination in a scaffold. Nanoscale fibers manufactured using electrospinning technology are competent to develop the cellular interactions of a wide-ranging diversity of cell types; furthermore, the cells are capable of conserving their functional and phenotypic characteristics on nanofibrous scaffolds. Generally, these electrospun scaffolds and nanofibers can be functionalized by anchoring mechanical and biochemical signals, to improve cellular interactions for stem cell therapy and tissue engineering applications.

Authors’ contributions

SG conceived of the study and participated in its design and coordination. AA participated in the sequence alignment and drafted the manuscript. All authors read and approved the final manuscript.

Acknowledgments

The authors thank the Department of Molecular Medicine, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, for all support provided. This work is funded by a 2015 grant by the Drug Applied Research Center, Tabriz University of Medical Sciences.

Declaration of interest

The authors have no declaration of interest. The authors alone are responsible for the content and writing of the paper.

References

  • Anton F.Process and apparatus for preparing artificial threads. 1934. Google Patents.
  • Badami AS, Kreke MR, Thompson MS, Riffle JS, Goldstein AS. 2006. Effect of fiber diameter on spreading, proliferation, and differentiation of osteoblastic cells on electrospun poly (lactic acid) substrates. Biomaterials. 27:596–606.
  • Bhattarai N, Edmondson D, Veiseh O, Matsen FA, Zhang M. 2005. Electrospun chitosan-based nanofibers and their cellular compatibility. Biomaterials. 26:6176–6184.
  • Binulal N, Deepthy M, Selvamurugan N, Shalumon K, Suja S, Mony U, et al. 2010. Role of nanofibrous poly (caprolactone) scaffolds in human mesenchymal stem cell attachment and spreading for in vitro bone tissue engineering—response to osteogenic regulators. Tissue Eng A. 16:393–404.
  • Burg KJ, Porter S, Kellam JF. 2000. Biomaterial developments for bone tissue engineering. Biomaterials. 21:2347–2359.
  • Daraee H, Eatemadi A, Abbasi E, Fekri Aval S, Kouhi M, Akbarzadeh A. 2014. Application of gold nanoparticles in biomedical and drug delivery. Artif Cells Nanomed Biotechnol. 1–13.
  • Daraee H, Etemadi A, Kouhi M, Alimirzalu S, Akbarzadeh A. 2014. Application of liposomes in medicine and drug delivery. Artif Cells Nanomed Biotechnol. 1–11.
  • Di J, Chen H, Wang X, Zhao Y, Jiang L, Yu J, et al. 2008. Fabrication of zeolite hollow fibers by coaxial electrospinning. Chem Mater. 20:3543–3545.
  • Doshi J, Reneker DH. (Eds.). 1993. Electrospinning process and applications of electrospun fibers. Industry Applications Society Annual Meeting. Conference Record of the 1993 IEEE; 1993: IEEE.
  • Dubois G, Segers VF, Bellamy V, Sabbah L, Peyrard S, Bruneval P, et al. 2008. Self‐assembling peptide nanofibers and skeletal myoblast transplantation in infarcted myocardium. J Biomed Mater Res B Appl Biomater. 87:222–228.
  • Eatemadi A, Daraee H, Karimkhanloo H, Kouhi M, Zarghami N, Akbarzadeh A, et al. 2014. Carbon nanotubes: properties, synthesis, purification, and medical applications. Nanoscale Res Lett. 9:1–13.
  • Eatemadi A, Daraee H, Zarghami N, Melat Yar H, Akbarzadeh A. 2014. Nanofiber: synthesis and biomedical applications. Artif Cells Nanomed Biotechnol. 1–11. [Epub ahead of print].
  • Ekaputra AK, Zhou Y, Cool SM, Hutmacher DW. 2009. Composite electrospun scaffolds for engineering tubular bone grafts. Tissue Eng A. 15:3779–3788.
  • Fujihara K, Kotaki M, Ramakrishna S. 2005. Guided bone regeneration membrane made of polycaprolactone/calcium carbonate composite nano-fibers. Biomaterials. 26:4139–4147.
  • Guarino V, Cirillo V, Altobelli R, Ambrosio L. 2014. Polymer-based platforms by electric field-assisted techniques for tissue engineering and cancer therapy. Expert Rev Med Devices. 12:113–129.
  • Han Y, Tao R, Sun T, Chai J, Xu G, Liu J. 2012. Advances and opportunities for stem cell research in skin tissue engineering. Eur Rev Med Pharmacol Sci. 16:1873–1877.
  • Hartgerink JD, Beniash E, Stupp SI. 2001. Self-assembly and mineralization of peptide-amphiphile nanofibers. Science. 294:1684–1688.
  • He W, Yong T, Teo WE, Ma Z, Ramakrishna S. 2005. Fabrication and endothelialization of collagen-blended biodegradable polymer nanofibers: potential vascular graft for blood vessel tissue engineering. Tissue Eng. 11:1574–1588.
  • Hohman MM, Shin M, Rutledge G, Brenner MP. 2001. Electrospinning and electrically forced jets. I. Stability theory. Phys Fluids. 13:2201–2220.
  • Huang ZM, Zhang YZ, Kotaki M, Ramakrishna S. 2003. A review on polymer nanofibers by electrospinning and their applications in nanocomposites. Comp Sci Technol. 63:2223–2253.
  • Jiang TS, Cai L, Ji WY, Hui YN, Wang YS, Hu D, Zhu J. 2010. Reconstruction of the corneal epithelium with induced marrow mesenchymal stem cells in rats. Mol Vis. 16:1304.
  • Jin HJ, Chen J, Karageorgiou V, Altman GH, Kaplan DL. 2004. Human bone marrow stromal cell responses on electrospun silk fibroin mats. Biomaterials. 25:1039–1047.
  • Ko EK, Jeong SI, Rim NG, Lee YM, Shin H, Lee BK. 2008. In vitro osteogenic differentiation of human mesenchymal stem cells and in vivo bone formation in composite nanofiber meshes. Tissue Eng A. 14:2105–2119.
  • Lee CH, Shin HJ, Cho IH, Kang YM, Kim IA, Park KD, et al. 2005. Nanofiber alignment and direction of mechanical strain affect the ECM production of human ACL fibroblast. Biomaterials. 26:1261–1270.
  • Li C, Vepari C, Jin HJ, Kim HJ, Kaplan DL. 2006. Electrospun silk-BMP-2 scaffolds for bone tissue engineering. Biomaterials. 27:3115–3124.
  • Li D, Xia Y. 2004. Electrospinning of nanofibers: reinventing the wheel? Advan Mater. 16:1151–1170.
  • Li M, Mondrinos MJ, Gandhi MR, Ko FK, Weiss AS, Lelkes PI. 2005. Electrospun protein fibers as matrices for tissue engineering. Biomaterials. 26:5999–6008.
  • Li WJ, Danielson KG, Alexander PG, Tuan RS. 2003. Biological response of chondrocytes cultured in three‐dimensional nanofibrous poly (ε‐caprolactone) scaffolds. J Biomed Mater Res A. 67:1105–1114.
  • Li WJ, Laurencin CT, Caterson EJ, Tuan RS, Ko FK. 2002. Electrospun nanofibrous structure: a novel scaffold for tissue engineering. J Biomed Mater Res. 60:613–621.
  • Liang D, Hsiao BS, Chu B. 2007. Functional electrospun nanofibrous scaffolds for biomedical applications. Advanc Drug Deliv Rev. 59:1392–1412.
  • Liu H, Zhang J, Liu CY, Wang IJ, Sieber M, Chang J, et al. 2010. Cell therapy of congenital corneal diseases with umbilical mesenchymal stem cells: lumican null mice. PloS One. 5:e10707.
  • Ma Y, Xu Y, Xiao Z, Yang W, Zhang C, Song E, et al. 2006. Reconstruction of chemically burned rat corneal surface by bone marrow–derived human mesenchymal stem cells. Stem Cells. 24:315–321.
  • Ma Z, He W, Yong T, Ramakrishna S. 2005a. Grafting of gelatin on electrospun poly (caprolactone) nanofibers to improve endothelial cell spreading and proliferation and to control cell orientation. Tissue Eng. 11:1149–1158.
  • Ma Z, Kotaki M, Inai R, Ramakrishna S. 2005b. Potential of nanofiber matrix as tissue-engineering scaffolds. Tissue Eng. 11:101–109.
  • Martínez-Conesa EM, Espel E, Reina M, Casaroli-Marano RP. 2012. Characterization of ocular surface epithelial and progenitor cell markers in human adipose stromal cells derived from lipoaspirates. Invest Ophthalmol Vis Sci. 53:513–520.
  • Meinel AJ, Kubow KE, Klotzsch E, Garcia-Fuentes M, Smith ML, Vogel V, et al. 2009. Optimization strategies for electrospun silk fibroin tissue engineering scaffolds. Biomaterials. 30:3058–3067.
  • Mellatyar H, Akbarzadeh A, Rahmati M, Ghalhar MG, Etemadi A, Nejati-Koshki K, et al. 2014. Comparison of inhibitory effect of 17-DMAG nanoparticles and free 17-DMAG in HSP90 gene expression in lung cancer. Asian Pac J Cancer Prev. 15:8693–8698.
  • Mo X, Xu C, Kotaki M, Ramakrishna S. 2004. Electrospun P (LLA-CL) nanofiber: a biomimetic extracellular matrix for smooth muscle cell and endothelial cell proliferation. Biomaterials. 25:1883–1890.
  • Naik S, Chiang A, Thompson R, Huang F. 2003. Formation of silicalite-1 hollow spheres by the self-assembly of nanocrystals. Chem Mater. 15:787–792.
  • Nakamura T, Inatomi T, Sotozono C, Amemiya T, Kanamura N, Kinoshita S. 2004. Transplantation of cultivated autologous oral mucosal epithelial cells in patients with severe ocular surface disorders. Br J Ophthalmol. 88:1280–1284.
  • Oh JY, Kim MK, Shin MS, Lee HJ, Ko JH, Wee WR, et al. 2008. The anti‐inflammatory and anti‐angiogenic role of mesenchymal stem cells in corneal wound healing following chemical injury. Stem Cells. 26:1047–1055.
  • Rama P, Bonini S, Lambiase A, Golisano O, Paterna P, De Luca M, et al. 2001. Autologous fibrin-cultured limbal stem cells permanently restore the corneal surface of patients with total limbal stem cell deficiency1. Transplantation. 72:1478–1485.
  • Reinshagen H, Auw‐Haedrich C, Sorg RV, Boehringer D, Eberwein P, Schwartzkopff J, et al. 2011. Corneal surface reconstruction using adult mesenchymal stem cells in experimental limbal stem cell deficiency in rabbits. Acta Ophthalmol. 89:741–748.
  • Ren L, Wang J, Yang FY, Wang L, Wang D, Wang TX, et al. 2010. Fabrication of gelatin–siloxane fibrous mats via sol–gel and electrospinning procedure and its application for bone tissue engineering. Mater Sci Eng C. 30:437–444.
  • Reneker DH, Chun I. 1996. Nanometre diameter fibres of polymer, produced by electrospinning. Nanotechnology. 7:216.
  • Riboldi SA, Sampaolesi M, Neuenschwander P, Cossu G, Mantero S. 2005. Electrospun degradable polyesterurethane membranes: potential scaffolds for skeletal muscle tissue engineering. Biomaterials. 26:4606–4615.
  • Ruckh TT, Kumar K, Kipper MJ, Popat KC. 2010. Osteogenic differentiation of bone marrow stromal cells on poly (ε-caprolactone) nanofiber scaffolds. Acta Biomater. 6:2949–2959.
  • Sahoo S, Ouyang H, Goh JCH, Tay T, Toh S. 2006. Characterization of a novel polymeric scaffold for potential application in tendon/ligament tissue engineering. Tissue Eng. 12:91–99.
  • Schwab IR, Johnson NT, Harkin DG. 2006. Inherent risks associated with manufacture of bioengineered ocular surface tissue. Arch Ophthalmol. 124:1734–1740.
  • Seidi K, Eatemadi A, Mansoori B, Jahanban-Esfahlan R, Farajzadeh D. 2014. Nanomagnet-based detoxifying machine: an alternative/complementary approach in HIV therapy. J AIDS Clin Res. 5:2.
  • Shields KJ, Beckman MJ, Bowlin GL, Wayne JS. 2004. Mechanical properties and cellular proliferation of electrospun collagen type II. Tissue Eng. 10:1510–1517.
  • Shih YRV, Chen CN, Tsai SW, Wang YJ, Lee OK. 2006. Growth of mesenchymal stem cells on electrospun type I collagen nanofibers. Stem Cells. 24:2391–2397.
  • Shimazaki J, Aiba M, Goto E, Kato N, Shimmura S, Tsubota K. 2002. Transplantation of human limbal epithelium cultivated on amniotic membrane for the treatment of severe ocular surface disorders. Ophthalmology. 109:1285–1290.
  • Shin M, Yoshimoto H, Vacanti JP. 2004. In vivo bone tissue engineering using mesenchymal stem cells on a novel electrospun nanofibrous scaffold. Tissue Eng. 10:33–41.
  • Son WK, Youk JH, Park WH. 2004. Preparation of ultrafine oxidized cellulose mats via electrospinning. Biomacromolecules. 5: 197–201.
  • Stitzel J, Liu J, Lee SJ, Komura M, Berry J, Soker S, et al. 2006. Controlled fabrication of a biological vascular substitute. Biomaterials. 27:1088–1094.
  • Syková E, Jendelová P, Urdzíková L, Lesný P, Hejčl A. 2006. Bone marrow stem cells and polymer hydrogels—two strategies for spinal cord injury repair. Cell Mol Neurobiol. 26:1111–1127.
  • Tuzlakoglu K, Bolgen N, Salgado A, Gomes ME, Piskin E, Reis R. 2005. Nano-and micro-fiber combined scaffolds: a new architecture for bone tissue engineering. J Mater Sci Mater Med. 16:1099–1104.
  • Venugopal J, Ramakrishna S. 2005. Biocompatible nanofiber matrices for the engineering of a dermal substitute for skin regeneration. Tissue Eng. 11:847–854.
  • Widmer MS, Gupta PK, Lu L, Meszlenyi RK, Evans GR, Brandt K, et al. 1998. Manufacture of porous biodegradable polymer conduits by an extrusion process for guided tissue regeneration. Biomaterials. 19:1945–1955.
  • Williamson MR, Coombes AG. 2004. Gravity spinning of polycaprolactone fibres for applications in tissue engineering. Biomaterials. 25:459–465.
  • Xu C, Inai R, Kotaki M, Ramakrishna S. 2004. Aligned biodegradable nanofibrous structure: a potential scaffold for blood vessel engineering. Biomaterials. 25:877–886.
  • Yang Y, Nikkola L, Ylikauppila H, Ashammakhi N. (Eds.). 2006. Investigation of cell attachment on the scaffolds manufactured by electrospun PCL-hyaluronan blend. TN.T. “Trends in Nanotechnology” Conference; 2006.
  • Yao L, Li ZR, Su WR, Li YP, Lin ML, Zhang WX, et al. 2012. Role of mesenchymal stem cells on cornea wound healing induced by acute alkali burn. PLoS One. 7:e30842.
  • Yoshimoto H, Shin Y, Terai H, Vacanti J. 2003. A biodegradable nanofiber scaffold by electrospinning and its potential for bone tissue engineering. Biomaterials. 24:2077–2082.
  • Zajicova A, Pokorna K, Lencova A, Krulova M, Svobodova E, Kubinova S, et al. 2010. Treatment of ocular surface injuries by limbal and mesenchymal stem cells growing on nanofiber scaffolds. Cell Transplant. 19:1281–1290.
  • Zhang H, Chen Z. 2010. Fabrication and characterization of electrospun PLGA/MWNTs/hydroxyapatite biocomposite scaffolds for bone tissue engineering. J Bioact Comp Poly. 25:241–259.
  • Zhang S. 2003. Fabrication of novel biomaterials through molecular self-assembly. Nat Biotechnol. 21:1171–1178.
  • Zhong S, Zhang Y, Lim CT. 2011. Fabrication of large pores in electrospun nanofibrous scaffolds for cellular infiltration: a review. Tissue Eng B Rev. 18:77–87.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.