1,377
Views
13
CrossRef citations to date
0
Altmetric
Reviews

New state of nanofibers in regenerative medicine

, , &
Pages 204-210 | Received 24 Jan 2016, Accepted 15 Mar 2016, Published online: 13 Apr 2016

Abstract

Regenerative medicine has discovered engineered nanofiber scaffolds enhancing regeneration process. These agents have an attractive property to mimic the native environment. They are excellent agents in binding the extracellular matrix of a cell to another cell. They help in the growth and multiplication of the cell and help in the differentiation of the cells which are required before the regeneration process. Regenerative medicine focuses on cellular therapies, origins of stem and progenitor cells, and on explaining how they persevere (or do not) in adult organisms and improvement of biomaterials. The focus of this review is on the application of nanofiber scaffolds.

Introduction: regenerative medicine

The field of regenerative medicine and tissue engineering has exploded in the last decade (Fisher and Mauck Citation2013). The approaches of tissue engineering and regenerative medicine are theoretically simple and attractive, yet these have proven to be challenging engineering tasks (Deluzio et al. Citation2013). In spite of rapid improvements made in this field (Fisher and Mauck Citation2013, Laflamme and Murry Citation2011), success is still limited because of major knowledge gaps in our ability to coordinate, control, and direct tissue formation, which are the ultimate goals for both tissue engineering and regenerative medicine (Deluzio et al. Citation2013).

In regenerative medicine approaches, cellular therapies remain a large focus, and the effectiveness of numerous cell types have been assessed in vivo. Furthermore, huge efforts in regenerative medicine and biology have focused on the origins of stem and progenitor cells, and on explaining how they persevere (or do not) in adult organisms. Another main focus has been on the improvement of biomaterials to either provide a scaffold that can promote appropriate tissue formation and/or release bioactive factors to aid in the healing response (Fisher and Mauck Citation2013).

Nanofibers and electrospinning

Using electrospinning or electrostatic spinning methods, nanofibrous scaffolds can be fabricated of biodegradable and biocompatible polymers (), which were reviewed smartly by Ravichandran et al. (Citation2012a).Various biodegradable polymers have been used to produce nanofiber scaffolds such as synthetic, natural, and composite of the both (Ravichandran et al. Citation2012a).

Table 1. Comparison of various nanofiber scaffold processing methods.

Natural polymer applied to produce nanofiber scaffolds have completed progressively more research interests, including oxidized cellulose (Son et al. Citation2004), collagen (Venugopal et al. Citation2005), hyaluronic acid (Um et al. Citation2004), elastin (Boland et al. Citation2004), fibrin (Jukola et al. Citation2008), silk protein (Jin et al. Citation2004), elastin-mimetic peptide (Huang et al. Citation2005), tropoelastin (Li et al. Citation2005), and fibrinogen (Sindelar et al. Citation2006).

The most important advantage of natural polymer is its identity and similarity to some molecular biomaterials that exist in the human body. However, the most important disadvantage of natural polymer is its reduced mechanical properties when isolated, thus this natural polymer has a need of further processing for handling. So, synthetic polymer was introduced.

Different synthetic polymers for synthesis of scaffolds have been used such as PCL (Khil et al. Citation2005, Luong-Van et al. Citation2006), PLGA (Badami et al. Citation2006, Chew et al. Citation2005), PLDLA (Cui et al. Citation2006), PLLA (Badami et al. Citation2006), and copolymers for example PCL-PLLA (Nikkola et al. Citation2005), PCL-PEG, PLGA-PEG (Daraee et al. Citation2016, Mellatyar et al. Citation2014), PLLA-PEG (Mellatyar et al. Citation2014), etc.

Blends of natural polymer and synthetic polymer have been used to encompass characteristics of both. Studies merging polymer involved gelatin-loaded PCL (Ma et al. Citation2005), composites of PLLA-PCL and collagen (He et al. Citation2005), collagen-loaded PLLA-PCL (Huang et al. Citation2003), composites of hyaluronic acid and PCL (Yang et al. Citation2006), blends of PEO and silk (Li et al. Citation2002), composites of PLGA, elastin and collagen (Stitzel et al. Citation2000), composites of PCL and starch (Yang et al. Citation2006), and composites of PLGA with PHBV (Zhu et al. Citation2009).

Nanofibers in regenerative medicine

Because of the significant potential of applying biomaterials in wide spectrum applications, field of nanofibers has achieved extensive interest in biotechnology, tissue engineering, and medicine.

Nanofibers have been extensively used in regenerative medicine and nano-biotechnology. summarizes several examples of nanofiber application in regenerative medicine.

Table 2. Example of nanofiber application in regenerative medicine.

Skin regeneration

Skin regeneration and repair are based on two important approaches: skin drug delivery to interested sites and ability of the cells surrounding the damaged tissue for regeneration and repair. Cells of these two classes have the potential to regenerate into molecular structures that are similar to the original tissues. The importance of nanofiber scaffolds for skin regeneration and repair and tissue-engineering applications has been greatly studied. Nanofiber scaffolds which have a larger surface to area and nano-scale molecular architectures adsorb efficiently proteins and skin drugs, presenting many binding sites to cell membrane receptors which would be more biomimetic to better support cell–matrix interactions (Fujihara et al. Citation2005, Venugopal et al. Citation2005, Venugopal et al. Citation2008, Yang et al. Citation2004). Electrospun fibers perform as engineered skin substitutes that are extensively studied as bioactive dressings (Guadalupe et al. Citation2015).

Wnek et al. engineered a nanofiber scaffold using fibrinogen for skin repair, hemostatic products, and wound dressing (Wnek et al. Citation2003). Blends of gelatin and chitosan nanofiber have enhanced the biological and cellular activity and skin regeneration, and this composition was assessed in regeneration of various tissues including skin and bone (Bhattarai et al. Citation2005). Hydrogel scaffolds are prepared for skin regeneration and repair and biological applications, because of their high water content and biocompatibility (Hoffman Citation2012).

Eross Guadalupe et al. report the fabrication and characterization of bioactive poly-e-caprolactone (PCL) nanofiber matrices incorporated with a model angiogenic factor, PNF1, and a model antibiotic drug, gentamicin (Guadalupe et al. Citation2015). This study shows that all the fiber matrices were able to support fibroblast growth and maintain normal cell morphology. Such bioactive bandages may serve as versatile and less expensive alternatives for the treatment of complex wounds.

In a study by Venugopal and coworkers, PCL and collagen were used to fabricate nanofiber matrices to observe cell attachment, morphology, proliferation, and cell–matrix interactions (Guadalupe et al. Citation2015). The results of this work show that this novel biodegradable PCL and collagen nanofiber matrices support the attachment and proliferation of human dermal fibroblasts and might have potential in tissue engineering as a dermal substitute for skin regeneration (Chandrasekaran et al. Citation2011).

Ravichandran et al. fabricated a mixture of nanofibrous scaffold comprising of PLLA/PAA/Col I&III with stem cell therapy along with the addition of bFGF to promote skin regeneration (Ravichandran et al. Citation2012b). This system induced the necessary paracrine signaling effect by faster regeneration of the damaged skin tissues.

Kalaipriya Madhaiyan et al. investigated the water soluble vitamin delivery with hydrophobic polymer nanofiber sustaining the release of the vitamin for the transdermal patch applications (Madhaiyan et al. Citation2013). They showed that the cyanocobalamin loaded nanofibers are suitable for transdermal patch according to the drug release profile in PBS buffer in vitro environment as the release of the energy supplement matches the requirement of the human need.

Bone regeneration

The promising application of electrospun nanofiber scaffolds in bone regeneration was initially investigated with fiber mesh of electrospun poly (ɛ-caprolactone) (PCL) to maintain ectopic bone formation in vivo (Shin et al. Citation2004) and osteogenic differentiation of rat bone marrow mesenchymal stem cells in vitro (Yoshimoto et al. Citation2003). Electrospun nanofiber scaffolds produced from a multiplicity of biomaterials were then used to maintain osteogenic differentiation of a variety of cell types in vitro, including mesenchymal and osteoblasts stem cells (Ngiam et al. Citation2009, Phipps et al. Citation2011, Zhang et al. Citation2008, Zhang et al. Citation2010). Interaction between cell-nanofiber scaffolds plays a significant role in osteogenesis process. The nanofiber scaffolds significantly improved the total amount of serum proteins adsorbed on the nanofiber scaffolds, particularly the key extracellular matrix proteins involved in cell adhesion (Woo et al. Citation2003), and enhanced the osteoblast differentiation, growth, and attachment (Woo et al. Citation2003, Citation2007). Embryonic stem (ES) cells (Thomson et al. Citation1998) are attractive enviable cell source for bone tissue regeneration, which is because of the pluripotency and unlimited expansion capacity (Handschel et al. Citation2010, Jukes et al. Citation2008). It has been verified that nanofiber matrix can considerably make possible the osteogenic differentiation of ES cells (Smith et al. Citation2009).

Bone regeneration, using a biomimetic scaffold to assemble a synthetic osteogenic microenvironment, makes possible the natural ossification procedures, which can improve clinical therapy (Shin et al. Citation2003).

Collagen is the main organic constituent of the bone extracellular matrix (ECM) (Elsdale and Bard Citation1972), and several studies informed that collagen nanofibers enhanced osteogenesis (Franceschi Citation1999, Xiao et al. Citation2002). As a result, complex of scaffolds-nanofibres were prepared to imitate the proosteogenic properties and structural features of collagenous ECM of the bone (Hartgerink et al. Citation2001, Li et al. Citation2002).

Bone regeneration is controlled by a variety of bioactive agents such as nucleic acids (Shea et al. Citation1999), integrin-binding ligands (Hern and Hubbell Citation1998), and growth factors (Richardson et al. Citation2001).

In one study, Wei and coworkers encapsulated nanofiber with bone morphogenetic protein-7 (BMP-7) and then implanted in rats subcutaneously (Wei et al. Citation2007). Then, BMP-7 was released in a regulated fashion with high biological activity and stimulated ectopic bone formation. But, scaffolds with passively adsorbed BMP-7 or without BMP-7 were incapable to provoke osteogenesis, probably due to the insufficient release duration and loss of bioactivity.

Rajeswari Ravichandran et al. investigated the in vitro responses of ADSCs to the surface mineralized PLLA/PBLG/Col/n-HA nanofibrous substrate, in terms of the initial cell adhesion, proliferation, and further osteogenic differentiation and mineralization (Ravichandran et al. Citation2012c). The results showed that the PLLA/PBLG/Col/n-HA scaffolds increased greater osteogenic differentiation of ADSC as evident from the enzyme activity and mineralization profiles for bone tissue engineering.

Chinnasamy Gandhimathi et al. investigated the in vitro response of hMSCs to the surface mineralized PCL/PAA/Col/n-HA hybrid nanofibrous scaffold, in terms of cell adhesion, proliferation, and further osteogenic differentiation and mineralization for bone tissue regeneration (Gandhimathi et al. Citation2013). They showed that the PCL/PAA/collagen/n-HA scaffolds promoted greater osteogenic differentiation of hMSCs, proving to be a potential hybrid scaffold for BTE.

Cartilage regeneration

In several studies of tissue-engineered auricle-shaped cartilage, natural or synthetic biodegradable polymers such as poly-ɛ-caprolactone, polylactic acid (PLA), collagen, polyglycolic acid (PGA), and hydrogel (fibrin gel, pluronics, alginate) have been prepared as scaffolds for cell seeding (Bichara et al. Citation2011, Isogai et al. Citation2005, Kamil et al. Citation2004, Vacanti et al. Citation1988).

Synthetic polymers, predominantly aliphatic polyesters such as polylactic-co-glycolic acid and PGA, have often been used in auricular cartilage engineering (Haisch et al. Citation2002, Isogai et al. Citation2005, Kamil et al. Citation2003, Liu et al. Citation2010).

Accumulating studies have focused on the interactions of these FDA-approved polymers with chondrocytes (Temenoff and Mikos Citation2000). For example, copolymer scaffolds of PGA/PLA have been extensively used in cartilage regeneration 17–20 and have revealed success in fabricating human auricle shaped constructs in vitro (Kamil et al. Citation2003).

Shieh and coworkers assessed scaffolds made of PGA coated with poly-4-hydroxybutyrate, PCL, and poly-l-lactic acid (PLLA) and compared the growth of engineered cartilage and ear-like shape preservation in vivo in both rabbit and nude mouse models (Shieh et al. Citation2004). The constructs were fabricated over 40 weeks in vivo in the nude mouse, thus detecting neocartilage creation in all specimens. PCL constructs revealed the best shape retention which is possibly because of the slowest degradation rate of this scaffold material, although they reduced in size over time.

Isogai and coworkers later informed numerous studies, one of which assessed the preservation of neocartilage integrity in an auricle-shaped construct in a nude mouse model, by using PLLA/PCL copolymer (Isogai et al. Citation2004). The results revealed that neocartilage had not been consistently dispersed all over the scaffold; insufficient cell-seeding methods were accountable, according to the authors. Isogai et al. prepared the similar PLLA/PCL copolymer blended with basic fibroblast growth factor (bFGF) encapsulated into microspheres generated by gelatin (Isogai et al. Citation2005). The prepared bFGF release enhanced chondrogenesis and improved vascularization of cell-seeded constructs blended in nude mice; shape maintenance was enhanced with addition of FGF because of enhanced chondrogenesis.

Ligament/tendon regeneration

The functions of the ligament are to prepare aid and support in joint movements. Basically ligament/tendon damages were treated by using autografts, allografts, or biological grafts (Bell Citation1995). In applications of ligament tissue engineering, scaffolds should make available appropriate shape and required mechanical strength during degrade and reconstruction at a rate similar to the regeneration rate of tissue. For ligament reconstruction both synthetic and natural materials have been used in the form of gels, membranes, or 3-D scaffolds (Figueroa et al. Citation2014, Lu et al. Citation2005, Rathbone et al. Citation2010, Wiig et al. Citation1990).

One of the most common ligament injuries of the knee is the rupture of the anterior cruciate ligament (ACL) (Fetto and Marshall Citation1980). Natalie Luanne Leong and coworkers examined the regenerative potential of tissue-engineered ACL grafts using immunohistochemistry, histology, and mechanical testing up to 16 weeks postoperatively (Leong et al. Citation2015). Mechanical testing of the grafts revealed significantly higher mechanical possessions than immediately post-implantation. Histology exhibited infiltration of the grafts with cells and immunohistochemistry revealed aligned collagen deposition with minimal inflammatory reaction. In this in vivo rodent model study for ACL reconstruction, the histological and mechanical evaluation revealed tremendous regeneration and healing possible of electrospun PCL ligament graft. The scaffold of electrospun polymer assisted both cell and matrix alignment in the regenerated ACL. These grafts caused inefficacious bone integration with augmented strength over time; load to failure augmented three-fold as compared with the recreated ACL instantly postoperatively. In an ovine study, it was established that at 52 weeks, the autografts had 67 and 38% the peak stiffness and load, respectively, of control ligaments (Scheffler et al. Citation2008).

Deepthi and coworkers, with the aim of developing a scaffold that could mimic the native ligament fibrous morphology, along with an ECM mimicking coating that can act as a cell or nutrient reservoir, developed aligned PCL micro/random nano- (PCL aligned multiscale) and random micro/nano (PCL random multiscale) fibers through electrospinning following which a coating with chitosan–hyaluronic acid hydrogel was given (Deepthi et al. Citation2015). This study implies the use of hydrogel coated systems to provide a reservoir for cells and nutrients and further modifications of these systems would make it promising for ligament regeneration.

Conclusion

Regenerative medicine and tissue engineering are the developing branches providing the requirement for regeneration of tissues damaged due to injury. The emerging field of regenerative medicine and tissue engineering targets to three main approaches: cellular therapies, origins of stem and progenitor cells, and on explaining how they persevere (or do not) in adult organisms and improvement of biomaterials to either provide a scaffold that can promote appropriate tissue formation and/or release bioactive factors to aid in the healing response. This filed regenerates damaged tissues by combining cell from the body with highly porous scaffold biomaterials which guide the growth of new tissues (Noorjahan and Sastry Citation2005, Pallela et al. Citation2012) and an effective technique in treating this is the use of nanofiber scaffolds. Finally, the recent progress in these fields needs to be combined to develop nanofiber scaffolds that would respond to the requirement of regenerative medicine.

Acknowledgements

The authors thank the Department of Medical Nanotechnology, Faculty of Advanced Medical Science of Tabriz University of Medical Sciences for all the support provided.

Disclosure statement

The authors declare that they have no competing interests.

References

  • 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.
  • Bell E. 1995. Strategy for the selection of scaffolds for tissue engineering. Tissue Eng. 1:163–179.
  • Bhattarai N, Edmondson D, Veiseh O, Matsen FA, Zhang M. 2005. Electrospun chitosan-based nanofibers and their cellular compatibility. Biomaterials. 26:6176–6184.
  • Bichara DA, O'Sullivan NA, Pomerantseva I, Zhao X, Sundback CA, Vacanti JP, Randolph MA. 2011. The tissue-engineered auricle: past, present, and future. Tissue Eng Part B Rev. 18:51–61.
  • Boland ED, Matthews JA, Pawlowski KJ, Simpson DG, Wnek GE, Bowlin GL. 2004. Electrospinning collagen and elastin: preliminary vascular tissue engineering. Front Biosci. 9:1422–1432.
  • Chandrasekaran A, Venugopal S, Sundarrajan S, Ramakrishna. 2011. Gelatin-enriched biomimetic scaffolds for skin-tissue engineering, Future Medicine Ltd Unitec House, 3rd Floor, 2 Albert Place, Finchley Central, London, N3 1QB, England.
  • Chew SY, Wen J, Yim EK, Leong KW. 2005. Sustained release of proteins from electrospun biodegradable fibers. Biomacromolecules. 6:2017–2024.
  • Cui W, Li X, Zhu X, Yu G, Zhou S, Weng J. 2006. Investigation of drug release and matrix degradation of electrospun poly(DL-lactide) fibers with paracetanol inoculation. Biomacromolecules. 7:1623–1629.
  • Daraee H, Etemadi A, Kouhi M, Alimirzalu S, Akbarzadeh A. 2016. Application of liposomes in medicine and drug delivery. Artif Cells Nanomed Biotechnol. 44, 381–391.
  • Deepthi S, Jeevitha K, Sundaram MN, Chennazhi K, Jayakumar R. 2015. Chitosan–hyaluronic acid hydrogel coated poly (caprolactone) multiscale bilayer scaffold for ligament regeneration. Chem Eng J. 260:478–485.
  • Deluzio TG, Seifu DG, Mequanint K. 2013. 3D scaffolds in tissue engineering and regenerative medicine: beyond structural templates?” Pharm Bioprocess. 1:267–281.
  • Elsdale T, Bard J. 1972. Collagen substrata for studies on cell behavior. J Cell Biol. 54:626–637.
  • Fetto JF, Marshall JL. 1980. The natural history and diagnosis of anterior cruciate ligament insufficiency. Clin Orthopaed Relat Res. 147:29–38.
  • Figueroa D, Espinosa M, Calvo R, Scheu M, Vaisman A, Gallegos M, Conget P. 2014. Anterior cruciate ligament regeneration using mesenchymal stem cells and collagen type I scaffold in a rabbit model. Knee Surg Sports Traumatol Arthrosc. 22:1196–1202.
  • Fisher MB, Mauck RL. 2013. Tissue engineering and regenerative medicine: recent innovations and the transition to translation. Tissue Eng B: Rev. 19:1–13.
  • Franceschi R. 1999. The developmental control of osteoblast-specific gene expression: role of specific transcription factors and the extracellular matrix environment. Crit Rev Oral Biol Med. 10:40–57.
  • Fujihara K, Kotaki M, Ramakrishna S. 2005. Guided bone regeneration membrane made of polycaprolactone/calcium carbonate composite nano-fibers. Biomaterials. 26:4139–4147.
  • Gandhimathi C, Venugopal J, Ravichandran R, Sundarrajan S, Suganya S, Ramakrishna S. 2013. Mimicking nanofibrous hybrid bone substitute for mesenchymal stem cells differentiation into osteogenesis. Macromol Biosci. 13:696–706.
  • Guadalupe E, Ramos D, Shelke NB, James R, Gibney C, Kumbar SG. 2015. Bioactive polymeric nanofiber matrices for skin regeneration. J Appl Polym Sci. 132:41879. DOI: 10.1002/app.41879.
  • Haisch A, Kläring S, Gröger A, Gebert C, Sittinger M. 2002. A tissue-engineering model for the manufacture of auricular-shaped cartilage implants. Eur Arch Otorhinolaryngol. 259:316–321.
  • Handschel J, Naujoks C, Langenbach F, Berr K, Depprich RA, Ommerborn MA, et al. 2010. Comparison of ectopic bone formation of embryonic stem cells and cord blood stem cells in vivo. Tissue Eng A. 16:2475–2483.
  • 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.
  • Hern DL, Hubbell JA. 1998. Incorporation of adhesion peptides into nonadhesive hydrogels useful for tissue resurfacing. J Biomed Mater Res. 39:266–276.
  • Hoffman AS. 2012. Hydrogels for biomedical applications. Adv Drug Deliv Rev. 64:18–23.
  • Huang Y, Onyeri S, Siewe M, Moshfeghian A, Madihally SV. 2005. In vitro characterization of chitosan-gelatin scaffolds for tissue engineering. Biomaterials. 26:7616–7627.
  • Huang ZM, Zhang YZ, Kotaki M, Ramakrishna S. 2003. A review on polymer nanofibers by electrospinning and their applications in nanocomposites. Compos Sci Technol. 63:2223–2253.
  • Isogai N, Asamura S, Higashi T, Ikada Y, Morita S, Hillyer J, Jacquet R, Landis WJ. 2004. Tissue engineering of an auricular cartilage model utilizing cultured chondrocyte-poly (L-lactide-ɛ-caprolactone) scaffolds. Tissue Eng. 10:673–687.
  • Isogai N, Morotomi T, Hayakawa S, Munakata H, Tabata Y, Ikada Y, Kamiishi H. 2005. Combined chondrocyte–copolymer implantation with slow release of basic fibroblast growth factor for tissue engineering an auricular cartilage construct. J Biomed Mater Res A. 74:408–418.
  • 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.
  • Jukes JM, Both SK, Leusink A, Lotus MT, Van Blitterswijk CA, De Boer J. 2008. Endochondral bone tissue engineering using embryonic stem cells. Proc Natl Acad Sci USA. 105:6840–6845.
  • Jukola H, Nikkola L, Gomes M, Reis R, Ashammakhi N. 2008. Electrospun Starch‐Polycaprolactone Nanofiber‐Based Constructs for Tissue Engineering. Multiscale and Functionally Graded Materials 2006 (M&FGM 2006), Oahu Island (Hawaii): AIP Conference Proceedings, 973:15–19.
  • Kamil S, Vacanti M, Aminuddin B, Jackson M, Vacanti C, Eavey R. 2004. Tissue engineering of a human sized and shaped auricle using a mold. Laryngoscope. 114:867–870.
  • Kamil SH, Kojima K, Vacanti MP, Bonassar LJ, Vacanti CA, Eavey RD. 2003. In vitro tissue engineering to generate a human-sized auricle and nasal tip. Laryngoscope. 113:90–94.
  • Khil MS, SR, Bhattarai HY, Kim SZ. Kim KH, Lee 2005. Novel fabricated matrix via electrospinning for tissue engineering. J Biomed Mater Res B Appl Biomater.72:117–124.
  • Laflamme MA, Murry CE. 2011. Heart regeneration. Nature. 473:326–335.
  • Lee CH, Shin HJ, Cho IH, Kang YM, Kim IA, Park KD, Shin JW. 2005. Nanofiber alignment and direction of mechanical strain affect the ECM production of human ACL fibroblast. Biomaterials. 26:1261–1270.
  • Leong NL, Kabir N, Arshi A, Nazemi A, Wu B, Petrigliano FA, McAllister DR. 2015. Evaluation of Polycaprolactone scaffold with basic fibroblast growth factor and fibroblasts in an athymic rat model for anterior cruciate ligament reconstruction. Tissue Eng A. 21:1859–1868.
  • 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 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 (ɛ‐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.
  • Liu Y, Zhang L, Zhou G, Li Q, Liu W, Yu Z, et al. 2010. In vitro engineering of human ear-shaped cartilage assisted with CAD/CAM technology. Biomaterials. 31:2176–2183.
  • Lu HH, Cooper JA, Manuel S, Freeman JW, Attawia MA, Ko FK, Laurencin CT. 2005. Anterior cruciate ligament regeneration using braided biodegradable scaffolds: in vitro optimization studies. Biomaterials. 26:4805–4816.
  • Luong-Van E, Grøndahl L, Chua KN, Leong KW, Nurcombe V, Cool SM. 2006. Controlled release of heparin from poly(epsilon-caprolactone) electrospun fibers. Biomaterials. 27:2042–2050.
  • Ma Z, He W, Yong T, Ramakrishna S. 2005. 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.
  • Madhaiyan K, Sridhar R, Sundarrajan S, Venugopal JR, Ramakrishna S. 2013. Vitamin B 12 loaded polycaprolactone nanofibers: a novel transdermal route for the water soluble energy supplement delivery. Int J Pharm. 444:70–76.
  • Mellatyar H, Akbarzadeh A, Rahmati M, Ghalhar MG, Etemadi A, Nejati-Koshki K, Zarghami N, Barkhordari A. 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 MEA, Ramakrishna S. 2004. Electrospun P(LLA-CL) nanofiber: a biomimetic extracellular matrix for smooth muscle cell and endothelial cell proliferation. Biomaterials. 25:1883–1890.
  • Ngiam M, Liao S, Patil AJ, Cheng Z, Chan CK, Ramakrishna S. 2009. The fabrication of nano-hydroxyapatite on PLGA and PLGA/collagen nanofibrous composite scaffolds and their effects in osteoblastic behavior for bone tissue engineering. Bone. 45:4–16.
  • Nikkola L, Viitanen P, Ashammakhi N. 2005. Multi-component implant for true controlled release of diclofenac sodium. 6th International Symposium on Frontiers in Biomedical Polymers.
  • Noorjahan S, Sastry T. 2005. Physiologically clotted fibrin‐calcined bone composite—a possible bone graft substitute. J Biomed Mater Res B: Appl Biomater. 75:343–350.
  • Pallela R, Venkatesan J, Janapala VR, Kim SK. 2012. Biophysicochemical evaluation of chitosan‐hydroxyapatite‐marine sponge collagen composite for bone tissue engineering. J Biomed Mater Res A. 100:486–495.
  • Phipps MC, Clem WC, Catledge SA, Xu Y, Hennessy KM, Thomas V, et al. 2011. Mesenchymal stem cell responses to bone-mimetic electrospun matrices composed of polycaprolactone, collagen I and nanoparticulate hydroxyapatite. PLoS One. 6:e16813.
  • Rathbone S, Furrer P, Lübben J, Zinn M, Cartmell S. 2010. Biocompatibility of polyhydroxyalkanoate as a potential material for ligament and tendon scaffold material. J Biomed Mater Res A. 93:1391–1403.
  • Ravichandran R, Sundarrajan S, Venugopal JR, Mukherjee S, Ramakrishna S. 2012a. Advances in polymeric systems for tissue engineering and biomedical applications. Macromol Biosci. 12:286–311.
  • Ravichandran R, JR, Venugopal S, Sundarrajan S. Mukherjee S, Ramakrishna 2012b. Precipitation of nanohydroxyapatite on PLLA/PBLG/collagen nanofibrous structures for the differentiation of adipose derived stem cells to osteogenic lineage. Biomaterials. 33:846–855.
  • Ravichandran R, Venugopal JR, Sundarrajan S, Mukherjee S, Sridhar R, Ramakrishna S. 2012c. Composite poly-l-lactic acid/poly-(α, β)-dl-aspartic acid/collagen nanofibrous scaffolds for dermal tissue regeneration. Mater Sci Eng: C. 32:1443–1451.
  • 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.
  • Richardson TP, Peters MC, Ennett AB, Mooney DJ. 2001. Polymeric system for dual growth factor delivery. Nat Biotechnol. 19:1029–1034.
  • 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.
  • Scheffler SU, Schmidt T, Gangéy I, Dustmann M, Unterhauser F, Weiler A. 2008. Fresh-frozen free-tendon allografts versus autografts in anterior cruciate ligament reconstruction: delayed remodeling and inferior mechanical function during long-term healing in sheep. Arthroscopy. 24:448–458.
  • Shea LD, Smiley E, Bonadio J, Mooney DJ. 1999. DNA delivery from polymer matrices for tissue engineering. Nat Biotechnol. 17:551–554.
  • Shieh SJ, Terada S, Vacanti JP. 2004. Tissue engineering auricular reconstruction: in vitro and in vivo studies. Biomaterials. 25:1545–1557.
  • Shields KJ, Beckman MJ, Bowlin GL, Wayne JS. 2004. Mechanical properties and cellular proliferation of electrospun collagen type II. Tissue Eng. 10:1510–1517.
  • Shin H, Jo S, Mikos AG. 2003. Biomimetic materials for tissue engineering. Biomaterials. 24:4353–4364.
  • 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.
  • Sindelar T, Nikkola L, Ashammakhi N, van Griensven M, Redl H. 2006. Electrospinning of fibrinogen nanofibers. Eur Cells Mater. 11:59.
  • Smith LA, Liu X, Hu J, Wang P, Ma PX. 2009. Enhancing osteogenic differentiation of mouse embryonic stem cells by nanofibers. Tissue Eng A. 15:1855–1864.
  • 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.
  • Stitzel JD, Bowlin GL, Mansfield K, Wnek GE, Simpson DG. 2000. Electrospraying and electrospinning of polymers for biomedical applications. Poly (lactic-co-glycolic acid) and poly (ethylene-co-vinylacetate). International SAMPE Technical Conference.
  • Temenoff JS, Mikos AG. 2000. Review: tissue engineering for regeneration of articular cartilage. Biomaterials. 21:431–440.
  • Thomson JA, Itskovitz-Eldor J, Shapiro SS, Waknitz MA, Swiergiel JJ, Marshall VS, Jones JM. 1998. Embryonic stem cell lines derived from human blastocysts. Science. 282:1145–1147.
  • 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.
  • Um IC, Fang D, Hsiao BS, Okamoto A, Chu B. 2004. Electro-spinning and electro-blowing of hyaluronic acid. Biomacromolecules. 5:1428–1436.
  • Vacanti JP, Morse MA, Saltzman WM, Domb AJ, Perez-Atayde A, Langer R. 1988. Selective cell transplantation using bioabsorbable artificial polymers as matrices. J Pediatr Surg. 23:3–9.
  • Venugopal J, Low S, Choon AT, Ramakrishna S. 2008. Interaction of cells and nanofiber scaffolds in tissue engineering. J Biomed Mater Res B Appl Biomater.84:34–48.
  • Venugopal J, Ma L, Yong T, Ramakrishna S. 2005. In vitro study of smooth muscle cells on polycaprolactone and collagen nanofibrous matrices. Cell Biol Int. 29:861–867.
  • Wei G, Jin Q, Giannobile WV, Ma PX. 2007. The enhancement of osteogenesis by nano-fibrous scaffolds incorporating rhBMP-7 nanospheres. Biomaterials. 28:2087–2096.
  • Wiig ME, Amiel D, Vandeberg J, Kitabayashi L, Harwood FL, Arfors KE. 1990. The early effect of high molecular weight hyaluronan (hyaluronic acid) on anterior cruciate ligament healing: an experimental study in rabbits. J Orthopaed Res. 8:425–434.
  • Wnek GE, Carr ME, Simpson DG, Bowlin GL. 2003. Electrospinning of nanofiber fibrinogen structures. Nano Lett. 3:213–216.
  • Woo KM, Chen VJ, Ma PX. 2003. Nano‐fibrous scaffolding architecture selectively enhances protein adsorption contributing to cell attachment. J Biomed Mater Res A. 67:531–537.
  • Woo KM, Jun JH, Chen VJ, Seo J, Baek JH, Ryoo HM, et al. 2007. Nano-fibrous scaffolding promotes osteoblast differentiation and biomineralization. Biomaterials. 28:335–343.
  • Xiao G, Gopalakrishnan R, Jiang D, Reith E, Benson MD, Franceschi RT. 2002. Bone morphogenetic proteins, extracellular matrix, and mitogen‐activated protein kinase signaling pathways are required for osteoblast‐specific gene expression and differentiation in MC3T3‐E1 cells. J Bone Miner Res. 17:101–110.
  • 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 F, Murugan R, Ramakrishna S, Wang X, Ma YX, Wang S. 2004. Fabrication of nano-structured porous PLLA scaffold intended for nerve tissue engineering. Biomaterials. 25:1891–1900.
  • Yang Y, Nikkola L, Ylikauppila H, Ashammakhi N. 2006. Investigation of cell attachment on the scaffolds manufactured by electrospun PCL-hyaluronan blend. TNT2006 Trends in Nanotechnology Conference.
  • 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.
  • Zhang Y, Reddy VJ, Wong SY, Li X, Su B, Ramakrishna S, Lim CT. 2010. Enhanced biomineralization in osteoblasts on a novel electrospun biocomposite nanofibrous substrate of hydroxyapatite/collagen/chitosan. Tissue Eng A. 16:1949–1960.
  • Zhang Y, JR, Venugopal A, El-Turki S, Ramakrishna B. Su CT, Lim 2008. Electrospun biomimetic nanocomposite nanofibers of hydroxyapatite/chitosan for bone tissue engineering. Biomaterials. 29:4314–4322.
  • Zhu XH, Wang CH, Tong YW. 2009. In vitro characterization of hepatocyte growth factor release from PHBV/PLGA microsphere scaffold. J Biomed Mater Res A. 89:411–423.

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.