1
|
Romero-Alemán MDM, Pérez-Galván JM, Hernández-Rodríguez JE, Monzón-Mayor M. The Potential of Aloe Vera in Solution and in Blended Nanofibers Containing Poly (3-Hydroxybutyrate-Co-3-Hydroxyvalerate) as Substrates for Neurite Outgrowth. J Biomed Mater Res A 2025; 113:e37825. [PMID: 39462944 DOI: 10.1002/jbm.a.37825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 09/22/2024] [Accepted: 10/14/2024] [Indexed: 10/29/2024]
Abstract
This pilot study investigated the potential of aloe vera (AV) to promote neurite outgrowth in organotypic dorsal root ganglia (DRG) explants (n = 230) from neonatal rats (n = 15). Using this in vitro model of acute axotomy, we assessed neurite outgrowth exceeding 1.5 times the explant diameter (viable explants) and measured the longest neurite length. These parameters were evaluated under control conditions and in cultures supplemented with commercial AV and four aligned scaffolds: poly-L-lactate (PLLA), polydioxanone (PDS), poly(3-hydroxybutyrate-co-3-hydroxyvalerate) (PHBV), and blended PHBV/AV. After 6 days of culture, explants were immunostained using neuron-specific marker Tuj1 and Schwann cell marker S100. Measurements were obtained with Image J software and analyzed using Jamovi 2.3. In control and AV dilution media, the study revealed radial tissue growth from the explant body with randomly oriented neurites, whereas in all scaffolds, bidirectional tissue growth occurred parallel to nanofibers. Binomial logistic regression analyses indicated that viable explants were more likely in the control group compared to PDS (p = 0.0042) and PHBV (p < 0.0001), with non-significant differences when compared to AV dilution, PLLA, and PHBV/AV. AV dilution showed a greater association with viable explants than PLLA (p = 0.0459), while non-significant difference was found between AV dilution and PHBV/AV. Additionally, the PHBV/AV scaffold predicted higher odds of viable explants than PLLA (p = 0.0479), PDS (p = 0.0001), and PHBV (p < 0.0001). Groups with similar probabilities of obtaining viable explants (control, AV dilution, and PHBV/AV) exhibited non-significant differences in their longest neurite lengths. In conclusion, control, AV dilution, and PHBV/AV yielded the highest probability of developing viable explants and the longest neurite lengths. This is the first study demonstrating the direct permissiveness of AV for axonal outgrowth. Furthermore, the blended PHBV/AV scaffold showed significant potential as a suitable scaffold for axonal regrowth and Schwann cell migration, ensuring controlled tissue formation for tissue engineering applications.
Collapse
Affiliation(s)
- María-Del-Mar Romero-Alemán
- Instituto Universitario de Investigaciones Biomédicas y Sanitarias, Universidad de las Palmas de Gran Canaria, Las Palmas, Spain
- Departamento de Morfología, Universidad de Las Palmas de Gran Canaria, Las Palmas, Spain
| | - José-Manuel Pérez-Galván
- Instituto Universitario de Investigaciones Biomédicas y Sanitarias, Universidad de las Palmas de Gran Canaria, Las Palmas, Spain
| | | | - Maximina Monzón-Mayor
- Departamento de Morfología, Universidad de Las Palmas de Gran Canaria, Las Palmas, Spain
| |
Collapse
|
2
|
Lian S, Lamprou D, Zhao M. Electrospinning technologies for the delivery of Biopharmaceuticals: Current status and future trends. Int J Pharm 2024; 651:123641. [PMID: 38029864 DOI: 10.1016/j.ijpharm.2023.123641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 11/15/2023] [Accepted: 11/26/2023] [Indexed: 12/01/2023]
Abstract
This review provides an in-depth exploration of electrospinning techniques employed to produce micro- or nanofibres of biopharmaceuticals using polymeric solutions or melts with high-voltage electricity. Distinct from prior reviews, the current work narrows its focus on the recent developments and advanced applications in biopharmaceutical formulations. It begins with an overview of electrospinning principles, covering both solution and melt modes. Various methods for incorporating biopharmaceuticals into electrospun fibres, such as surface adsorption, blending, emulsion, co-axial, and high-throughput electrospinning, are elaborated. The review also surveys a wide array of biopharmaceuticals formulated through electrospinning, thereby identifying both opportunities and challenges in this emerging field. Moreover, it outlines the analytical techniques for characterizing electrospun fibres and discusses the legal and regulatory requirements for their production. This work aims to offer valuable insights into the evolving realm of electrospun biopharmaceutical delivery systems.
Collapse
Affiliation(s)
- Shangjie Lian
- School of Pharmacy, Queen's University Belfast, Belfast BT9 7BL, UK
| | | | - Min Zhao
- School of Pharmacy, Queen's University Belfast, Belfast BT9 7BL, UK; China Medical University- Queen's University Belfast Joint College (CQC), China Medical University, Shenyang 110000, China
| |
Collapse
|
3
|
Behtaj S, Karamali F, Najafian S, Masaeli E, Rybachuk M. Ciliary neurotrophic factor mediated growth of retinal ganglion cell axons on PGS/PCL scaffolds. Biomed Mater 2024; 19:025001. [PMID: 38181445 DOI: 10.1088/1748-605x/ad1bae] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 01/05/2024] [Indexed: 01/07/2024]
Abstract
Ciliary neurotrophic factor (CNTF) promotes survival and/or differentiation of a variety of neuronal cells including retinal ganglion cells (RGCs). Delivery of CNTF requires a suitable medium capable of mediating diffusion and premature release of CNTF within the target tissue. Polymeric tissue-engineered scaffolds have been readily used as substrates for cell transplantation, expansion, and differentiation and, as carriers of cell growth factors. Their functions to CNTF release for RGC proliferation have remained so far unexplored, especially to CNTF affinity to the scaffold and subsequent RGC fate. Electrospunpoly(glycerol sebacate)/poly(ϵ-caprolactone) (PGS/PCL) biopolymer scaffolds have recently shown promising results in terms of supporting regeneration of RGC neurites. This work explores covalent immobilization of CNTF on PGS/PCL scaffold and the way immobilised CNTF mediates growth of RGC axons on the scaffold. Anex-vivothree-dimensional model of rodent optic nerve on PGS/PCL revealed that RGC explants cultured in CNTF mediated environment increased their neurite extensions after 20 d of cell culture employing neurite outgrowth measurements. The CNTF secretion on PGS/PCL scaffold was found bio-mimicking natural extracellular matrix of the cell target tissue and, consequently, has shown a potential to improve the overall efficacy of the RGC regeneration process.
Collapse
Affiliation(s)
- Sanaz Behtaj
- School of Engineering and Built Environment, Griffith University, Engineering Drive, Southport QLD 4222, Australia
- Department of Animal Biotechnology, Cell Science Research Centre, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
- Clem Jones Centre for Neurobiology and Stem Cell Research, Menzies Health Institute Queensland, Griffith University, Gold Coast QLD 4222, Australia
| | - Fereshteh Karamali
- Department of Animal Biotechnology, Cell Science Research Centre, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Samaneh Najafian
- Department of Animal Biotechnology, Cell Science Research Centre, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Elahe Masaeli
- Department of Animal Biotechnology, Cell Science Research Centre, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Maksym Rybachuk
- School of Engineering and Built Environment, Griffith University, 170 Kessels Road, Nathan QLD 4111, Australia
- Centre for Quantum Dynamics and Australian Attosecond Science Facility, Griffith University, Science Road, Nathan QLD 4111, Australia
| |
Collapse
|
4
|
Dos Santos FV, Siqueira RL, de Morais Ramos L, Yoshioka SA, Branciforti MC, Correa DS. Silk fibroin-derived electrospun materials for biomedical applications: A review. Int J Biol Macromol 2024; 254:127641. [PMID: 37913875 DOI: 10.1016/j.ijbiomac.2023.127641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 10/14/2023] [Accepted: 10/22/2023] [Indexed: 11/03/2023]
Abstract
Electrospinning is a versatile technique for fabricating polymeric fibers with diameters ranging from micro- to nanoscale, exhibiting multiple morphologies and arrangements. By combining silk fibroin (SF) with synthetic and/or natural polymers, electrospun materials with outstanding biological, chemical, electrical, physical, mechanical, and optical properties can be achieved, fulfilling the evolving biomedical demands. This review highlights the remarkable versatility of SF-derived electrospun materials, specifically focusing on their application in tissue regeneration (including cartilage, cornea, nerves, blood vessels, bones, and skin), disease treatment (such as cancer and diabetes), and the development of controlled drug delivery systems. Additionally, we explore the potential future trends in utilizing these nanofibrous materials for creating intelligent biomaterials, incorporating biosensors and wearable sensors for monitoring human health, and also discuss the bottlenecks for its widespread use. This comprehensive overview illuminates the significant impact and exciting prospects of SF-derived electrospun materials in advancing biomedical research and applications.
Collapse
Affiliation(s)
- Francisco Vieira Dos Santos
- Nanotechnology National Laboratory for Agriculture, Embrapa Instrumentação, 13560-970 São Carlos, SP, Brazil; Materials Engineering Department, São Carlos School of Engineering, University of São Paulo, 13563-120 São Carlos, SP, Brazil
| | - Renato Luiz Siqueira
- Materials Engineering Department, Federal University of São Carlos, 13565-905 São Carlos, SP, Brazil
| | - Lucas de Morais Ramos
- São Carlos Institute of Physics, University of São Paulo, 13560-970 São Carlos, SP, Brazil
| | - Sérgio Akinobu Yoshioka
- Laboratory of Biochemistry and Biomaterials, São Carlos Institute of Chemistry, University of São Paulo, 13560-970 São Carlos, SP, Brazil
| | - Márcia Cristina Branciforti
- Materials Engineering Department, São Carlos School of Engineering, University of São Paulo, 13563-120 São Carlos, SP, Brazil
| | - Daniel Souza Correa
- Nanotechnology National Laboratory for Agriculture, Embrapa Instrumentação, 13560-970 São Carlos, SP, Brazil; Materials Engineering Department, São Carlos School of Engineering, University of São Paulo, 13563-120 São Carlos, SP, Brazil.
| |
Collapse
|
5
|
Bianchini M, Micera S, Redolfi Riva E. Recent Advances in Polymeric Drug Delivery Systems for Peripheral Nerve Regeneration. Pharmaceutics 2023; 15:pharmaceutics15020640. [PMID: 36839962 PMCID: PMC9965241 DOI: 10.3390/pharmaceutics15020640] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 02/03/2023] [Accepted: 02/06/2023] [Indexed: 02/17/2023] Open
Abstract
When a traumatic event causes complete denervation, muscle functional recovery is highly compromised. A possible solution to this issue is the implantation of a biodegradable polymeric tubular scaffold, providing a biomimetic environment to support the nerve regeneration process. However, in the case of consistent peripheral nerve damage, the regeneration capabilities are poor. Hence, a crucial challenge in this field is the development of biodegradable micro- nanostructured polymeric carriers for controlled and sustained release of molecules to enhance nerve regeneration. The aim of these systems is to favor the cellular processes that support nerve regeneration to increase the functional recovery outcome. Drug delivery systems (DDSs) are interesting solutions in the nerve regeneration framework, due to the possibility of specifically targeting the active principle within the site of interest, maximizing its therapeutical efficacy. The scope of this review is to highlight the recent advances regarding the study of biodegradable polymeric DDS for nerve regeneration and to discuss their potential to enhance regenerative performance in those clinical scenarios characterized by severe nerve damage.
Collapse
Affiliation(s)
- Marta Bianchini
- The BioRobotics Institute, Department of Excellence in Robotics and AI, Scuola Superiore Sant’Anna, 56127 Pisa, Italy
| | - Silvestro Micera
- The BioRobotics Institute, Department of Excellence in Robotics and AI, Scuola Superiore Sant’Anna, 56127 Pisa, Italy
- Translational Neuroengineering, Centre for Neuroprosthetics and Institute of Bioengineering, School of Engineering, École Polytechnique Fédérale de Lausanne (EPFL), 1000 Lausanne, Switzerland
| | - Eugenio Redolfi Riva
- The BioRobotics Institute, Department of Excellence in Robotics and AI, Scuola Superiore Sant’Anna, 56127 Pisa, Italy
- Correspondence:
| |
Collapse
|
6
|
Khosropanah MH, Majidi Zolbin M, Kajbafzadeh AM, Amani L, Harririan I, Azimzadeh A, Nejatian T, Alizadeh Vaghsloo M, Hassannejad Z. Evaluation and Comparison of the Effects of Mature Silkworm ( Bombyx mori) and Silkworm Pupae Extracts on Schwann Cell Proliferation and Axon Growth: An In Vitro Study. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2022; 21:e133552. [PMID: 36896320 PMCID: PMC9990520 DOI: 10.5812/ijpr-133552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 01/18/2023] [Accepted: 01/23/2023] [Indexed: 02/23/2023]
Abstract
Background Silkworm products were first used by physicians more than 8500 years ago, in the early Neolithic period. In Persian medicine, silkworm extract has several uses for treating and preventing neurological, cardiac, and liver diseases. Mature silkworms (Bombyx mori) and their pupae contain a variety of growth factors and proteins that can be used in many repair processes, including nerve regeneration. Objectives The study aimed to evaluate the effects of mature silkworm (Bombyx mori), and silkworm pupae extract on Schwann cell proliferation and axon growth. Methods Silkworm (Bombyx mori) and silkworm pupae extracts were prepared. Then, the concentration and type of amino acids and proteins in the extracts were evaluated by Bradford assay, sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE), and liquid chromatograph-mass spectrometer (LC-MS/MS). Also, the regenerative potential of extracts for improving Schwann cell proliferation and axon growth was examined by 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyl tetrazolium bromide (MTT) assay, electron microscopy, and NeuroFilament-200 (NF-200) immunostaining. Results According to the results of the Bradford test, the total protein content of pupae extract was almost twice that of mature worm extract. Also, SDS-PAGE analysis revealed numerous proteins and growth factors, such as bombyrin and laminin, in extracts that are involved in the repair of the nervous system. In accordance with Bradford's results, the evaluation of extracts using LC-MS/MS revealed that the number of amino acids in pupae extract was higher than in mature silkworm extract. It was found that the proliferation of Schwann cells at a concentration of 0.25 mg/mL in both extracts was higher than the concentrations of 0.01 and 0.05 mg/mL. When using both extracts on dorsal root ganglion (DRGs), an increase in length and number was observed in axons. Conclusions The findings of this study demonstrated that extracts obtained from silkworms, especially pupae, can play an effective role in Schwann cell proliferation and axonal growth, which can be strong evidence for nerve regeneration, and, consequently, repairing peripheral nerve damage.
Collapse
Affiliation(s)
- Mohammad Hossein Khosropanah
- Department of Traditional Medicine, School of Persian Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Masoumeh Majidi Zolbin
- Pediatric Urology and Regenerative Medicine Research Center, Children’s Medical Center, Gene, Cell and Tissue Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Abdol-Mohammad Kajbafzadeh
- Pediatric Urology and Regenerative Medicine Research Center, Children’s Medical Center, Gene, Cell and Tissue Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Leili Amani
- Department of Traditional Pharmacy, School of Persian Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ismaeil Harririan
- Department of Pharmaceutical Biomaterials, Medical Biomaterials Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Ashkan Azimzadeh
- Pediatric Urology and Regenerative Medicine Research Center, Children’s Medical Center, Gene, Cell and Tissue Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Touraj Nejatian
- AFHEA Prosthodontics and ORE University College London, London, England
| | - Mahdi Alizadeh Vaghsloo
- Department of Traditional Medicine, School of Persian Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Persian Medicine Network, Universal Scientific Education and Research Network, Tehran, Iran
- Corresponding Author: Department of Traditional Medicine, School of Persian Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Zahra Hassannejad
- Pediatric Urology and Regenerative Medicine Research Center, Children’s Medical Center, Gene, Cell and Tissue Research Institute, Tehran University of Medical Sciences, Tehran, Iran
- Corresponding Author: Pediatric Urology and Regenerative Medicine Research Center, Children’s Medical Center, Gene, Cell and Tissue Research Institute, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
7
|
Lee S, Patel M, Patel R. Electrospun nanofiber nerve guidance conduits for peripheral nerve regeneration: A review. Eur Polym J 2022. [DOI: 10.1016/j.eurpolymj.2022.111663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
8
|
De I, Sharma P, Singh M. Emerging approaches of neural regeneration using physical stimulations solely or coupled with smart piezoelectric nano-biomaterials. Eur J Pharm Biopharm 2022; 173:73-91. [DOI: 10.1016/j.ejpb.2022.02.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 02/18/2022] [Accepted: 02/22/2022] [Indexed: 01/20/2023]
|
9
|
Fitzpatrick V, Martín-Moldes Z, Deck A, Torres-Sanchez R, Valat A, Cairns D, Li C, Kaplan DL. Functionalized 3D-printed silk-hydroxyapatite scaffolds for enhanced bone regeneration with innervation and vascularization. Biomaterials 2021; 276:120995. [PMID: 34256231 PMCID: PMC8408341 DOI: 10.1016/j.biomaterials.2021.120995] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 06/20/2021] [Accepted: 06/24/2021] [Indexed: 02/07/2023]
Abstract
Our goal was to generate functionalized 3D-printed scaffolds for bone regeneration using silk-hydroxyapatite bone cements and osteoinductive, proangiogenic and neurotrophic growth factors or morphogens for accelerated bone formation. 3D printing was utilized to generate macroporous scaffolds with controlled geometries and architectures that promote osseointegration. We build on the knowledge that the osteoinductive factor Bone Morphogenetic Protein-2 (BMP2) can also positively impact vascularization, Vascular Endothelial Growth Factor (VEGF) can impact osteoblastic differentiation, and that Neural Growth Factor (NGF)-mediated signaling can influence bone regeneration. We assessed functions on the 3D printed construct via the osteogenic differentiation of human mesenchymal stem cells; migration and proliferation of human umbilical vein endothelial cells; and proliferation of human induced neural stem cells. The scaffolds provided mechanical properties suitable for bone and the materials were cytocompatible, osteoconductive and maintained the activity of the morphogens and cytokines. Synergistic outcomes between BMP-2, VEGF and NGF in terms of osteoblastic differentiation in vitro were identified, based on the upregulation of genes associated with osteoblastic differentiation (Runt-related transcription factor-2, Osteopontin, Bone Sialoprotein). Additional studies will be required to assess these scaffold designs in vivo. These results are expected to have a strong impact in bone regeneration in dental, oral and maxillofacial surgery.
Collapse
Affiliation(s)
- Vincent Fitzpatrick
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Zaira Martín-Moldes
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Anna Deck
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | | | - Anne Valat
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Dana Cairns
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Chunmei Li
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA.
| |
Collapse
|
10
|
Hu X, Ricci S, Naranjo S, Hill Z, Gawason P. Protein and Polysaccharide-Based Electroactive and Conductive Materials for Biomedical Applications. Molecules 2021; 26:4499. [PMID: 34361653 PMCID: PMC8348981 DOI: 10.3390/molecules26154499] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/23/2021] [Accepted: 07/24/2021] [Indexed: 11/16/2022] Open
Abstract
Electrically responsive biomaterials are an important and emerging technology in the fields of biomedical and material sciences. A great deal of research explores the integral role of electrical conduction in normal and diseased cell biology, and material scientists are focusing an even greater amount of attention on natural and hybrid materials as sources of biomaterials which can mimic the properties of cells. This review establishes a summary of those efforts for the latter group, detailing the current materials, theories, methods, and applications of electrically conductive biomaterials fabricated from protein polymers and polysaccharides. These materials can be used to improve human life through novel drug delivery, tissue regeneration, and biosensing technologies. The immediate goal of this review is to establish fabrication methods for protein and polysaccharide-based materials that are biocompatible and feature modular electrical properties. Ideally, these materials will be inexpensive to make with salable production strategies, in addition to being both renewable and biocompatible.
Collapse
Affiliation(s)
- Xiao Hu
- Department of Physics and Astronomy, Rowan University, Glassboro, NJ 08028, USA; (S.R.); (Z.H.)
- Department of Biomedical Engineering, Rowan University, Glassboro, NJ 08028, USA; (S.N.); (P.G.)
- Department of Molecular and Cellular Biosciences, Rowan University, Glassboro, NJ 08028, USA
| | - Samuel Ricci
- Department of Physics and Astronomy, Rowan University, Glassboro, NJ 08028, USA; (S.R.); (Z.H.)
| | - Sebastian Naranjo
- Department of Biomedical Engineering, Rowan University, Glassboro, NJ 08028, USA; (S.N.); (P.G.)
| | - Zachary Hill
- Department of Physics and Astronomy, Rowan University, Glassboro, NJ 08028, USA; (S.R.); (Z.H.)
| | - Peter Gawason
- Department of Biomedical Engineering, Rowan University, Glassboro, NJ 08028, USA; (S.N.); (P.G.)
| |
Collapse
|
11
|
Kochhar D, DeBari MK, Abbott RD. The Materiobiology of Silk: Exploring the Biophysical Influence of Silk Biomaterials on Directing Cellular Behaviors. Front Bioeng Biotechnol 2021; 9:697981. [PMID: 34239865 PMCID: PMC8259510 DOI: 10.3389/fbioe.2021.697981] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 05/31/2021] [Indexed: 11/13/2022] Open
Abstract
Biophysical properties of the extracellular environment dynamically regulate cellular fates. In this review, we highlight silk, an indispensable polymeric biomaterial, owing to its unique mechanical properties, bioactive component sequestration, degradability, well-defined architectures, and biocompatibility that can regulate temporospatial biochemical and biophysical responses. We explore how the materiobiology of silks, both mulberry and non-mulberry based, affect cell behaviors including cell adhesion, cell proliferation, cell migration, and cell differentiation. Keeping in mind the novel biophysical properties of silk in film, fiber, or sponge forms, coupled with facile chemical decoration, and its ability to match functional requirements for specific tissues, we survey the influence of composition, mechanical properties, topography, and 3D geometry in unlocking the body's inherent regenerative potential.
Collapse
Affiliation(s)
- Dakshi Kochhar
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, United States
| | - Megan K. DeBari
- Department of Materials Science and Engineering, Carnegie Mellon University, Pittsburgh, PA, United States
| | - Rosalyn D. Abbott
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, United States
| |
Collapse
|
12
|
Anup N, Chavan T, Chavan S, Polaka S, Kalyane D, Abed SN, Venugopala KN, Kalia K, Tekade RK. Reinforced electrospun nanofiber composites for drug delivery applications. J Biomed Mater Res A 2021; 109:2036-2064. [PMID: 33834610 DOI: 10.1002/jbm.a.37187] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 02/15/2021] [Accepted: 03/24/2021] [Indexed: 01/10/2023]
Abstract
Electrospun technology becomes a valuable means of fabricating functional polymeric nanofibers with distinctive morphological properties for drug delivery applications. Nanofibers are prepared from the polymer solution, which allows the direct incorporation of therapeutics such as small drug molecules, genes, and proteins by merely mixing them into the polymeric solution. Due to their biocompatibility, adhesiveness, sterility, and efficiency in delivering diverse cargoes, electrospun nanofibers have gained much attention. This review discusses the capabilities of the electrospun nanofibers in delivering different therapeutics like small molecules, genes, and proteins to their desired target site for treating various ailments. The potential of nanofibers in administering through multiple administration routes and the associated challenges has also been expounded along with a cross-talk about the commercial products of nanofibers for biomedical applications.
Collapse
Affiliation(s)
- Neelima Anup
- Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), An Institute of National Importance, Government of India, Gandhinagar, India
| | - Tejas Chavan
- Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), An Institute of National Importance, Government of India, Gandhinagar, India
| | - Shruti Chavan
- Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), An Institute of National Importance, Government of India, Gandhinagar, India
| | - Suryanarayana Polaka
- Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), An Institute of National Importance, Government of India, Gandhinagar, India
| | - Dnyaneshwar Kalyane
- Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), An Institute of National Importance, Government of India, Gandhinagar, India
| | - Sara Nidal Abed
- School of Science, Department of Chemistry, Loughborough University, Loughborough, LE11 3TU, UK
| | - Katharigatta N Venugopala
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa, Saudi Arabia.,Departments of Biotechnology and Food Technology, Durban University of Technology, Durban, South Africa
| | - Kiran Kalia
- Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), An Institute of National Importance, Government of India, Gandhinagar, India
| | - Rakesh K Tekade
- Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), An Institute of National Importance, Government of India, Gandhinagar, India
| |
Collapse
|
13
|
Yonesi M, Garcia-Nieto M, Guinea GV, Panetsos F, Pérez-Rigueiro J, González-Nieto D. Silk Fibroin: An Ancient Material for Repairing the Injured Nervous System. Pharmaceutics 2021; 13:429. [PMID: 33806846 PMCID: PMC8004633 DOI: 10.3390/pharmaceutics13030429] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 03/16/2021] [Accepted: 03/17/2021] [Indexed: 12/25/2022] Open
Abstract
Silk refers to a family of natural fibers spun by several species of invertebrates such as spiders and silkworms. In particular, silkworm silk, the silk spun by Bombyx mori larvae, has been primarily used in the textile industry and in clinical settings as a main component of sutures for tissue repairing and wound ligation. The biocompatibility, remarkable mechanical performance, controllable degradation, and the possibility of producing silk-based materials in several formats, have laid the basic principles that have triggered and extended the use of this material in regenerative medicine. The field of neural soft tissue engineering is not an exception, as it has taken advantage of the properties of silk to promote neuronal growth and nerve guidance. In addition, silk has notable intrinsic properties and the by-products derived from its degradation show anti-inflammatory and antioxidant properties. Finally, this material can be employed for the controlled release of factors and drugs, as well as for the encapsulation and implantation of exogenous stem and progenitor cells with therapeutic capacity. In this article, we review the state of the art on manufacturing methodologies and properties of fiber-based and non-fiber-based formats, as well as the application of silk-based biomaterials to neuroprotect and regenerate the damaged nervous system. We review previous studies that strategically have used silk to enhance therapeutics dealing with highly prevalent central and peripheral disorders such as stroke, Alzheimer's disease, Parkinson's disease, and peripheral trauma. Finally, we discuss previous research focused on the modification of this biomaterial, through biofunctionalization techniques and/or the creation of novel composite formulations, that aim to transform silk, beyond its natural performance, into more efficient silk-based-polymers towards the clinical arena of neuroprotection and regeneration in nervous system diseases.
Collapse
Affiliation(s)
- Mahdi Yonesi
- Center for Biomedical Technology, Universidad Politécnica de Madrid, 28223 Pozuelo de Alarcón, Spain; (M.Y.); (G.V.G.)
- Silk Biomed SL, 28260 Madrid, Spain;
| | | | - Gustavo V. Guinea
- Center for Biomedical Technology, Universidad Politécnica de Madrid, 28223 Pozuelo de Alarcón, Spain; (M.Y.); (G.V.G.)
- Silk Biomed SL, 28260 Madrid, Spain;
- Departamento de Ciencia de Materiales, ETSI Caminos, Canales y Puertos, Universidad Politécnica de Madrid, 28040 Madrid, Spain
- Biomedical Research Networking Center in Bioengineering Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
| | - Fivos Panetsos
- Silk Biomed SL, 28260 Madrid, Spain;
- Neurocomputing and Neurorobotics Research Group, Faculty of Biology and Faculty of Optics, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Innovation Group, Institute for Health Research San Carlos Clinical Hospital (IdISSC), 28040 Madrid, Spain
| | - José Pérez-Rigueiro
- Center for Biomedical Technology, Universidad Politécnica de Madrid, 28223 Pozuelo de Alarcón, Spain; (M.Y.); (G.V.G.)
- Silk Biomed SL, 28260 Madrid, Spain;
- Departamento de Ciencia de Materiales, ETSI Caminos, Canales y Puertos, Universidad Politécnica de Madrid, 28040 Madrid, Spain
- Biomedical Research Networking Center in Bioengineering Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
| | - Daniel González-Nieto
- Center for Biomedical Technology, Universidad Politécnica de Madrid, 28223 Pozuelo de Alarcón, Spain; (M.Y.); (G.V.G.)
- Silk Biomed SL, 28260 Madrid, Spain;
- Biomedical Research Networking Center in Bioengineering Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
- Departamento de Tecnología Fotónica y Bioingeniería, ETSI Telecomunicaciones, Universidad Politécnica de Madrid, 28040 Madrid, Spain
| |
Collapse
|
14
|
Lan X, Wang H, Bai J, Miao X, Lin Q, Zheng J, Ding S, Li X, Tang Y. Multidrug-loaded electrospun micro/nanofibrous membranes: Fabrication strategies, release behaviors and applications in regenerative medicine. J Control Release 2021; 330:1264-1287. [PMID: 33232749 DOI: 10.1016/j.jconrel.2020.11.036] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 11/18/2020] [Accepted: 11/18/2020] [Indexed: 01/02/2023]
Abstract
Electrospun micro/nanofibrous membranes (EFMs) have been widely investigated as local drug delivery systems. Multiple drugs can be simultaneously incorporated into one EFM to create synergistic effects, reduce side effects, and play their respective roles in the complex physiological processes of tissue regeneration and postoperative adhesion prevention. Due to the versatile electrospinning techniques, sustained and programmed release behaviors of multiple drugs could be achieved by modulating the structure of the EFMs and the location of the drugs. In this review, various multidrug incorporation approaches based on electrospinning are overviewed. In particular, the advantages and limitations of each drug incorporation technique, the methods to control drug release and the effect of one drug release on another are discussed. Then the applications of multidrug-loaded EFMs in regenerative medicine, including wound healing, bone regeneration, vascular tissue engineering, nerve regeneration, periodontal regeneration and adhesion prevention are comprehensively reviewed. Finally, the future perspectives and challenges in the research of multidrug-loaded EFMs are discussed.
Collapse
Affiliation(s)
- Xingzi Lan
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Han Wang
- State Key Laboratory of Precision Electronic Manufacturing Technology and Equipment, School of Electromechanical Engineering, Guangdong University of Technology, Guangzhou 510006, China
| | - Jianfu Bai
- State Key Laboratory of Precision Electronic Manufacturing Technology and Equipment, School of Electromechanical Engineering, Guangdong University of Technology, Guangzhou 510006, China
| | - Xiaomin Miao
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Quan Lin
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Jianpei Zheng
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Shukai Ding
- Materials Institute of Atomic and Molecular Science, ShaanXi University of Science and Technology, Xi'an 710021, China
| | - Xiaoran Li
- Innovation Center for Textile Science and Technology, Donghua University, Shanghai 200051, China
| | - Yadong Tang
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China; School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, China.
| |
Collapse
|
15
|
Millesi F, Weiss T, Mann A, Haertinger M, Semmler L, Supper P, Pils D, Naghilou A, Radtke C. Defining the regenerative effects of native spider silk fibers on primary Schwann cells, sensory neurons, and nerve-associated fibroblasts. FASEB J 2021; 35:e21196. [PMID: 33210360 PMCID: PMC7894153 DOI: 10.1096/fj.202001447r] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 09/25/2020] [Accepted: 10/30/2020] [Indexed: 01/09/2023]
Abstract
The search for a suitable material to promote regeneration after long-distance peripheral nerve defects turned the spotlight on spider silk. Nerve conduits enriched with native spider silk fibers as internal guiding structures previously demonstrated a regenerative outcome similar to autologous nerve grafts in animal studies. Nevertheless, spider silk is a natural material with associated limitations for clinical use. A promising alternative is the production of recombinant silk fibers that should mimic the outstanding properties of their native counterpart. However, in vitro data on the regenerative features that native silk fibers provide for cells involved in nerve regeneration are scarce. Thus, there is a lack of reference parameters to evaluate whether recombinant silk fiber candidates will be eligible for nerve repair in vivo. To gain insight into the regenerative effect of native spider silk, our study aims to define the behavioral response of primary Schwann cells (SCs), nerve-associated fibroblasts (FBs), and dorsal root ganglion (DRG) neurons cultured on native dragline silk from the genus Nephila and on laminin coated dishes. The established multi-color immunostaining panels together with confocal microscopy and live cell imaging enabled the analysis of cell identity, morphology, proliferation, and migration on both substrates in detail. Our findings demonstrated that native spider silk rivals laminin coating as it allowed attachment and proliferation and supported the characteristic behavior of all tested cell types. Axonal out-growth of DRG neurons occurred along longitudinally aligned SCs that formed sustained bundled structures resembling Bungner bands present in regenerating nerves. The migration of SCs along the silk fibers achieved the reported distance of regenerating axons of about 1 mm per day, but lacked directionality. Furthermore, rFBs significantly reduced the velocity of rSCs in co-cultures on silk fibers. In summary, this study (a) reveals features recombinant silk must possess and what modifications or combinations could be useful for enhanced nerve repair and (b) provides assays to evaluate the regenerative performance of silk fibers in vitro before being applied as internal guiding structure in nerve conduits in vivo.
Collapse
Affiliation(s)
- Flavia Millesi
- Research Laboratory of the Division of Plastic and Reconstructive SurgeryDepartment of SurgeryMedical University of ViennaViennaAustria
- Austrian Cluster for Tissue RegenerationViennaAustria
| | - Tamara Weiss
- Research Laboratory of the Division of Plastic and Reconstructive SurgeryDepartment of SurgeryMedical University of ViennaViennaAustria
- Austrian Cluster for Tissue RegenerationViennaAustria
| | - Anda Mann
- Research Laboratory of the Division of Plastic and Reconstructive SurgeryDepartment of SurgeryMedical University of ViennaViennaAustria
| | - Maximilian Haertinger
- Research Laboratory of the Division of Plastic and Reconstructive SurgeryDepartment of SurgeryMedical University of ViennaViennaAustria
- Austrian Cluster for Tissue RegenerationViennaAustria
| | - Lorenz Semmler
- Research Laboratory of the Division of Plastic and Reconstructive SurgeryDepartment of SurgeryMedical University of ViennaViennaAustria
| | - Paul Supper
- Research Laboratory of the Division of Plastic and Reconstructive SurgeryDepartment of SurgeryMedical University of ViennaViennaAustria
| | - Dietmar Pils
- Division of General SurgeryDepartment of SurgeryComprehensive Cancer Center ViennaMedical University of ViennaViennaAustria
| | - Aida Naghilou
- Research Laboratory of the Division of Plastic and Reconstructive SurgeryDepartment of SurgeryMedical University of ViennaViennaAustria
| | - Christine Radtke
- Research Laboratory of the Division of Plastic and Reconstructive SurgeryDepartment of SurgeryMedical University of ViennaViennaAustria
- Austrian Cluster for Tissue RegenerationViennaAustria
- Division of Plastic and Reconstructive SurgeryDepartment of SurgeryMedical University of ViennaViennaAustria
| |
Collapse
|
16
|
Meena P, Kakkar A, Kumar M, Khatri N, Nagar RK, Singh A, Malhotra P, Shukla M, Saraswat SK, Srivastava S, Datt R, Pandey S. Advances and clinical challenges for translating nerve conduit technology from bench to bed side for peripheral nerve repair. Cell Tissue Res 2020; 383:617-644. [PMID: 33201351 DOI: 10.1007/s00441-020-03301-x] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 09/14/2020] [Indexed: 12/19/2022]
Abstract
Injuries to the peripheral nervous system remain a large-scale clinical problem. These injuries often lead to loss of motor and/or sensory function that significantly affects patients' quality of life. The current neurosurgical approach for peripheral nerve repair involves autologous nerve transplantation, which often leads to clinical complications. The most pressing need is to increase the regenerative capacity of existing tubular constructs in the repair of large nerve gaps through development of tissue-engineered approaches that can surpass the performance of autografts. To fully realize the clinical potential of nerve conduit technology, there is a need to reconsider design strategies, biomaterial selection, fabrication techniques and the various potential modifications to optimize a conduit microenvironment that can best mimic the natural process of regeneration. In recent years, a significant progress has been made in the designing and functionality of bioengineered nerve conduits to bridge long peripheral nerve gaps in various animal models. However, translation of this work from lab to commercial scale has not been achieve. The current review summarizes recent advances in the development of tissue engineered nerve guidance conduits (NGCs) with regard to choice of material, novel fabrication methods, surface modifications and regenerative cues such as stem cells and growth factors to improve regeneration performance. Also, the current clinical potential and future perspectives to achieve therapeutic benefits of NGCs will be discussed in context of peripheral nerve regeneration.
Collapse
Affiliation(s)
- Poonam Meena
- Department of Life Sciences, Datt Mediproducts Pvt. Ltd., Roz Ka Meo Industrial Area, District Mewat, Nuh, 122103, District Haryana, India
| | - Anupama Kakkar
- Department of Life Sciences, Datt Mediproducts Pvt. Ltd., Roz Ka Meo Industrial Area, District Mewat, Nuh, 122103, District Haryana, India
| | - Mukesh Kumar
- Department of Life Sciences, Datt Mediproducts Pvt. Ltd., Roz Ka Meo Industrial Area, District Mewat, Nuh, 122103, District Haryana, India
| | - Nitin Khatri
- Department of Life Sciences, Datt Mediproducts Pvt. Ltd., Roz Ka Meo Industrial Area, District Mewat, Nuh, 122103, District Haryana, India
| | - Rakesh Kumar Nagar
- Department of Life Sciences, Datt Mediproducts Pvt. Ltd., Roz Ka Meo Industrial Area, District Mewat, Nuh, 122103, District Haryana, India
| | - Aarti Singh
- Department of Life Sciences, Datt Mediproducts Pvt. Ltd., Roz Ka Meo Industrial Area, District Mewat, Nuh, 122103, District Haryana, India
| | - Poonam Malhotra
- Department of Life Sciences, Datt Mediproducts Pvt. Ltd., Roz Ka Meo Industrial Area, District Mewat, Nuh, 122103, District Haryana, India
| | - Manish Shukla
- Department of Life Sciences, Datt Mediproducts Pvt. Ltd., Roz Ka Meo Industrial Area, District Mewat, Nuh, 122103, District Haryana, India
| | - Sumit Kumar Saraswat
- Department of Life Sciences, Datt Mediproducts Pvt. Ltd., Roz Ka Meo Industrial Area, District Mewat, Nuh, 122103, District Haryana, India
| | - Supriya Srivastava
- Department of Life Sciences, Datt Mediproducts Pvt. Ltd., Roz Ka Meo Industrial Area, District Mewat, Nuh, 122103, District Haryana, India
| | - Rajan Datt
- Department of Life Sciences, Datt Mediproducts Pvt. Ltd., Roz Ka Meo Industrial Area, District Mewat, Nuh, 122103, District Haryana, India
| | - Siddharth Pandey
- Department of Life Sciences, Datt Mediproducts Pvt. Ltd., Roz Ka Meo Industrial Area, District Mewat, Nuh, 122103, District Haryana, India.
| |
Collapse
|
17
|
Mercado J, Pérez-Rigueiro J, González-Nieto D, Lozano-Picazo P, López P, Panetsos F, Elices M, Gañán-Calvo AM, Guinea GV, Ramos-Gómez M. Regenerated Silk Fibers Obtained by Straining Flow Spinning for Guiding Axonal Elongation in Primary Cortical Neurons. ACS Biomater Sci Eng 2020; 6:6842-6852. [PMID: 33320622 DOI: 10.1021/acsbiomaterials.0c00985] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The recovery of injured nervous tissue, one of the main goals for regenerative therapeutic approaches, is often hindered by the limited axonal regeneration ability of the central nervous system (CNS). In this regard, the identification of scaffolds that support the reconstruction of functional neuronal tissues and guide the alignment of regenerating neurons is a major challenge in tissue engineering. Ideally, the usage of such scaffolds would promote and guide the axonal growth, a crucial phase for the restoration of neuronal connections and, consequently, the nerve function. Among the materials proposed as scaffolds for CNS regeneration, silk has been used to exploit its outstanding features as a biomaterial to promote axonal regeneration. In this study, we explore, for the first time, the possibility of using high-performance regenerated silk fibers obtained by straining flow spinning (SFS) to serve as scaffolds for inducing and guiding the axonal growth. It is shown that SFS fibers promote the spontaneous organization of dissociated cortical primary cells into highly interconnected cellular spheroid-like tissue formations. Neuronal projections (i.e., axons) from these cellular spheroids span hundreds of microns along the SFS fibers that act as guides and allow the connection of distant spheroids. In addition, it is also shown that SFS fibers serve as scaffolds for neuronal migration covering short and long distances. As a consequence, the usage of high-performance SFS fibers appears as a promising basis for the development of novel therapies, leading to directed axonal regeneration.
Collapse
Affiliation(s)
- Juan Mercado
- Center for Biomedical Technology (CTB), Universidad Politécnica de Madrid, Pozuelo de Alarcón, 28223 Madrid, Spain
| | - José Pérez-Rigueiro
- Center for Biomedical Technology (CTB), Universidad Politécnica de Madrid, Pozuelo de Alarcón, 28223 Madrid, Spain.,Departamento de Ciencia de Materiales, ETSI Caminos, Canales y Puertos, Universidad Politécnica de Madrid, 28040 Madrid, Spain.,Biomedical Research Networking Center in Bioengineering Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
| | - Daniel González-Nieto
- Center for Biomedical Technology (CTB), Universidad Politécnica de Madrid, Pozuelo de Alarcón, 28223 Madrid, Spain.,Biomedical Research Networking Center in Bioengineering Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain.,Departamento de Tecnología Fotónica y Bioingeniería, ETSI Telecomunicaciones, Universidad Politécnica de Madrid, 28040 Madrid, Spain
| | - Paloma Lozano-Picazo
- Center for Biomedical Technology (CTB), Universidad Politécnica de Madrid, Pozuelo de Alarcón, 28223 Madrid, Spain.,Departamento de Ciencia de Materiales, ETSI Caminos, Canales y Puertos, Universidad Politécnica de Madrid, 28040 Madrid, Spain
| | - Patricia López
- Center for Biomedical Technology (CTB), Universidad Politécnica de Madrid, Pozuelo de Alarcón, 28223 Madrid, Spain.,Departamento de Ciencia de Materiales, ETSI Caminos, Canales y Puertos, Universidad Politécnica de Madrid, 28040 Madrid, Spain
| | - Fivos Panetsos
- Neurocomputing and Neurorobotics Research Group, Faculty of Biology and Faculty of Optics, Universidad Complutense de Madrid, 28040 Madrid, Spain.,Brain Plasticity Group, Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
| | - Manuel Elices
- Departamento de Ciencia de Materiales, ETSI Caminos, Canales y Puertos, Universidad Politécnica de Madrid, 28040 Madrid, Spain
| | - Alfonso M Gañán-Calvo
- Escuela Técnica Superior de Ingenieros, Universidad de Sevilla, 41092 Sevilla, Spain
| | - Gustavo V Guinea
- Center for Biomedical Technology (CTB), Universidad Politécnica de Madrid, Pozuelo de Alarcón, 28223 Madrid, Spain.,Departamento de Ciencia de Materiales, ETSI Caminos, Canales y Puertos, Universidad Politécnica de Madrid, 28040 Madrid, Spain.,Biomedical Research Networking Center in Bioengineering Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
| | - Milagros Ramos-Gómez
- Center for Biomedical Technology (CTB), Universidad Politécnica de Madrid, Pozuelo de Alarcón, 28223 Madrid, Spain.,Biomedical Research Networking Center in Bioengineering Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain.,Departamento de Tecnología Fotónica y Bioingeniería, ETSI Telecomunicaciones, Universidad Politécnica de Madrid, 28040 Madrid, Spain
| |
Collapse
|
18
|
Li R, Li DH, Zhang HY, Wang J, Li XK, Xiao J. Growth factors-based therapeutic strategies and their underlying signaling mechanisms for peripheral nerve regeneration. Acta Pharmacol Sin 2020; 41:1289-1300. [PMID: 32123299 PMCID: PMC7608263 DOI: 10.1038/s41401-019-0338-1] [Citation(s) in RCA: 114] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 11/20/2019] [Indexed: 12/21/2022] Open
Abstract
Peripheral nerve injury (PNI), one of the most common concerns following trauma, can result in a significant loss of sensory or motor function. Restoration of the injured nerves requires a complex cellular and molecular response to rebuild the functional axons so that they can accurately connect with their original targets. However, there is no optimized therapy for complete recovery after PNI. Supplementation with exogenous growth factors (GFs) is an emerging and versatile therapeutic strategy for promoting nerve regeneration and functional recovery. GFs activate the downstream targets of various signaling cascades through binding with their corresponding receptors to exert their multiple effects on neurorestoration and tissue regeneration. However, the simple administration of GFs is insufficient for reconstructing PNI due to their short half‑life and rapid deactivation in body fluids. To overcome these shortcomings, several nerve conduits derived from biological tissue or synthetic materials have been developed. Their good biocompatibility and biofunctionality made them a suitable vehicle for the delivery of multiple GFs to support peripheral nerve regeneration. After repairing nerve defects, the controlled release of GFs from the conduit structures is able to continuously improve axonal regeneration and functional outcome. Thus, therapies with growth factor (GF) delivery systems have received increasing attention in recent years. Here, we mainly review the therapeutic capacity of GFs and their incorporation into nerve guides for repairing PNI. In addition, the possible receptors and signaling mechanisms of the GF family exerting their biological effects are also emphasized.
Collapse
Affiliation(s)
- Rui Li
- Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
- School of Chemistry, Sun Yat-sen University, Guangzhou, 510275, China
| | - Duo-Hui Li
- Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Hong-Yu Zhang
- Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Jian Wang
- Department of Peripheral Neurosurgery, The First Affiliated Hospital, Wenzhou, Medical University, Wenzhou, 325000, China
| | - Xiao-Kun Li
- Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China.
| | - Jian Xiao
- Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China.
- Department of Peripheral Neurosurgery, The First Affiliated Hospital, Wenzhou, Medical University, Wenzhou, 325000, China.
| |
Collapse
|
19
|
You R, Zhang Q, Li X, Yan S, Luo Z, Qu J, Li M. Multichannel Bioactive Silk Nanofiber Conduits Direct and Enhance Axonal Regeneration after Spinal Cord Injury. ACS Biomater Sci Eng 2020; 6:4677-4686. [PMID: 33455191 DOI: 10.1021/acsbiomaterials.0c00698] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
After a spinal cord injury, axonal regeneration over long distances is challenging due to the lack of physical guidance cues and bioactive signals. In this study, a multichannel bioactive silk fibroin nanofiber conduit was fabricated to improve spinal cord injury repair by enhancing axonal regeneration. The conduit was composed of longitudinally oriented silk fibroin nanofibers and then functionalized with laminin. In vitro, the bioactive conduits could promote neuron-like development and directional neurite extension of PC12 cells by providing a bioactive stimulus and physical guidance. In a spinal cord injury model in Sprague-Dawley rats, the biofunctionalized conduits displayed superior integration with the host tissue due to enhanced cell infiltration and tissue ingrowth. The glial scar was significantly reduced, allowing axonal ingrowth along with the channel direction. Compared to a single-channel conduit, the multichannel conduit improved spinal cord regeneration by boosting tissue ingrowth and axonal regeneration, indicating that the conduit architectures play critical roles in spinal cord regeneration. These silk fibroin conduits, along with the multichannel architecture, nanoscale cues, and the ability to bind bioactive compounds, represent promising candidates for spinal cord regeneration.
Collapse
Affiliation(s)
- Renchuan You
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou 215123, China.,State Key Laboratory of New Textile Materials and Advanced Processing Technologies, School of Textile Science and Engineering, Wuhan Textile University, Wuhan 430200, China
| | - Qiang Zhang
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou 215123, China.,State Key Laboratory of New Textile Materials and Advanced Processing Technologies, School of Textile Science and Engineering, Wuhan Textile University, Wuhan 430200, China
| | - Xiufang Li
- State Key Laboratory of New Textile Materials and Advanced Processing Technologies, School of Textile Science and Engineering, Wuhan Textile University, Wuhan 430200, China
| | - Shuqin Yan
- State Key Laboratory of New Textile Materials and Advanced Processing Technologies, School of Textile Science and Engineering, Wuhan Textile University, Wuhan 430200, China
| | - Zuwei Luo
- State Key Laboratory of New Textile Materials and Advanced Processing Technologies, School of Textile Science and Engineering, Wuhan Textile University, Wuhan 430200, China
| | - Jing Qu
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou 215123, China
| | - Mingzhong Li
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou 215123, China
| |
Collapse
|
20
|
Gough CR, Rivera-Galletti A, Cowan DA, Salas-de la Cruz D, Hu X. Protein and Polysaccharide-Based Fiber Materials Generated from Ionic Liquids: A Review. Molecules 2020; 25:E3362. [PMID: 32722182 PMCID: PMC7435976 DOI: 10.3390/molecules25153362] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 07/19/2020] [Accepted: 07/24/2020] [Indexed: 02/06/2023] Open
Abstract
Natural biomacromolecules such as structural proteins and polysaccharides are composed of the basic building blocks of life: amino acids and carbohydrates. Understanding their molecular structure, self-assembly and interaction in solvents such as ionic liquids (ILs) is critical for unleashing a flora of new materials, revolutionizing the way we fabricate multi-structural and multi-functional systems with tunable physicochemical properties. Ionic liquids are superior to organic solvents because they do not produce unwanted by-products and are considered green substitutes because of their reusability. In addition, they will significantly improve the miscibility of biopolymers with other materials while maintaining the mechanical properties of the biopolymer in the final product. Understanding and controlling the physicochemical properties of biopolymers in ionic liquids matrices will be crucial for progress leading to the ability to fabricate robust multi-level structural 1D fiber materials. It will also help to predict the relationship between fiber conformation and protein secondary structures or carbohydrate crystallinity, thus creating potential applications for cell growth signaling, ionic conductivity, liquid diffusion and thermal conductivity, and several applications in biomedicine and environmental science. This will also enable the regeneration of biopolymer composite fiber materials with useful functionalities and customizable options critical for additive manufacturing. The specific capabilities of these fiber materials have been shown to vary based on their fabrication methods including electrospinning and post-treatments. This review serves to provide basic knowledge of these commonly utilized protein and polysaccharide biopolymers and their fiber fabrication methods from various ionic liquids, as well as the effect of post-treatments on these fiber materials and their applications in biomedical and pharmaceutical research, wound healing, environmental filters and sustainable and green chemistry research.
Collapse
Affiliation(s)
- Christopher R. Gough
- Department of Physics and Astronomy, Rowan University, Glassboro, NJ 08028, USA; (C.R.G.); (A.R.-G.); (D.A.C.)
- Department of Chemistry and Biochemistry, Rowan University, Glassboro, NJ 08028, USA
| | - Ashley Rivera-Galletti
- Department of Physics and Astronomy, Rowan University, Glassboro, NJ 08028, USA; (C.R.G.); (A.R.-G.); (D.A.C.)
- Department of Chemistry and Biochemistry, Rowan University, Glassboro, NJ 08028, USA
| | - Darrel A. Cowan
- Department of Physics and Astronomy, Rowan University, Glassboro, NJ 08028, USA; (C.R.G.); (A.R.-G.); (D.A.C.)
- Department of Chemistry and Biochemistry, Rowan University, Glassboro, NJ 08028, USA
| | - David Salas-de la Cruz
- Department of Chemistry, and Center for Computational and Integrative Biology, Camden, NJ 08102, USA;
| | - Xiao Hu
- Department of Physics and Astronomy, Rowan University, Glassboro, NJ 08028, USA; (C.R.G.); (A.R.-G.); (D.A.C.)
- Department of Biomedical Engineering, Rowan University, Glassboro, NJ 08028, USA
- Department of Molecular and Cellular Biosciences, Rowan University, Glassboro, NJ 08028, USA
| |
Collapse
|
21
|
Farokhi M, Mottaghitalab F, Reis RL, Ramakrishna S, Kundu SC. Functionalized silk fibroin nanofibers as drug carriers: Advantages and challenges. J Control Release 2020; 321:324-347. [DOI: 10.1016/j.jconrel.2020.02.022] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 02/11/2020] [Accepted: 02/11/2020] [Indexed: 12/13/2022]
|
22
|
Jang SR, Kim JI, Park CH, Kim CS. The controlled design of electrospun PCL/silk/quercetin fibrous tubular scaffold using a modified wound coil collector and L-shaped ground design for neural repair. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 111:110776. [PMID: 32279813 DOI: 10.1016/j.msec.2020.110776] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 02/18/2020] [Accepted: 02/24/2020] [Indexed: 12/28/2022]
Abstract
Asymmetrically porous and aligned fibrous tubular conduit with selective permeability as a biomimetic neural scaffold was manufactured using polycaprolactone (PCL), silk, and quercetin by a modified electrospinning method. The outer surface of the randomly oriented fibrous scaffold had microscale pores that could prevent fibrous tissue invasion (FTI), but could permeate neurotrophic factors, nutrients, and oxygen. The inner surface of the aligned fibrous scaffold can be favorable for neurite outgrowth, because of their superior neural cell attachment, migration, and directional growth. In vitro and in vivo studies have demonstrated the therapeutic effect of Quercetin, a ubiquitous flavonoid widely distributed in plants, on neuropathy, by modulating the expression of NRF-2-dependent antioxidant responsive elements. In this study, the controlled inner and outer surface geometry of the 0.5, 1.0, and 2.0 wt% quercetin-containing electrospun PCL/silk fibrous tubular scaffold fabricated via a modified wound coil collector and L-shaped ground design (WCC-LG) was characterized by FE-SEM, TEM, FFT, FT-IR, and XRD. In addition, two types of neural cell lines, PC12 and S42, were used to evaluate the cell proliferation rate of the different amount of quercetin-loaded PCL/silk tubular scaffolds.
Collapse
Affiliation(s)
- Se Rim Jang
- Department of Bionanosystem Engineering, Graduate School, Jeonbuk National University, Jeonju 561-756, Republic of Korea
| | - Jeong In Kim
- Department of Bionanosystem Engineering, Graduate School, Jeonbuk National University, Jeonju 561-756, Republic of Korea
| | - Chan Hee Park
- Department of Bionanosystem Engineering, Graduate School, Jeonbuk National University, Jeonju 561-756, Republic of Korea; Division of Mechanical Design Engineering, College of Engineering, Jeonbuk National University, Jeonju 561-756, Republic of Korea.
| | - Cheol Sang Kim
- Department of Bionanosystem Engineering, Graduate School, Jeonbuk National University, Jeonju 561-756, Republic of Korea; Division of Mechanical Design Engineering, College of Engineering, Jeonbuk National University, Jeonju 561-756, Republic of Korea.
| |
Collapse
|
23
|
Ikegami Y, Ijima H. Development of heparin-conjugated nanofibers and a novel biological signal by immobilized growth factors for peripheral nerve regeneration. J Biosci Bioeng 2019; 129:354-362. [PMID: 31601468 DOI: 10.1016/j.jbiosc.2019.09.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 08/12/2019] [Accepted: 09/05/2019] [Indexed: 11/29/2022]
Abstract
Aligned fibers have been used as a scaffold of nerve guidance conduit owing to their guiding function of neural cells for peripheral nerve regeneration. However, the recovery performance of nerve guidance conduits using aligned fibrous scaffold is insufficient, and further improvements in scaffold function is required for promoting regeneration. In this study, we developed aligned heparin-conjugated fibers and supplied a biological signal to neural cells by the growth factors immobilized through heparin. Results indicated that neural model cells (PC12 cells) were cultured well on the scaffold without inhibiting cell adhesion by heparin conjugation and exhibited more vigorous cell proliferation than in a heparin-free condition. The cells extended their neurites along the fiber direction. Furthermore, PC12 cells on the heparin-conjugated fibrous scaffold pre-exposed to a nerve growth factor solution sprouted more neurites compared to those of heparin-free condition. These results verified that our scaffold exhibited high biocompatibility to neural cells and could maintain an effective local concentration of growth factors on the scaffold surface. Therefore, aligned heparin-conjugated fibers are promising scaffolds of nerve guidance conduits for promoting peripheral nerve regeneration by the combinatorial effect of topological and biological signals.
Collapse
Affiliation(s)
- Yasuhiro Ikegami
- Department of Chemical Engineering, Faculty of Engineering, Graduate School, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Hiroyuki Ijima
- Department of Chemical Engineering, Faculty of Engineering, Graduate School, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan.
| |
Collapse
|
24
|
Pawar K, Welzel G, Haynl C, Schuster S, Scheibel T. Recombinant Spider Silk and Collagen-Based Nerve Guidance Conduits Support Neuronal Cell Differentiation and Functionality in Vitro. ACS APPLIED BIO MATERIALS 2019; 2:4872-4880. [DOI: 10.1021/acsabm.9b00628] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Kiran Pawar
- Department for Biomaterials, University of Bayreuth, Prof.-Rüdiger-Bormann-Strasse 1, 95447 Bayreuth, Germany
| | | | - Christian Haynl
- Department for Biomaterials, University of Bayreuth, Prof.-Rüdiger-Bormann-Strasse 1, 95447 Bayreuth, Germany
| | | | - Thomas Scheibel
- Department for Biomaterials, University of Bayreuth, Prof.-Rüdiger-Bormann-Strasse 1, 95447 Bayreuth, Germany
| |
Collapse
|
25
|
Porzionato A, Barbon S, Stocco E, Dalzoppo D, Contran M, De Rose E, Parnigotto PP, Macchi V, Grandi C, De Caro R. Development of Oxidized Polyvinyl Alcohol-Based Nerve Conduits Coupled with the Ciliary Neurotrophic Factor. MATERIALS (BASEL, SWITZERLAND) 2019; 12:E1996. [PMID: 31234386 PMCID: PMC6631399 DOI: 10.3390/ma12121996] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 06/09/2019] [Accepted: 06/19/2019] [Indexed: 12/14/2022]
Abstract
Functionalized synthetic conduits represent a promising strategy to enhance peripheral nerve regeneration by guiding axon growth while delivering therapeutic neurotrophic factors. In this work, hollow nerve conduits made of polyvinyl alcohol partially oxidized with bromine (OxPVA_Br2) and potassium permanganate (OxPVA_KMnO4) were investigated for their structural/biological properties and ability to absorb/release the ciliary neurotrophic factor (CNTF). Chemical oxidation enhanced water uptake capacity of the polymer, with maximum swelling index of 60.5% ± 2.5%, 71.3% ± 3.6% and 19.5% ± 4.0% for OxPVA_Br2, OxPVA_KMnO4 and PVA, respectively. Accordingly, hydrogel porosity increased from 15.27% ± 1.16% (PVA) to 62.71% ± 8.63% (OxPVA_Br2) or 77.50% ± 3.39% (OxPVA_KMnO4) after oxidation. Besides proving that oxidized PVA conduits exhibited mechanical resistance and a suture holding ability, they did not exert a cytotoxic effect on SH-SY5Y and Schwann cells and biodegraded over time when subjected to enzymatic digestion, functionalization with CNTF was performed. Interestingly, higher amounts of neurotrophic factor were detected in the lumen of OxPVA_Br2 (0.22 ± 0.029 µg) and OxPVA_KMnO4 (0.29 ± 0.033 µg) guides rather than PVA (0.11 ± 0.021 µg) tubular scaffolds. In conclusion, we defined a promising technology to obtain drug delivery conduits based on functionalizable oxidized PVA hydrogels.
Collapse
Affiliation(s)
- Andrea Porzionato
- Department of Neurosciences, Section of Human Anatomy, University of Padova, Via A. Gabelli 65, 35121 Padova, Italy.
- L.i.f.e.L.a.b. Program, Consorzio per la Ricerca Sanitaria (CORIS), Veneto Region, Via N. Giustiniani 2, 35128 Padova, Italy.
| | - Silvia Barbon
- Department of Neurosciences, Section of Human Anatomy, University of Padova, Via A. Gabelli 65, 35121 Padova, Italy.
- L.i.f.e.L.a.b. Program, Consorzio per la Ricerca Sanitaria (CORIS), Veneto Region, Via N. Giustiniani 2, 35128 Padova, Italy.
| | - Elena Stocco
- Department of Neurosciences, Section of Human Anatomy, University of Padova, Via A. Gabelli 65, 35121 Padova, Italy.
- L.i.f.e.L.a.b. Program, Consorzio per la Ricerca Sanitaria (CORIS), Veneto Region, Via N. Giustiniani 2, 35128 Padova, Italy.
| | - Daniele Dalzoppo
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35128 Padova, Italy.
| | - Martina Contran
- Department of Neurosciences, Section of Human Anatomy, University of Padova, Via A. Gabelli 65, 35121 Padova, Italy.
| | - Enrico De Rose
- Department of Neurosciences, Section of Human Anatomy, University of Padova, Via A. Gabelli 65, 35121 Padova, Italy.
| | - Pier Paolo Parnigotto
- Foundation for Biology and Regenerative Medicine, Tissue Engineering and Signaling (T.E.S.) Onlus, 35030 Padua, Italy.
| | - Veronica Macchi
- Department of Neurosciences, Section of Human Anatomy, University of Padova, Via A. Gabelli 65, 35121 Padova, Italy.
- L.i.f.e.L.a.b. Program, Consorzio per la Ricerca Sanitaria (CORIS), Veneto Region, Via N. Giustiniani 2, 35128 Padova, Italy.
| | - Claudio Grandi
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35128 Padova, Italy.
| | - Raffaele De Caro
- Department of Neurosciences, Section of Human Anatomy, University of Padova, Via A. Gabelli 65, 35121 Padova, Italy.
- L.i.f.e.L.a.b. Program, Consorzio per la Ricerca Sanitaria (CORIS), Veneto Region, Via N. Giustiniani 2, 35128 Padova, Italy.
| |
Collapse
|
26
|
Electrospun polymer micro/nanofibers as pharmaceutical repositories for healthcare. J Control Release 2019; 302:19-41. [PMID: 30922946 DOI: 10.1016/j.jconrel.2019.03.020] [Citation(s) in RCA: 180] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 03/22/2019] [Accepted: 03/23/2019] [Indexed: 12/19/2022]
|
27
|
Mehrotra S, Chouhan D, Konwarh R, Kumar M, Jadi PK, Mandal BB. Comprehensive Review on Silk at Nanoscale for Regenerative Medicine and Allied Applications. ACS Biomater Sci Eng 2019; 5:2054-2078. [PMID: 33405710 DOI: 10.1021/acsbiomaterials.8b01560] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Shreya Mehrotra
- Biomaterial and Tissue Engineering Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati−781039, Assam, India
| | - Dimple Chouhan
- Biomaterial and Tissue Engineering Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati−781039, Assam, India
| | - Rocktotpal Konwarh
- Biotechnology Department, Addis Ababa Science and Technology University, Addis Ababa−16417, Ethiopia
| | - Manishekhar Kumar
- Biomaterial and Tissue Engineering Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati−781039, Assam, India
| | - Praveen Kumar Jadi
- Biomaterial and Tissue Engineering Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati−781039, Assam, India
| | - Biman B. Mandal
- Biomaterial and Tissue Engineering Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati−781039, Assam, India
| |
Collapse
|
28
|
Yen CM, Shen CC, Yang YC, Liu BS, Lee HT, Sheu ML, Tsai MH, Cheng WY. Novel electrospun poly(ε-caprolactone)/type I collagen nanofiber conduits for repair of peripheral nerve injury. Neural Regen Res 2019; 14:1617-1625. [PMID: 31089062 PMCID: PMC6557087 DOI: 10.4103/1673-5374.255997] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Recent studies have shown the potential of artificially synthesized conduits in the repair of peripheral nerve injury. Natural biopolymers have received much attention because of their biocompatibility. To investigate the effects of novel electrospun absorbable poly(ε-caprolactone)/type I collagen nanofiber conduits (biopolymer nanofiber conduits) on the repair of peripheral nerve injury, we bridged 10-mm-long sciatic nerve defects with electrospun absorbable biopolymer nanofiber conduits, poly(ε-caprolactone) or silicone conduits in Sprague-Dawley rats. Rat neurologica1 function was weekly evaluated using sciatic function index within 8 weeks after repair. Eight weeks after repair, sciatic nerve myelin sheaths and axon morphology were observed by osmium tetroxide staining, hematoxylin-eosin staining, and transmission electron microscopy. S-100 (Schwann cell marker) and CD4 (inflammatory marker) immunoreactivities in sciatic nerve were detected by immunohistochemistry. In rats subjected to repair with electrospun absorbable biopolymer nanofiber conduits, no serious inflammatory reactions were observed in rat hind limbs, the morphology of myelin sheaths in the injured sciatic nerve was close to normal. CD4 immunoreactivity was obviously weaker in rats subjected to repair with electrospun absorbable biopolymer nanofiber conduits than in those subjected to repair with poly(ε-caprolactone) or silicone. Rats subjected to repair with electrospun absorbable biopolymer nanofiber conduits tended to have greater sciatic nerve function recovery than those receiving poly(ε-caprolactone) or silicone repair. These results suggest that electrospun absorbable poly(ε-caprolactone)/type I collagen nanofiber conduits have the potential of repairing sciatic nerve defects and exhibit good biocompatibility. All experimental procedures were approved by Institutional Animal Care and Use Committee of Taichung Veteran General Hospital, Taiwan, China (La-1031218) on October 2, 2014.
Collapse
Affiliation(s)
- Chun-Ming Yen
- Department of Neurosurgery, Neurological Institute, Taichung Veterans General Hospital; Ph.D. Program in Translational Medicine, National Chung Hsing University, Taichung, Taiwan, China
| | - Chiung-Chyi Shen
- Department of Neurosurgery, Neurological Institute, Taichung Veterans General Hospital; Department of Physical Therapy, Hungkuang University; Basic Medical Education Center, Central Taiwan University of Science and Technology, Taichung, Taiwan, China
| | - Yi-Chin Yang
- Department of Neurosurgery, Neurological Institute, Taichung Veterans General Hospital, Taichung, Taiwan, China
| | - Bai-Shuan Liu
- Department of Medical Imaging and Radiological Sciences, Central Taiwan University of Science and Technology, Taichung, Taiwan, China
| | - Hsu-Tung Lee
- Department of Neurosurgery, Neurological Institute, Taichung Veterans General Hospital, Taichung, Taiwan, China
| | - Meei-Ling Sheu
- Institute of Biomedical Sciences, National Chung Hsing University; Department of Medical Research, Taichung Veterans General Hospital; Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung, Taiwan, China
| | - Meng-Hsiun Tsai
- Department of Management Information System, National Chung Hsing University, Taichung, Taiwan, China
| | - Wen-Yu Cheng
- Department of Neurosurgery, Neurological Institute, Taichung Veterans General Hospital; Department of Physical Therapy, Hungkuang University, Taichung, Taiwan, China
| |
Collapse
|
29
|
Wang L, Song D, Zhang X, Ding Z, Kong X, Lu Q, Kaplan DL. Silk-Graphene Hybrid Hydrogels with Multiple Cues to Induce Nerve Cell Behavior. ACS Biomater Sci Eng 2018; 5:613-622. [PMID: 33405825 DOI: 10.1021/acsbiomaterials.8b01481] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Cell behavior is dependent in part on chemical and physical cues from the extracellular matrix. Although the influence of various cues on cell behavior has been studied, challenges remain to incorporate multiple cues to matrix systems to optimize and control cell outcomes. Here, aligned silk fibroin (SF)-graphene hydrogels with preferable stiffness were developed through arranging SF nanofibers and SF-modified graphene sheets under an electric field. Different signals, such as bioactive graphene, nanofibrous structure, aligned topography, and mechanical stiffness, were tailored into the hydrogel system, providing niches for nerve cell responses. The desired adhesion, proliferation, differentiation, extensio,n and growth factor secretion of multiple nerve-related cells was achieved on these hydrogels, suggesting strong synergistic action through the combination of different cues. Based on the fabrication strategy, our present study provides a useful materials engineering platform for revealing cooperative influences of different signals on nerve cell behavior, to help in the understanding of cell-biomaterial interactions, with potential toward studies related to nerve regeneration.
Collapse
Affiliation(s)
- Lili Wang
- National Engineering Laboratory for Modern Silk & Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou, Jiangsu 215123, People's Republic of China
| | - Dawei Song
- Tai'an City Central Hospital, Taian, 271000, People's Republic of China
| | - Xiaoyi Zhang
- National Engineering Laboratory for Modern Silk & Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou, Jiangsu 215123, People's Republic of China
| | - Zhaozhao Ding
- National Engineering Laboratory for Modern Silk & Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou, Jiangsu 215123, People's Republic of China
| | - Xiangdong Kong
- College of Materials and Textiles, Zhejiang Sci-Tech University, Hangzhou 310018, People's Republic of China
| | - Qiang Lu
- National Engineering Laboratory for Modern Silk & Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou, Jiangsu 215123, People's Republic of China
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, United States
| |
Collapse
|
30
|
Belanger K, Schlatter G, Hébraud A, Marin F, Testelin S, Dakpé S, Devauchelle B, Egles C. A multi-layered nerve guidance conduit design adapted to facilitate surgical implantation. Health Sci Rep 2018; 1:e86. [PMID: 30623049 PMCID: PMC6295612 DOI: 10.1002/hsr2.86] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2017] [Revised: 07/16/2018] [Accepted: 07/18/2018] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND AND AIMS The gold standard procedure after a severe nerve injury is the nerve autograft, yet this technique has drawbacks. In recent years, progress has been made in the development of artificial nerve guides to replace the autograft, but no device has been able to demonstrate superiority. The present study introduces an adaptable foundation design for peripheral nerve regeneration. METHODS Silk fibroin was electrospun, creating a tri-layered material with aligned fiber surfaces and a randomly deposited fiber interior. This material was rolled into a micro-channeled conduit, which was then enveloped by a jacket layer of the same tri-layered material. RESULTS The proposed implant design succeeds in incorporating various desirable aspects of synthetic nerve guides, while facilitating the surgical implantation process for medical application. The aligned fiber surfaces of the conduit support axon guidance, while the tri-layered architecture improves its structural integrity compared with a fully aligned fiber material. Moreover, the jacket layer creates a small niche on each end which facilitates surgical implantation. An in vivo study in rats showed that nerve regeneration using this device was comparable to results after direct suture. CONCLUSION This proof-of-principle study, therefore, advances the development of tissue engineered nerve grafts by creating an optimized guidance conduit design capable of successful nerve regeneration.
Collapse
Affiliation(s)
- Kayla Belanger
- UMR 7338, Biomécanique et Bioingénierie, Centre de recherches de RoyallieuSorbonne Universités, Université de Technologie de Compiègne, CNRSCompiègne cedexFrance
| | - Guy Schlatter
- ICPEES Institut de Chimie et Procédés pour l'Energie, l'Environnement et la Santé, UMR 7515, CNRSUniversité de StrasbourgStrasbourg cedexFrance
| | - Anne Hébraud
- ICPEES Institut de Chimie et Procédés pour l'Energie, l'Environnement et la Santé, UMR 7515, CNRSUniversité de StrasbourgStrasbourg cedexFrance
| | - Frédéric Marin
- UMR 7338, Biomécanique et Bioingénierie, Centre de recherches de RoyallieuSorbonne Universités, Université de Technologie de Compiègne, CNRSCompiègne cedexFrance
| | - Sylvie Testelin
- Facing Faces Institute, Amiens University Hospital CenterAmiens Cedex 1France
| | - Stéphanie Dakpé
- Facing Faces Institute, Amiens University Hospital CenterAmiens Cedex 1France
| | - Bernard Devauchelle
- Facing Faces Institute, Amiens University Hospital CenterAmiens Cedex 1France
| | - Christophe Egles
- UMR 7338, Biomécanique et Bioingénierie, Centre de recherches de RoyallieuSorbonne Universités, Université de Technologie de Compiègne, CNRSCompiègne cedexFrance
- Tufts University, School of Dental MedicineBostonMAUSA
| |
Collapse
|
31
|
Aijie C, Xuan L, Huimin L, Yanli Z, Yiyuan K, Yuqing L, Longquan S. Nanoscaffolds in promoting regeneration of the peripheral nervous system. Nanomedicine (Lond) 2018; 13:1067-1085. [PMID: 29790811 DOI: 10.2217/nnm-2017-0389] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The ability to surgically repair peripheral nerve injuries is urgently needed. However, traditional tissue engineering techniques, such as autologous nerve transplantation, have some limitations. Therefore, tissue engineered autologous nerve grafts have become a suitable choice for nerve repair. Novel tissue engineering techniques derived from nanostructured conduits have been shown to be superior to other successful functional neurological structures with different scaffolds in terms of providing the required structures and properties. Additionally, different biomaterials and growth factors have been added to nerve scaffolds to produce unique biological effects that promote nerve regeneration and functional recovery. This review summarizes the application of different nanoscaffolds in peripheral nerve repair and further analyzes how the nanoscaffolds promote peripheral nerve regeneration.
Collapse
Affiliation(s)
- Chen Aijie
- Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou, Guangdong 510515, China
- Guangdong Provincial Key Laboratory of Construction & Detection in Tissue Engineering, Guangzhou 510515, China
| | - Lai Xuan
- Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou, Guangdong 510515, China
| | - Liang Huimin
- Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou, Guangdong 510515, China
| | - Zhang Yanli
- Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou, Guangdong 510515, China
| | - Kang Yiyuan
- Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou, Guangdong 510515, China
| | - Lin Yuqing
- Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou, Guangdong 510515, China
| | - Shao Longquan
- Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou, Guangdong 510515, China
- Guangdong Provincial Key Laboratory of Construction & Detection in Tissue Engineering, Guangzhou 510515, China
| |
Collapse
|
32
|
Lecomte A, Descamps E, Bergaud C. A review on mechanical considerations for chronically-implanted neural probes. J Neural Eng 2018; 15:031001. [DOI: 10.1088/1741-2552/aa8b4f] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
33
|
Hsu CC, Serio A, Amdursky N, Besnard C, Stevens MM. Fabrication of Hemin-Doped Serum Albumin-Based Fibrous Scaffolds for Neural Tissue Engineering Applications. ACS APPLIED MATERIALS & INTERFACES 2018; 10:5305-5317. [PMID: 29381329 PMCID: PMC5814958 DOI: 10.1021/acsami.7b18179] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 01/12/2018] [Indexed: 05/06/2023]
Abstract
Neural tissue engineering (TE) represents a promising new avenue of therapy to support nerve recovery and regeneration. To recreate the complex environment in which neurons develop and mature, the ideal biomaterials for neural TE require a number of properties and capabilities including the appropriate biochemical and physical cues to adsorb and release specific growth factors. Here, we present neural TE constructs based on electrospun serum albumin (SA) fibrous scaffolds. We doped our SA scaffolds with an iron-containing porphyrin, hemin, to confer conductivity, and then functionalized them with different recombinant proteins and growth factors to ensure cell attachment and proliferation. We demonstrated the potential for these constructs combining topographical, biochemical, and electrical stimuli by testing them with clinically relevant neural populations derived from human induced pluripotent stem cells (hiPSCs). Our scaffolds could support the attachment, proliferation, and neuronal differentiation of hiPSC-derived neural stem cells (NSCs), and were also able to incorporate active growth factors and release them over time, which modified the behavior of cultured cells and substituted the need for growth factor supplementation by media change. Electrical stimulation on the doped SA scaffold positively influenced the maturation of neuronal populations, with neurons exhibiting more branched neurites compared to controls. Through promotion of cell proliferation, differentiation, and neurite branching of hiPSC-derived NSCs, these conductive SA fibrous scaffolds are of broad application in nerve regeneration strategies.
Collapse
Affiliation(s)
- Chia-Chen Hsu
- Department of Materials, Imperial College London, London SW7 2AZ, U.K.
- Department of Bioengineering, Imperial College London, London SW7 2AZ, U.K.
- Institute
of Biomedical Engineering, Imperial College
London, London SW7 2AZ, U.K.
| | - Andrea Serio
- Department of Materials, Imperial College London, London SW7 2AZ, U.K.
- Department of Bioengineering, Imperial College London, London SW7 2AZ, U.K.
- Institute
of Biomedical Engineering, Imperial College
London, London SW7 2AZ, U.K.
| | - Nadav Amdursky
- Department of Materials, Imperial College London, London SW7 2AZ, U.K.
- Department of Bioengineering, Imperial College London, London SW7 2AZ, U.K.
- Institute
of Biomedical Engineering, Imperial College
London, London SW7 2AZ, U.K.
| | - Cyril Besnard
- Department of Materials, Imperial College London, London SW7 2AZ, U.K.
| | - Molly M. Stevens
- Department of Materials, Imperial College London, London SW7 2AZ, U.K.
- Department of Bioengineering, Imperial College London, London SW7 2AZ, U.K.
- Institute
of Biomedical Engineering, Imperial College
London, London SW7 2AZ, U.K.
| |
Collapse
|
34
|
Guarino V, Benfenati V, Cruz-Maya I, Saracino E, Zamboni R, Ambrosio L. Instructive proteins for tissue regeneration. FUNCTIONAL 3D TISSUE ENGINEERING SCAFFOLDS 2018:23-49. [DOI: 10.1016/b978-0-08-100979-6.00002-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
35
|
Li C, Yang M, Zhu L, Zhu Y. Honeysuckle flowers extract loaded Bombyx mori silk fibroin films for inducing apoptosis of HeLa cells. Microsc Res Tech 2017; 80:1297-1303. [PMID: 28841768 PMCID: PMC5763328 DOI: 10.1002/jemt.22928] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2016] [Revised: 08/08/2017] [Accepted: 08/08/2017] [Indexed: 01/30/2023]
Abstract
This study aimed to prepare silk fibroin (SF) films loaded with honeysuckle flowers extract (HFE) for inducing apoptosis of HeLa cells. We mixed solution of SF and HFE by air-drying for preparing the honeysuckle flowers extract loaded silk fibroin (SFH) films. The physical properties including morphologies, contact angle, roughness, and Z range were characterized. MTS assay and fluorescence micrographs proved that SFH films inhibited the proliferation rate of HeLa cells due to induction of HFE into SF films. Furthermore, cell apoptosis assay and cell cycle analysis confirmed that the apoptosis of HeLa cells resulted from SFH films. Therefore, SFH films designed in our study might be a promising candidate material for cancer therapy.
Collapse
Affiliation(s)
- Chenlin Li
- Institute of Applied Bioresource, College of Animal ScienceZhejiang UniversityHangzhou, Zhejiang 310058People's Republic of China
| | - Mingying Yang
- Institute of Applied Bioresource, College of Animal ScienceZhejiang UniversityHangzhou, Zhejiang 310058People's Republic of China
| | - Liangjun Zhu
- Institute of Applied Bioresource, College of Animal ScienceZhejiang UniversityHangzhou, Zhejiang 310058People's Republic of China
| | - Yongqiang Zhu
- Zhejiang Academy of Traditional Chinese MedicineHangzhou, Zhejiang 310058People's Republic of China
| |
Collapse
|
36
|
Xue C, Zhu H, Tan D, Ren H, Gu X, Zhao Y, Zhang P, Sun Z, Yang Y, Gu J, Gu Y, Gu X. Electrospun silk fibroin-based neural scaffold for bridging a long sciatic nerve gap in dogs. J Tissue Eng Regen Med 2017; 12:e1143-e1153. [PMID: 28485084 DOI: 10.1002/term.2449] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 01/22/2017] [Accepted: 05/04/2017] [Indexed: 12/20/2022]
Abstract
Silk fibroin (SF)-derived silkworms represent a type of highly biocompatible biomaterial for tissue engineering. We have previously investigated biocompatibility of SF with neural cells isolated from the central nervous system or peripheral nerve system in vitro, and also developed a SF-based nerve graft conduit or tissue-engineered nerve grafts by introducing bone marrow mesenchymal stem cells, as support cells, into SF-based scaffold and evaluated the outcomes of peripheral nerve repair in a rat model. As an extension of the previous study, the electrospun technique was performed here to fabricate SF-based neural scaffold inserted with silk fibres for bridging a 30-mm-long sciatic nerve gap in dogs. Assessments including functional, histological and morphometrical analyses were applied 12 months after surgery. All the results indicated that the SF-based neural scaffold group achieved satisfactory regenerative outcomes, which were close to those achieved by autologous nerve grafts as the golden-standard for peripheral nerve repair. Overall, our results raise a potential possibility for the translation of SF-based electrospun neural scaffolds as an alternative to nerve autografts into the clinic.
Collapse
Affiliation(s)
- Chengbin Xue
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing Biomedical Research Institute, Nanjing University, Nanjing, China.,Key Laboratory of Neuroregeneration, Ministry of Education and Jiangsu Province, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, JS, PR China
| | - Hui Zhu
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing Biomedical Research Institute, Nanjing University, Nanjing, China.,Key Laboratory of Neuroregeneration, Ministry of Education and Jiangsu Province, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, JS, PR China.,Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, JS, PR China
| | - Dehua Tan
- Key Laboratory of Neuroregeneration, Ministry of Education and Jiangsu Province, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, JS, PR China
| | - Hechun Ren
- Key Laboratory of Neuroregeneration, Ministry of Education and Jiangsu Province, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, JS, PR China
| | - Xiaokun Gu
- Department of Hand Surgery, Affiliated Hospital of Nantong University, Nantong, JS, PR China
| | - Yahong Zhao
- Key Laboratory of Neuroregeneration, Ministry of Education and Jiangsu Province, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, JS, PR China
| | - Ping Zhang
- Key Laboratory of Neuroregeneration, Ministry of Education and Jiangsu Province, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, JS, PR China
| | - Zhichao Sun
- Key Laboratory of Neuroregeneration, Ministry of Education and Jiangsu Province, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, JS, PR China
| | - Yumin Yang
- Key Laboratory of Neuroregeneration, Ministry of Education and Jiangsu Province, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, JS, PR China
| | - Jianhui Gu
- Department of Hand Surgery, Affiliated Hospital of Nantong University, Nantong, JS, PR China
| | - Yun Gu
- Key Laboratory of Neuroregeneration, Ministry of Education and Jiangsu Province, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, JS, PR China
| | - Xiaosong Gu
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing Biomedical Research Institute, Nanjing University, Nanjing, China.,Key Laboratory of Neuroregeneration, Ministry of Education and Jiangsu Province, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, JS, PR China
| |
Collapse
|
37
|
|
38
|
Tonazzini I, Moffa M, Pisignano D, Cecchini M. Neuregulin 1 functionalization of organic fibers for Schwann cell guidance. NANOTECHNOLOGY 2017; 28:155303. [PMID: 28303795 DOI: 10.1088/1361-6528/aa6316] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
The repair of peripheral nerve lesions is a clinical problem where the functional recovery is often far from being satisfactory, although peripheral nerves generally retain good potential for regeneration. Here, we develop a novel scaffold approach based on bioactive fibers of poly(ε-caprolactone) where nanotopographical guidance and neuregulin 1 (NRG1) cues are combined. We interface them with rat primary Schwann cells (SCs), the peripheral glial cells that drive initial regeneration of injured nerves, and found that the combination of NRG1 with parallel nano-fibrous topographies is effective in improving SC growth up to 72 h, alignment to fiber topography, and bipolar differentiation, opening original perspectives for nerve repair applications.
Collapse
Affiliation(s)
- Ilaria Tonazzini
- NEST, Istituto Nanoscienze-CNR and Scuola Normale Superiore, Piazza San Silvestro 12, Pisa I-56127, Italy. Fondazione Umberto Veronesi, Piazza Velasca 5, Milan I-20122, Italy
| | | | | | | |
Collapse
|
39
|
Girard D, Laverdet B, Buhé V, Trouillas M, Ghazi K, Alexaline MM, Egles C, Misery L, Coulomb B, Lataillade JJ, Berthod F, Desmoulière A. Biotechnological Management of Skin Burn Injuries: Challenges and Perspectives in Wound Healing and Sensory Recovery. TISSUE ENGINEERING PART B-REVIEWS 2017; 23:59-82. [DOI: 10.1089/ten.teb.2016.0195] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Dorothée Girard
- University of Limoges, Myelin Maintenance and Peripheral Neuropathies (EA 6309), Faculties of Medicine and Pharmacy, Limoges, France
| | - Betty Laverdet
- University of Limoges, Myelin Maintenance and Peripheral Neuropathies (EA 6309), Faculties of Medicine and Pharmacy, Limoges, France
| | - Virginie Buhé
- University of Western Brittany, Laboratory of Neurosciences of Brest (EA 4685), Brest, France
| | - Marina Trouillas
- Paris Sud University, Unité mixte Inserm/SSA 1197, IRBA/CTSA/HIA Percy, École du Val de Grâce, Clamart, France
| | - Kamélia Ghazi
- Sorbonne University, Université de Technologie de Compiègne, CNRS UMR 7338 Biomechanics and Bioengineering, Centre de Recherche Royallieu, Compiègne, France
| | - Maïa M. Alexaline
- Paris Sud University, Unité mixte Inserm/SSA 1197, IRBA/CTSA/HIA Percy, École du Val de Grâce, Clamart, France
| | - Christophe Egles
- Sorbonne University, Université de Technologie de Compiègne, CNRS UMR 7338 Biomechanics and Bioengineering, Centre de Recherche Royallieu, Compiègne, France
| | - Laurent Misery
- University of Western Brittany, Laboratory of Neurosciences of Brest (EA 4685), Brest, France
| | - Bernard Coulomb
- Paris Sud University, Unité mixte Inserm/SSA 1197, IRBA/CTSA/HIA Percy, École du Val de Grâce, Clamart, France
| | - Jean-Jacques Lataillade
- Paris Sud University, Unité mixte Inserm/SSA 1197, IRBA/CTSA/HIA Percy, École du Val de Grâce, Clamart, France
| | - François Berthod
- Centre LOEX de l'Université Laval, Centre de recherche du CHU de Québec and Département de Chirurgie, Faculté de Médecine, Université Laval, Québec, Canada
| | - Alexis Desmoulière
- University of Limoges, Myelin Maintenance and Peripheral Neuropathies (EA 6309), Faculties of Medicine and Pharmacy, Limoges, France
| |
Collapse
|
40
|
Hansen C, Dinis TM, Vidal G, Ben-Mansour K, Bresson D, Egles C, Marin F. In-vivo analysis of nerve regeneration after sciatic nerve injury in a rat model. Int Biomech 2016. [DOI: 10.1080/23335432.2016.1233077] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Affiliation(s)
- Clint Hansen
- Sorbonne Universités, Université de Technologie de Compiègne (UTC), Compiègne, France
| | - Tony M. Dinis
- Sorbonne Universités, Université de Technologie de Compiègne (UTC), Compiègne, France
| | - Guillaume Vidal
- Sorbonne Universités, Université de Technologie de Compiègne (UTC), Compiègne, France
| | - Khalil Ben-Mansour
- Sorbonne Universités, Université de Technologie de Compiègne (UTC), Compiègne, France
| | - Damien Bresson
- Sorbonne Universités, Université de Technologie de Compiègne (UTC), Compiègne, France
| | - Christophe Egles
- Sorbonne Universités, Université de Technologie de Compiègne (UTC), Compiègne, France
- Department of Oral and Maxillofacial Pathology, School of Dental Medicine, Tufts University, Boston, MA, USA
| | - Frédéric Marin
- Sorbonne Universités, Université de Technologie de Compiègne (UTC), Compiègne, France
| |
Collapse
|
41
|
Quan Q, Chang B, Meng HY, Liu RX, Wang Y, Lu SB, Peng J, Zhao Q. Use of electrospinning to construct biomaterials for peripheral nerve regeneration. Rev Neurosci 2016; 27:761-768. [PMID: 27428846 DOI: 10.1515/revneuro-2016-0032] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 05/26/2016] [Indexed: 12/16/2022]
Abstract
AbstractA number of limitations associated with the use of hollow nerve guidance conduits (NGCs) require further discussion. Most importantly, the functional recovery outcomes after the placement of hollow NGCs are poor even after the successful bridging of peripheral nerve injuries. However, nerve regeneration scaffolds built using electric spinning have several advantages that may improve functional recovery. Thus, the present study summarizes recent developments in this area, including the key cells that are combined with the scaffold and associated with nerve regeneration, the structure and configuration of the electrospinning design (which determines the performance of the electrospinning scaffold), the materials the electrospinning fibers are composed of, and the methods used to control the morphology of a single fiber. Additionally, this study also discusses the processes underlying peripheral nerve regeneration. The primary goals of the present review were to evaluate and consolidate the findings of studies that used scaffolding biomaterials built by electrospinning used for peripheral nerve regeneration support. It is amazing that the field of peripheral nerve regeneration continues to consistently produce such a wide variety of innovative techniques and novel types of equipment, because the introduction of every new process creates an opportunity for advances in materials for nerve repair.
Collapse
|
42
|
Approaches to Peripheral Nerve Repair: Generations of Biomaterial Conduits Yielding to Replacing Autologous Nerve Grafts in Craniomaxillofacial Surgery. BIOMED RESEARCH INTERNATIONAL 2016; 2016:3856262. [PMID: 27556032 PMCID: PMC4983313 DOI: 10.1155/2016/3856262] [Citation(s) in RCA: 128] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 06/29/2016] [Indexed: 01/09/2023]
Abstract
Peripheral nerve injury is a common clinical entity, which may arise due to traumatic, tumorous, or even iatrogenic injury in craniomaxillofacial surgery. Despite advances in biomaterials and techniques over the past several decades, reconstruction of nerve gaps remains a challenge. Autografts are the gold standard for nerve reconstruction. Using autografts, there is donor site morbidity, subsequent sensory deficit, and potential for neuroma development and infection. Moreover, the need for a second surgical site and limited availability of donor nerves remain a challenge. Thus, increasing efforts have been directed to develop artificial nerve guidance conduits (ANCs) as new methods to replace autografts in the future. Various synthetic conduit materials have been tested in vitro and in vivo, and several first- and second-generation conduits are FDA approved and available for purchase, while third-generation conduits still remain in experimental stages. This paper reviews the current treatment options, summarizes the published literature, and assesses future prospects for the repair of peripheral nerve injury in craniomaxillofacial surgery with a particular focus on facial nerve regeneration.
Collapse
|
43
|
Johnson BN, Jia X. 3D printed nerve guidance channels: computer-aided control of geometry, physical cues, biological supplements and gradients. Neural Regen Res 2016; 11:1568-1569. [PMID: 27904481 PMCID: PMC5116829 DOI: 10.4103/1673-5374.193230] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Affiliation(s)
- Blake N Johnson
- Department of Industrial and Systems Engineering, School of Neuroscience, Virginia Tech, Blacksburg, VA, USA
| | - Xiaofeng Jia
- Department of Neurosurgery, Orthopaedics, Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA; Department of Biomedical Engineering, Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
44
|
Razavi S, Karbasi S, Morshed M, Zarkesh Esfahani H, Golozar M, Vaezifar S. Cell Attachment and Proliferation of Human Adipose-Derived Stem Cells on PLGA/Chitosan Electrospun Nano-Biocomposite. CELL JOURNAL 2015; 17:429-37. [PMID: 26464814 PMCID: PMC4601863 DOI: 10.22074/cellj.2015.4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Accepted: 10/04/2014] [Indexed: 11/04/2022]
Abstract
Objective In this study, nano-biocomposite composed of poly (lactide-co-glycolide)
(PLGA) and chitosan (CS) were electrospun through a single nozzle by dispersing the CS
nano-powders in PLGA solution. The cellular behavior of human adipose derived stem
cells (h-ADSCs) on random and aligned scaffolds was then evaluated.
Materials and Methods In this experimental study, the PLGA/CS scaffolds were prepared
at the different ratios of 90/10, 80/20, and 70/30 (w/w) %. Morphology, cell adhesion and prolif-
eration rate of h-ADSCs on the scaffolds were assessed using scanning electron microscope
(SEM), 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyl tetrazolium bromide (MTT) assay and trypan
blue staining respectively.
Results H-ADSCs seeded on the matrices indicated that the PLGA/CS composite matrix
with aligned nanofibres and higher content of CS nano-powders gave significantly better
performance than others in terms of cell adhesion and proliferation rate (P<0.05).
Conclusion We found that CS enhanced cell adhesion and proliferation rate, and
aligned nanofibers guided cell growth along the longitudinal axis of the nanofibers,
which would provide a beneficial approach for tissue engineering.
Collapse
Affiliation(s)
- Shahnaz Razavi
- Department of Anatomical Sciences and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Saeed Karbasi
- Department of Medical Physics and Biomedical Engineering, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammad Morshed
- Department of Textile Engineering, Isfahan University of Technology, Isfahan, Iran
| | | | - Mohammad Golozar
- Department of Materials Engineering, Isfahan University of Technology, Isfahan, Iran
| | - Sedigheh Vaezifar
- Department of Anatomical Sciences and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran ; Department of Materials Engineering, Isfahan University of Technology, Isfahan, Iran
| |
Collapse
|
45
|
Sugimura-Wakayama Y, Katagiri W, Osugi M, Kawai T, Ogata K, Sakaguchi K, Hibi H. Peripheral Nerve Regeneration by Secretomes of Stem Cells from Human Exfoliated Deciduous Teeth. Stem Cells Dev 2015; 24:2687-99. [PMID: 26154068 DOI: 10.1089/scd.2015.0104] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Peripheral nerve regeneration across nerve gaps is often suboptimal, with poor functional recovery. Stem cell transplantation-based regenerative therapy is a promising approach for axon regeneration and functional recovery of peripheral nerve injury; however, the mechanisms remain controversial and unclear. Recent studies suggest that transplanted stem cells promote tissue regeneration through a paracrine mechanism. We investigated the effects of conditioned media derived from stem cells from human exfoliated deciduous teeth (SHED-CM) on peripheral nerve regeneration. In vitro, SHED-CM-treated Schwann cells exhibited significantly increased proliferation, migration, and the expression of neuron-, extracellular matrix (ECM)-, and angiogenesis-related genes. SHED-CM stimulated neuritogenesis of dorsal root ganglia and increased cell viability. Similarly, SHED-CM enhanced tube formation in an angiogenesis assay. In vivo, a 10-mm rat sciatic nerve gap model was bridged by silicon conduits containing SHED-CM or serum-free Dulbecco's modified Eagle's medium. Light and electron microscopy confirmed that the number of myelinated axons and axon-to-fiber ratio (G-ratio) were significantly higher in the SHED-CM group at 12 weeks after nerve transection surgery. The sciatic functional index (SFI) and gastrocnemius (target muscle) wet weight ratio demonstrated functional recovery. Increased compound muscle action potentials and increased SFI in the SHED-CM group suggested sciatic nerve reinnervation of the target muscle and improved functional recovery. We also observed reduced muscle atrophy in the SHED-CM group. Thus, SHEDs may secrete various trophic factors that enhance peripheral nerve regeneration through multiple mechanisms. SHED-CM may therefore provide a novel therapy that creates a more desirable extracellular microenvironment for peripheral nerve regeneration.
Collapse
Affiliation(s)
- Yukiko Sugimura-Wakayama
- 1 Department of Oral and Maxillofacial Surgery, Nagoya University Graduate School of Medicine , Nagoya, Japan
| | - Wataru Katagiri
- 1 Department of Oral and Maxillofacial Surgery, Nagoya University Graduate School of Medicine , Nagoya, Japan
| | - Masashi Osugi
- 1 Department of Oral and Maxillofacial Surgery, Nagoya University Graduate School of Medicine , Nagoya, Japan
| | - Takamasa Kawai
- 1 Department of Oral and Maxillofacial Surgery, Nagoya University Graduate School of Medicine , Nagoya, Japan
| | - Kenichi Ogata
- 1 Department of Oral and Maxillofacial Surgery, Nagoya University Graduate School of Medicine , Nagoya, Japan .,2 Section of Oral and Maxillofacial Oncology, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University , Fukuoka, Japan
| | - Kohei Sakaguchi
- 1 Department of Oral and Maxillofacial Surgery, Nagoya University Graduate School of Medicine , Nagoya, Japan
| | - Hideharu Hibi
- 1 Department of Oral and Maxillofacial Surgery, Nagoya University Graduate School of Medicine , Nagoya, Japan
| |
Collapse
|
46
|
Hansen C, Dinis TM, Vidal G, Ben-Mansour K, Bresson D, Egles C, Marin F. Motion capture for functional analysis of new biomaterials in a small animal model. Comput Methods Biomech Biomed Engin 2015; 18 Suppl 1:1956-7. [PMID: 26230375 DOI: 10.1080/10255842.2015.1069589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Affiliation(s)
- C Hansen
- a Sorbonne Unversités, Université de technologie de Compiégne, UMR CNRS 7338 , Compiègne , France
| | - T M Dinis
- a Sorbonne Unversités, Université de technologie de Compiégne, UMR CNRS 7338 , Compiègne , France
| | - G Vidal
- a Sorbonne Unversités, Université de technologie de Compiégne, UMR CNRS 7338 , Compiègne , France
| | - K Ben-Mansour
- a Sorbonne Unversités, Université de technologie de Compiégne, UMR CNRS 7338 , Compiègne , France
| | - D Bresson
- a Sorbonne Unversités, Université de technologie de Compiégne, UMR CNRS 7338 , Compiègne , France
| | - C Egles
- a Sorbonne Unversités, Université de technologie de Compiégne, UMR CNRS 7338 , Compiègne , France.,b Department of Oral and Maxillofacial Pathology, School of Dental Medicine , Tufts University , Boston , MS , USA
| | - F Marin
- a Sorbonne Unversités, Université de technologie de Compiégne, UMR CNRS 7338 , Compiègne , France
| |
Collapse
|
47
|
Kilinc D, Blasiak A, Lee GU. Microtechnologies for studying the role of mechanics in axon growth and guidance. Front Cell Neurosci 2015; 9:282. [PMID: 26283918 PMCID: PMC4515553 DOI: 10.3389/fncel.2015.00282] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 07/10/2015] [Indexed: 12/18/2022] Open
Abstract
The guidance of axons to their proper targets is not only a crucial event in neurodevelopment, but also a potential therapeutic target for neural repair. Axon guidance is mediated by various chemo- and haptotactic cues, as well as the mechanical interactions between the cytoskeleton and the extracellular matrix (ECM). Axonal growth cones, dynamic ends of growing axons, convert external stimuli to biochemical signals, which, in turn, are translated into behavior, e.g., turning or retraction, via cytoskeleton-matrix linkages. Despite the inherent mechanical nature of the problem, the role of mechanics in axon guidance is poorly understood. Recent years has witnessed the application of a range of microtechnologies in neurobiology, from microfluidic circuits to single molecule force spectroscopy. In this mini-review, we describe microtechnologies geared towards dissecting the mechanical aspects of axon guidance, divided into three categories: controlling the growth cone microenvironment, stimulating growth cones with externally applied forces, and measuring forces exerted by the growth cones. A particular emphasis is given to those studies that combine multiple techniques, as dictated by the complexity of the problem.
Collapse
Affiliation(s)
- Devrim Kilinc
- Bionanosciences Group, School of Chemisty and Chemical Biology, University College Dublin Belfield, Dublin, Ireland
| | - Agata Blasiak
- Bionanosciences Group, School of Chemisty and Chemical Biology, University College Dublin Belfield, Dublin, Ireland
| | - Gil U Lee
- Bionanosciences Group, School of Chemisty and Chemical Biology, University College Dublin Belfield, Dublin, Ireland
| |
Collapse
|
48
|
Shen G, Hu X, Guan G, Wang L. Surface Modification and Characterisation of Silk Fibroin Fabric Produced by the Layer-by-Layer Self-Assembly of Multilayer Alginate/Regenerated Silk Fibroin. PLoS One 2015; 10:e0124811. [PMID: 25919690 PMCID: PMC4412632 DOI: 10.1371/journal.pone.0124811] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 03/17/2015] [Indexed: 02/05/2023] Open
Abstract
Silk-based medical products have a long history of use as a material for surgical sutures because of their desirable mechanical properties. However, silk fibroin fabric has been reported to be haemolytic when in direct contact with blood. The layer-by-layer self-assembly technique provides a method for surface modification to improve the biocompatibility of silk fibroin fabrics. Regenerated silk fibroin and alginate, which have excellent biocompatibility and low immunogenicity, are outstanding candidates for polyelectrolyte deposition. In this study, silk fabric was degummed and positively charged to create a silk fibroin fabric that could undergo self-assembly. The multilayer self-assembly of the silk fibroin fabric was achieved by alternating the polyelectrolyte deposition of a negatively charged alginate solution (pH = 8) and a positively charged regenerated silk fibroin solution (pH = 2). Finally, the negatively charged regenerated silk fibroin solution (pH = 8) was used to assemble the outermost layer of the fabric so that the surface would be negatively charged. A stable structural transition was induced using 75% ethanol. The thickness and morphology were characterised using atomic force microscopy. The properties of the self-assembled silk fibroin fabric, such as the bursting strength, thermal stability and flushing stability, indicated that the fabric was stable. In addition, the cytocompatibility and haemocompatibility of the self-assembled silk fibroin fabrics were evaluated. The results indicated that the biocompatibility of the self-assembled multilayers was acceptable and that it improved markedly. In particular, after the self-assembly, the fabric was able to prevent platelet adhesion. Furthermore, other non-haemolytic biomaterials can be created through self-assembly of more than 1.5 bilayers, and we propose that self-assembled silk fibroin fabric may be an attractive candidate for anticoagulation applications and for promoting endothelial cell adhesion for vascular prostheses.
Collapse
Affiliation(s)
- Gaotian Shen
- Key Laboratory of Textile Science and Technology, Ministry of Education, College of Textiles, Donghua University, Songjiang District, Shanghai 201620, China
| | - Xingyou Hu
- Key Laboratory of Textile Science and Technology, Ministry of Education, College of Textiles, Donghua University, Songjiang District, Shanghai 201620, China
| | - Guoping Guan
- Key Laboratory of Textile Science and Technology, Ministry of Education, College of Textiles, Donghua University, Songjiang District, Shanghai 201620, China
- Engineering Research Center of Technical Textiles, Ministry of Education, Shanghai 201620, China
| | - Lu Wang
- Key Laboratory of Textile Science and Technology, Ministry of Education, College of Textiles, Donghua University, Songjiang District, Shanghai 201620, China
| |
Collapse
|
49
|
Tian L, Prabhakaran MP, Hu J, Chen M, Besenbacher F, Ramakrishna S. Coaxial electrospun poly(lactic acid)/silk fibroin nanofibers incorporated with nerve growth factor support the differentiation of neuronal stem cells. RSC Adv 2015. [DOI: 10.1039/c5ra05773f] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Plasma treated PLA/silk fibroin/NGF nanofibers with core–shell structure could enhance the neuronal differentiation of PC12 cells.
Collapse
Affiliation(s)
- Lingling Tian
- Center for Nanofibers and Nanotechnology
- E3-05-14
- Department of Mechanical Engineering
- Faculty of Engineering
- National University of Singapore
| | - Molamma P. Prabhakaran
- Center for Nanofibers and Nanotechnology
- E3-05-14
- Department of Mechanical Engineering
- Faculty of Engineering
- National University of Singapore
| | - Jue Hu
- Center for Nanofibers and Nanotechnology
- E3-05-14
- Department of Mechanical Engineering
- Faculty of Engineering
- National University of Singapore
| | - Menglin Chen
- Interdisciplinary Nanoscience Center (iNANO)
- Aarhus University
- Aarhus
- Denmark
| | | | - Seeram Ramakrishna
- Center for Nanofibers and Nanotechnology
- E3-05-14
- Department of Mechanical Engineering
- Faculty of Engineering
- National University of Singapore
| |
Collapse
|