1
|
Chansoria P, Winkelbauer M, Zhang S, Janiak J, Liu H, Boev D, Morandi A, Grange R, Zenobi-Wong M. Structured Light Projection Using Image Guide Fibers for In Situ Photo-biofabrication. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025:e2419350. [PMID: 40297914 DOI: 10.1002/adma.202419350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 03/30/2025] [Indexed: 04/30/2025]
Abstract
Light-based biofabrication techniques have revolutionized the field of tissue engineering and regenerative medicine. Specifically, the projection of structured light, where the spatial distribution of light is controlled at both macro and microscale, has enabled precise fabrication of complex three dimensional structures with high resolution and speed. However, despite tremendous progress, biofabrication processes are mostly limited to benchtop devices which limit the flexibility in terms of where the fabrication can occur. Here, a Fiber-assisted Structured Light (FaSt-Light) projection apparatus for rapid in situ crosslinking of photoresins is demonstrated. This approach uses image-guide fiber bundles which can project bespoke images at multiple wavelengths, enabling flexibility and spatial control of different photoinitiation systems and crosslinking chemistries and also the location of fabrication. Coupling of different sizes of fibers and different lenses attached to the fibers to project small (several mm) or large (several cm) images for material crosslinking is demonstrated. FaSt-Light allows control over the cross-section of the crosslinked resins and enables the introduction of microfilaments which can further guide cellular infiltration, differentiation, and anisotropic matrix production. The proposed approach can lead to a new range of in situ biofabrication techniques which improve the translational potential of photofabricated tissues and grafts.
Collapse
Affiliation(s)
- Parth Chansoria
- Department of Health Sciences and Technology, Institute for Biomechanics, Tissue Engineering and Biofabrication Group, ETH Zürich, 8093, Switzerland
| | - Michael Winkelbauer
- Department of Health Sciences and Technology, Institute for Biomechanics, Tissue Engineering and Biofabrication Group, ETH Zürich, 8093, Switzerland
| | - Shipin Zhang
- Department of Health Sciences and Technology, Institute for Biomechanics, Tissue Engineering and Biofabrication Group, ETH Zürich, 8093, Switzerland
| | - Jakub Janiak
- Department of Health Sciences and Technology, Institute for Biomechanics, Tissue Engineering and Biofabrication Group, ETH Zürich, 8093, Switzerland
| | - Hao Liu
- Department of Health Sciences and Technology, Institute for Biomechanics, Tissue Engineering and Biofabrication Group, ETH Zürich, 8093, Switzerland
| | - Dimitar Boev
- Department of Health Sciences and Technology, Institute for Biomechanics, Tissue Engineering and Biofabrication Group, ETH Zürich, 8093, Switzerland
| | - Andrea Morandi
- Department of Physics, Institute for Quantum Electronics, Optical Nanomaterial Group, ETH Zürich, 8093, Switzerland
| | - Rachel Grange
- Department of Physics, Institute for Quantum Electronics, Optical Nanomaterial Group, ETH Zürich, 8093, Switzerland
| | - Marcy Zenobi-Wong
- Department of Health Sciences and Technology, Institute for Biomechanics, Tissue Engineering and Biofabrication Group, ETH Zürich, 8093, Switzerland
| |
Collapse
|
2
|
Su S, Wang J. A Comprehensive Review on Bioprinted Graphene-Based Material (GBM)-Enhanced Scaffolds for Nerve Guidance Conduits. Biomimetics (Basel) 2025; 10:213. [PMID: 40277612 PMCID: PMC12024949 DOI: 10.3390/biomimetics10040213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 03/21/2025] [Accepted: 03/27/2025] [Indexed: 04/26/2025] Open
Abstract
Peripheral nerve injuries (PNIs) pose significant challenges to recovery, often resulting in impaired function and quality of life. To address these challenges, nerve guidance conduits (NGCs) are being developed as effective strategies to promote nerve regeneration by providing a supportive framework that guides axonal growth and facilitates reconnection of severed nerves. Among the materials being explored, graphene-based materials (GBMs) have emerged as promising candidates due to their unique properties. Their unique properties-such as high mechanical strength, excellent electrical conductivity, and favorable biocompatibility-make them ideal for applications in nerve repair. The integration of 3D printing technologies further enhances the development of GBM-based NGCs, enabling the creation of scaffolds with complex architectures and precise topographical cues that closely mimic the natural neural environment. This customization significantly increases the potential for successful nerve repair. This review offers a comprehensive overview of properties of GBMs, the principles of 3D printing, and key design strategies for 3D-printed NGCs. Additionally, it discusses future perspectives and research directions that could advance the application of 3D-printed GBMs in nerve regeneration therapies.
Collapse
Affiliation(s)
- Siheng Su
- Department of Mechanical Engineering, California State University, Fullerton, CA 92831, USA
| | - Jilong Wang
- Key Laboratory of Clean Dyeing and Finishing Technology of Zhejiang Province, College of Textile and Garment, Shaoxing University, Shaoxing 312000, China
- Shaoxing Sub-Center of National Engineering Research Center for Fiber-Based Composites, Shaoxing University, Shaoxing 312000, China
- Shaoxing Key Laboratory of High Performance Fibers & Products, Shaoxing University, Shaoxing 312000, China
| |
Collapse
|
3
|
Wu Z, Sun J, Liao Z, Sun T, Huang L, Qiao J, Ling C, Chen C, Zhang B, Wang H. Activation of PAR1 contributes to ferroptosis of Schwann cells and inhibits regeneration of myelin sheath after sciatic nerve crush injury in rats via Hippo-YAP/ACSL4 pathway. Exp Neurol 2025; 384:115053. [PMID: 39542339 DOI: 10.1016/j.expneurol.2024.115053] [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: 08/16/2024] [Revised: 11/06/2024] [Accepted: 11/09/2024] [Indexed: 11/17/2024]
Abstract
OBJECTIVE Peripheral nerve injury (PNI) is characterized by high incidence and sequela rate. Recently, there was increasing evidence that has shown ferroptosis may impede functional recovery. Our objective is to explore the novel mechanism that regulates ferroptosis after PNI. METHODS LC-MS/MS proteomics was used to explore the possible differential signals, while PCR array was performed to investigate the differential factors. Besides, we also tried to activate or inhibit the key factors and then observe the level of ferroptosis. Regeneration of myelin sheath was finally examined in vivo via transmission electron microscopy. RESULTS Proteomics analysis suggested coagulation signal was activated after sciatic nerve crush injury, in which high expression of F2 (encoding thrombin) and F2r (encoding PAR1) were observed. Both thrombin and PAR1-targeted activator TRAP6 can induce ferroptosis in RSC96 cells, which can be rescued by Vorapaxar (PAR1 targeted inhibitor) in vitro. Further PCR array revealed that activation of PAR1 induced ferroptosis in RSC96 cells by increasing expression of YAP and ACSL4. Immunofluorescence of sciatic nerve confirmed that the expression of YAP and ACSL4 were simultaneously reduced after PAR1 inhibition, which may contribute to myelin regeneration after injury in SD rats. CONCLUSION Inhibition of PAR1 can relieve ferroptosis after sciatic nerve crush injury in SD rats through Hippo-YAP/ACSL4 pathway, thereby regulating myelin regeneration after injury. In summary, PAR1/Hippo-YAP/ACSL4 pathway may be a promising therapeutic target for promoting functional recovery post-sciatic crush injury.
Collapse
Affiliation(s)
- Zhimin Wu
- Department of Neurosurgery, the Third Affiliated Hospital, Sun Yat-Sen University, 600 Tian He Road, Tian He District, Guangzhou, Guangdong 510630, China.
| | - Jun Sun
- Department of Neurosurgery, the Third Affiliated Hospital, Sun Yat-Sen University, 600 Tian He Road, Tian He District, Guangzhou, Guangdong 510630, China.
| | - Zhi Liao
- Department of Neurosurgery, the Third Affiliated Hospital, Sun Yat-Sen University, 600 Tian He Road, Tian He District, Guangzhou, Guangdong 510630, China.
| | - Tao Sun
- Department of Neurosurgery, the Third Affiliated Hospital, Sun Yat-Sen University, 600 Tian He Road, Tian He District, Guangzhou, Guangdong 510630, China.
| | - Lixin Huang
- Department of Neurosurgery, the Third Affiliated Hospital, Sun Yat-Sen University, 600 Tian He Road, Tian He District, Guangzhou, Guangdong 510630, China.
| | - Jia Qiao
- Department of Rehabilitation Medicine, the Third Affiliated Hospital, Sun Yat-Sen University, 600 Tian He Road, Tian He District, Guangzhou, Guangdong 510630, China.
| | - Cong Ling
- Department of Neurosurgery, the Third Affiliated Hospital, Sun Yat-Sen University, 600 Tian He Road, Tian He District, Guangzhou, Guangdong 510630, China.
| | - Chuan Chen
- Department of Neurosurgery, the Third Affiliated Hospital, Sun Yat-Sen University, 600 Tian He Road, Tian He District, Guangzhou, Guangdong 510630, China.
| | - Baoyu Zhang
- Department of Neurosurgery, the Third Affiliated Hospital, Sun Yat-Sen University, 600 Tian He Road, Tian He District, Guangzhou, Guangdong 510630, China.
| | - Hui Wang
- Department of Neurosurgery, the Third Affiliated Hospital, Sun Yat-Sen University, 600 Tian He Road, Tian He District, Guangzhou, Guangdong 510630, China.
| |
Collapse
|
4
|
Sawada E, Yamamoto T, Kishimoto N, Ooishi D, Sasakura H, Takeuchi K, Seo K. Creating Alternative Afferent Input to Facilitate the Regeneration of Injured Primary Afferent Neurons. Cureus 2025; 17:e78708. [PMID: 39926623 PMCID: PMC11805595 DOI: 10.7759/cureus.78708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/07/2025] [Indexed: 02/11/2025] Open
Abstract
The trigeminal spinal tract nucleus receives primary afferent input from the orofacial region, serving as a relay between peripheral terminals and secondary neurons. The trigeminal nerve is divided into ophthalmic, maxillary, and mandibular. While it is known that primary afferent terminals synapse with secondary neurons, the interaction between different primary terminals remains unclear. Recent studies have shown that trigeminal neurons with lost input can be activated through electrical stimulation of other afferent terminals. Therefore, we examined the possibility of inducing neural activity using synaptic organizers to promote circuit reorganization. To assess the regeneration of the injured inferior alveolar nerve (third division of the trigeminal nerve), the potential involvement of input from the infraorbital nerve (second division of the trigeminal nerve) in the regeneration of the injured inferior alveolar nerve (third division of the trigeminal nerve) was investigated. Intact and injured groups were created for the second and third divisions to facilitate comparative analysis. A synapse organizer was applied to establish input between the primary afferent terminals of these divisions. This study aimed to determine if central connections between different terminals can activate trigeminal neurons with lost input, ultimately promoting peripheral nerve regeneration. In this research, male C57BL/6J mice (seven to nine weeks old) (total n=40) underwent transection of the inferior alveolar nerve. They were divided into three groups: intact (n=10), injured (saline control) (n=10), and synapse organizer (n=10). In addition, the mice were divided into two groups: one group underwent inferior alveolar nerve transection only (II, intact; III, injured, n=5), and the other group underwent transection of both the infraorbital and inferior alveolar nerves (II, injured; III, injured, n=5), followed by local administration of a synapse organizer. Regeneration was assessed using immunostaining, sensory tests, and retrograde tracing. Regeneration was confirmed by retrograde tracing and functional recovery of sensory thresholds in the skin of the mental region. These findings align with previous observations that infraorbital nerve transection reduced regeneration activity, suggesting that infraorbital input triggered regeneration in the mandibular nerve. Thus, the results propose a novel therapeutic approach where mandibular nerve injury can be treated by stimulating the infraorbital nerve immediately after injury, enhancing peripheral nerve regeneration.
Collapse
Affiliation(s)
- Emi Sawada
- Division of Dental Anesthesiology, Faculty of Dentistry and Graduate School of Medical and Dental Sciences, Niigata University, Niigata, JPN
| | - Toru Yamamoto
- Division of Dental Anesthesiology, Faculty of Dentistry and Graduate School of Medical and Dental Sciences, Niigata University, Niigata, JPN
| | - Naotaka Kishimoto
- Division of Dental Anesthesiology, Faculty of Dentistry and Graduate School of Medical and Dental Sciences, Niigata University, Niigata, JPN
| | - Dai Ooishi
- Department of Medical Cell Biology, School of Medicine, Aichi Medical University, Nagakute, JPN
| | - Hiroyuki Sasakura
- Department of Medical Cell Biology, School of Medicine, Aichi Medical University, Nagakute, JPN
| | - Kosei Takeuchi
- Department of Medical Cell Biology, School of Medicine, Aichi Medical University, Nagakute, JPN
- Medical Research Creation Center, Aichi Medical University, Nagakute, JPN
| | - Kenji Seo
- Division of Dental Anesthesiology, Faculty of Dentistry and Graduate School of Medical and Dental Sciences, Niigata University, Niigata, JPN
| |
Collapse
|
5
|
Matsuo T, Kimura H, Nishijima T, Kiyota Y, Suzuki T, Nagoshi N, Shibata S, Shindo T, Moritoki N, Sasaki M, Noguchi S, Tamada Y, Nakamura M, Iwamoto T. Peripheral nerve regeneration using a bioresorbable silk fibroin-based artificial nerve conduit fabricated via a novel freeze-thaw process. Sci Rep 2025; 15:3797. [PMID: 39885362 PMCID: PMC11782519 DOI: 10.1038/s41598-025-88221-y] [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: 09/20/2024] [Accepted: 01/23/2025] [Indexed: 02/01/2025] Open
Abstract
While silk fibroin (SF) obtained from silkworm cocoons is expected to become a next-generation natural polymer, a fabrication method for SF-based artificial nerve conduits (SFCs) has not yet been established. Here, we report a bioresorbable SFC, fabricated using a novel freeze-thaw process, which ensures biosafety by avoiding any harmful chemical additives. The SFC demonstrated favorable biocompatibility (high hydrophilicity and porosity with a water content of > 90%), structural stability (stiffness, toughness, and elasticity), and biodegradability, making it an ideal candidate for nerve regeneration. We evaluated the nerve-regenerative effects of the SFC in a rat sciatic-nerve-defect model, including its motor and sensory function recovery as well as histological regeneration. We found that SFC transplantation significantly promoted functional recovery and nerve regeneration compared to silicone tubes and was almost equally effective as autologous nerve transplantation. Histological analyses indicated that vascularization and M2 macrophage recruitment were pronounced inside the SFC. These results suggest that the unique properties of the SFC further enhanced the peripheral nerve regeneration mechanism. As no SFC has been applied in clinical practice, the SFC reported herein may be a promising candidate for repairing extensive peripheral nerve defects.
Collapse
Affiliation(s)
- Tomoki Matsuo
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-Ku, Tokyo, 160-8582, Japan
| | - Hiroo Kimura
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-Ku, Tokyo, 160-8582, Japan.
- Department of Orthopaedic Surgery, Hand and Upper Extremity Surgery Center, Kitasato Institute Hospital, 9-1, Shirokane 5-Chome, Minato-Ku, Tokyo, 108-8642, Japan.
| | - Takayuki Nishijima
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-Ku, Tokyo, 160-8582, Japan
| | - Yasuhiro Kiyota
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-Ku, Tokyo, 160-8582, Japan
| | - Taku Suzuki
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-Ku, Tokyo, 160-8582, Japan
| | - Narihito Nagoshi
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-Ku, Tokyo, 160-8582, Japan
| | - Shinsuke Shibata
- Division of Microscopic Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, 951-8510, Japan
- Electron Microscope Laboratory, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-Ku, Tokyo, 160-8582, Japan
| | - Tomoko Shindo
- Electron Microscope Laboratory, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-Ku, Tokyo, 160-8582, Japan
| | - Nobuko Moritoki
- Electron Microscope Laboratory, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-Ku, Tokyo, 160-8582, Japan
| | - Makoto Sasaki
- Faculty of Advanced Science and Technology, Kumamoto University, 2-39-1 Kurokami, Chuo-Ku, Kumamoto, 860-8555, Japan
- Charlie Lab Inc., 2-39-1 Kurokami, Chuou-Ku, Kumamoto, 860-8555, Japan
| | - Sarara Noguchi
- Materials Development Department, Kumamoto Industrial Research Institute, 3-11-38 Higashimachi, Higashi-Ku, Kumamoto, 862-0901, Japan
| | - Yasushi Tamada
- Faculty of Textile Science and Technology, Shinshu University, 3-15-1 Tokida, Ueda City, Nagano, 386-8567, Japan
| | - Masaya Nakamura
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-Ku, Tokyo, 160-8582, Japan
| | - Takuji Iwamoto
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-Ku, Tokyo, 160-8582, Japan.
| |
Collapse
|
6
|
Sharifi M, Salehi M, Ebrahimi-Barough S, Alizadeh M, Jahromi HK, Kamalabadi-Farahani M. Synergic effects of core-shell nanospheres and magnetic field for sciatic nerve regeneration in decellularized artery conduits with Schwann cells. J Nanobiotechnology 2024; 22:776. [PMID: 39696412 DOI: 10.1186/s12951-024-03048-5] [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: 10/09/2024] [Accepted: 11/29/2024] [Indexed: 12/20/2024] Open
Abstract
Numerous conduits have been developed to improve peripheral nerve regeneration. However, challenges remain, including remote control of conduit function, and programmed cell behaviors like orientation. We synthesized Fe3O4-MnO2@Zirconium-based Metal-organic frameworks@Retinoic acid (FMZMR) core-shell and assessed their impact on Schwann cell function and behavior within conduits made from decellularized human umbilical arteries (DHUCA) under magnetic field (MF). FMZMR core-shell, featuring a spherical porous structure and catalytic properties, effectively scavenges radicals and facilitates controlled drug release under MF. The histology of the DHUCA indicates effective decellularization with adequate tensile strength and Young's modulus for sciatic nerve regeneration. In-vitro results demonstrate that FMZMR core-shell is biocompatible and promotes Schwann cell proliferation through remotely controlled drug release. Furthermore, its synergy with MF enhances cell orientation and increases neurite length by ~ 1.93-fold. Functional and histological evaluations indicate that the FMZMR core-shell combined with MF promotes nerve regeneration, decreases muscle atrophy, and enhances new neuron growth and myelin formation, without negatively affecting vital tissues. This study suggests that the synergistic effect of FMZMR core-shell with MF can alleviate some of the treatment challenges.
Collapse
Affiliation(s)
- Majid Sharifi
- Student Research Committee, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Majid Salehi
- Tissue Engineering and Stem Cells Research Center, Shahroud University of Medical Sciences, Shahroud, Iran
- Department of Tissue Engineering, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Somayeh Ebrahimi-Barough
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Morteza Alizadeh
- Department of Tissue Engineering and Biomaterials, School of Advanced Medical Sciences and Technologies, Hamadan University of Medical Sciences, Hamadan, Iran
| | | | - Mohammad Kamalabadi-Farahani
- Tissue Engineering and Stem Cells Research Center, Shahroud University of Medical Sciences, Shahroud, Iran.
- Department of Tissue Engineering, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran.
| |
Collapse
|
7
|
Nawrotek K, Chyb M, Gatkowska J, Rudnicka K, Michlewska S, Jóźwiak P. Effect of sodium L-lactate on bioactive properties of chitosan-hydroxyapatite/polycaprolactone conduits for peripheral nerve tissue engineering. Int J Biol Macromol 2024; 281:136254. [PMID: 39366606 DOI: 10.1016/j.ijbiomac.2024.136254] [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: 02/22/2024] [Revised: 09/26/2024] [Accepted: 10/01/2024] [Indexed: 10/06/2024]
Abstract
Biomaterials and synthetic polymers have been widely used to replicate the regenerative microenvironment of the peripheral nervous system. Chitosan-based conduits have shown promise in the regeneration of nerve injuries. However, to mimic the regenerative microenvironment, the scaffold structure should possess bioactive properties. This can be achieved by the incorporation of biomolecules (e.g., proteins, peptides) or trophic factors that should preferably be aligned and/or released with controlled kinetics to activate the process of positive axon chemotaxis. In this study, sodium L-lactate has been used to enhance the bioactive properties of chitosan-hydroxyapatite/polycaprolactone electrodeposits. Next, two methods have been developed to incorporate NGF-loaded microspheres - Method 1 involves entrapment and co-deposition of NGF-loaded microspheres, while Method 2 is based on absorption of NGF-loaded microspheres. The study shows that modification of chitosan-hydroxyapatite/polycaprolactone conduits by sodium L-lactate significantly improves their bioactive, biological, and physicochemical properties. The obtained implants are cytocompatible, enhancing the neurite regeneration process by stimulating its elongation. The absorption of NGF-loaded microspheres into the conduit structure may be considered more favorable for the stimulation of axonal elongation compared to entrapment, as it allows for trophic factor dose-dependent controlled release. The developed conduits possess properties essential for the successful treatment of peripheral nerve discontinuities.
Collapse
Affiliation(s)
- Katarzyna Nawrotek
- Lodz University of Technology, Faculty of Process and Environmental Engineering, Department of Environmental Engineering, Wolczanska 213, 93-005 Lodz, Poland; Lodz University of Technology, International Centre for Research on Innovative Bio-based Materials, 2/22 Stefanowskiego, 90-537, Poland; Warsaw University of Technology, Centre for Advanced Materials and Technology (CEZAMAT), 19 Poleczki, 02-822 Warsaw, Poland.
| | - Maciej Chyb
- University of Lodz, Faculty of Biology and Environmental Protection, Department of Molecular Microbiology, 12/16 Banacha, 90-237 Lodz, Poland; Bio-Med-Chem Doctoral School of the University of Lodz and Lodz Institutes of the Polish Academy of Sciences Banacha 12/16, 90-237 Lodz, Poland.
| | - Justyna Gatkowska
- University of Lodz, Faculty of Biology and Environmental Protection, Department of Molecular Microbiology, 12/16 Banacha, 90-237 Lodz, Poland.
| | - Karolina Rudnicka
- University of Lodz, Faculty of Biology and Environmental Protection, Department of Immunology and Infectious Biology, 90-237 Lodz, Poland.
| | - Sylwia Michlewska
- University of Lodz, Faculty of Biology and Environmental Protection, Laboratory of Microscopic Imaging and Specialized Biological Techniques, Banacha 12/16, 90-237 Lodz, Poland.
| | - Piotr Jóźwiak
- University of Lodz, Faculty of Biology and Environmental Protection, Department of Invertebrates Zoology and Hydrobiology, Banacha 12/16, 90-324 Lodz, Poland.
| |
Collapse
|
8
|
Wang X, Chen S, Chen X, Wu J, Huang Z, Wang J, Chen F, Liu C. Biomimetic multi-channel nerve conduits with micro/nanostructures for rapid nerve repair. Bioact Mater 2024; 41:577-596. [PMID: 39257673 PMCID: PMC11384339 DOI: 10.1016/j.bioactmat.2024.07.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 07/13/2024] [Accepted: 07/13/2024] [Indexed: 09/12/2024] Open
Abstract
Peripheral nervous system (PNS) injuries often lead to significant sensory and motor impairments. Traditional artificial nerve conduits, lacking anisotropic structures, have been associated with prolonged repair time and failures in nerve regeneration. This study aimed to address these challenges by developing a novel approach for rapid repair of peripheral nerve injuries (PNI). A 3D oriented fibers scaffold featuring distinct radial (RFs) and longitudinal (LFs) fibers orientations was engineered using coaxial electrospinning and gas directional foaming techniques. This scaffold was then integrated with a shape memory conduit to form a directional multi-channel nerve conduit with micro/nanostructures. The results revealed that the grooved surface of the fibers significantly improved cellular directional guidance, effectively facilitating the migration of SCs from the periphery towards the center and from the base to the apex of the scaffold. In a rat model with a 10 mm nerve defect, the ND-PLATMC/LF ND-PCL scaffold significantly enhanced nerve regeneration and motor function recovery within 4 weeks. These results suggest the potential of this innovative scaffold for efficient repair of the nerve injuries.
Collapse
Affiliation(s)
- Xinqing Wang
- Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Shuo Chen
- Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Xiaolei Chen
- Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Juan Wu
- Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Zhenhua Huang
- Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Jing Wang
- Key Laboratory for Ultrafine Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
- Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Fangping Chen
- Key Laboratory for Ultrafine Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
- Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Changsheng Liu
- Key Laboratory for Ultrafine Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
- Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China
| |
Collapse
|
9
|
Hattori Y, Takeda S, Usami T, Shibata R, Takahashi H, Joyo Y, Kawaguchi Y, Okamoto H, Murakami H, Paholpak P, Ota H. Tensile Strength of Nerve Bridging Models Using Collagen Nerve Conduits. J Reconstr Microsurg 2024. [PMID: 39142345 DOI: 10.1055/a-2387-3282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/16/2024]
Abstract
BACKGROUND In the treatment of peripheral nerve injuries with nerve defects, second-generation collagen-based conduits, such as Renerve® (Nipro, Osaka, Japan), have shown the potential for promoting nerve regeneration. However, there is concern related to the weak material properties. No previous studies have addressed the strength of the bridging model using collagen conduits. This study aimed to investigate the tensile strength and failure patterns in nerve defect models bridged with Renerve® conduits through biomechanical research. METHODS Using fresh chicken sciatic nerves, we examined the maximum failure load of four groups: bridging models using Renerve® with one suture (group A), with two sutures (group B), with three sutures (group C), and end-to-end neurorrhaphy models with two sutures (group N). Each group had eight specimens. We also evaluated failure patterns of the specimens. RESULTS Group N showed a significantly higher maximum failure load (0.96 ± 0.13 N) compared to groups A (0.23 ± 0.06 N, p < 0.0001), B (0.29 ± 0.05 N, p < 0.0001), and C (0.40 ± 0.10 N, p < 0.0001). Regarding failure patterns, all specimens in group A showed nerve-end dislocation from the conduit. Two specimens in group B and three specimens in group C failed due to circumferential cracks in the conduit. Six specimens in group B and five specimens in group C exhibited cutting out of sutures from the conduit. CONCLUSION This study suggests that the number of sutures in synthetic collagen nerve conduits has little effect on the maximum failure load. To take advantage of its biomaterial benefits, a period of postoperative range of motion restriction may be required.
Collapse
Affiliation(s)
- Yusuke Hattori
- Department of Orthopedic Surgery, Nagoya City University Graduate School of Medical Science, Nagoya, Japan
- Department of Orthopedic Surgery, Nagoya City University East Medical Center, Nagoya, Japan
| | - Shinsuke Takeda
- Department of Orthopedic Surgery, Nagoya City University Graduate School of Medical Science, Nagoya, Japan
| | - Takuya Usami
- Department of Orthopedic Surgery, Nagoya City University Graduate School of Medical Science, Nagoya, Japan
| | - Ryutaro Shibata
- Department of Orthopedic Surgery, Nagoya City University Graduate School of Medical Science, Nagoya, Japan
| | - Hiroshi Takahashi
- Department of Orthopedic Surgery, Nagoya City University Graduate School of Medical Science, Nagoya, Japan
| | - Yuji Joyo
- Department of Orthopedic Surgery, Nagoya City University East Medical Center, Nagoya, Japan
| | - Yohei Kawaguchi
- Department of Orthopedic Surgery, Nagoya City University Graduate School of Medical Science, Nagoya, Japan
| | - Hideki Okamoto
- Department of Orthopedic Surgery, Nagoya City University Graduate School of Medical Science, Nagoya, Japan
| | - Hideki Murakami
- Department of Orthopedic Surgery, Nagoya City University Graduate School of Medical Science, Nagoya, Japan
| | - Permsak Paholpak
- Department of Orthopedics, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Hideyuki Ota
- Department of Orthopedic Surgery and Hand Surgery, Nagoya Ekisaikai Hospital, Nagoya, Japan
| |
Collapse
|
10
|
Buckley C, Montgomery TR, Szank T, Major I. Hyaluronic acid hybrid formulations optimised for 3D printing of nerve conduits and the delivery of the novel neurotrophic-like compound tyrosol to enhance peripheral nerve regeneration via Schwann cell proliferation. Int J Pharm 2024; 661:124477. [PMID: 39013530 DOI: 10.1016/j.ijpharm.2024.124477] [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: 02/28/2024] [Revised: 07/11/2024] [Accepted: 07/13/2024] [Indexed: 07/18/2024]
Abstract
Peripheral nerve injuries, predominantly affecting individuals aged 20-40, pose significant healthcare challenges, with current surgical methods often failing to achieve complete functional recovery. This study focuses on the development of 3D printed hydrogel nerve conduits using modified hyaluronic acid (HA) for potentially enhancing peripheral nerve regeneration. Hyaluronic acid was chemically altered with cysteamine HCl and methacrylic anhydride to create thiolated HA (HA-SH) and methacrylated HA (HA-MA), achieving a modification degree of approximately 20 %. This modification was crucial to maintain the receptor interaction of HA. The modified HA was rigorously tested to ensure cytocompatibility in neuronal and glial cell lines. Subsequently, various 3D printed HA formulations were evaluated, focusing on improving HA's inherent mechanical weaknesses. These formulations were assessed for cytotoxicity through direct contact and elution extract testing, confirming their safety over a 24-h period. Among the neurotrophic compounds tested, Tyrosol emerged as the most effective in promoting Schwann cell proliferation in vitro. The 3D printed HA system demonstrated proficiency in loading and releasing Tyrosol at physiological pH. The findings from this research highlight the promising role of 3D printed HA and Tyrosol in the field of nerve tissue engineering, offering a novel approach to peripheral nerve regeneration.
Collapse
Affiliation(s)
- Ciara Buckley
- PRISM Research Institute, Technological University of the Shannon, Athlone N37 HD68, Ireland
| | - Therese R Montgomery
- School of Science and Computing, Atlantic Technological University, Galway H91 T8NW, Ireland
| | - Tomasz Szank
- Bioscience Research Institute, Technological University of the Shannon, Athlone N37 HD68, Ireland
| | - Ian Major
- PRISM Research Institute, Technological University of the Shannon, Athlone N37 HD68, Ireland.
| |
Collapse
|
11
|
Wong GC, Chung KC. Bioengineered Nerve Conduits and Wraps. Hand Clin 2024; 40:379-387. [PMID: 38972682 DOI: 10.1016/j.hcl.2024.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/09/2024]
Abstract
Peripheral nerve injuries are prevalent and their treatments present significant challenges. Among the various reconstructive options, nerve conduits and wraps are popular choices. Advances in bioengineering and regenerative medicine have led to the development of new biocompatible materials and implant designs that offer the potential for enhanced neural recovery. Cost, nerve injury type, and implant size must be considered when deciding on the ideal reconstructive option.
Collapse
Affiliation(s)
- Gordon C Wong
- University of Michigan Comprehensive Hand Center, Michigan Medicine, 1500 East Medical Center Drive, 2130 Taubman Center, SPC 5340, Ann Arbor, MI 48109, USA
| | - Kevin C Chung
- University of Michigan Comprehensive Hand Center, Michigan Medicine, 1500 East Medical Center Drive, 2130 Taubman Center, SPC 5340, Ann Arbor, MI 48109, USA.
| |
Collapse
|
12
|
Crabtree JR, Mulenga CM, Tran K, Feinberg K, Santerre JP, Borschel GH. Biohacking Nerve Repair: Novel Biomaterials, Local Drug Delivery, Electrical Stimulation, and Allografts to Aid Surgical Repair. Bioengineering (Basel) 2024; 11:776. [PMID: 39199733 PMCID: PMC11352148 DOI: 10.3390/bioengineering11080776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 07/15/2024] [Accepted: 07/26/2024] [Indexed: 09/01/2024] Open
Abstract
The regenerative capacity of the peripheral nervous system is limited, and peripheral nerve injuries often result in incomplete healing and poor outcomes even after repair. Transection injuries that induce a nerve gap necessitate microsurgical intervention; however, even the current gold standard of repair, autologous nerve graft, frequently results in poor functional recovery. Several interventions have been developed to augment the surgical repair of peripheral nerves, and the application of functional biomaterials, local delivery of bioactive substances, electrical stimulation, and allografts are among the most promising approaches to enhance innate healing across a nerve gap. Biocompatible polymers with optimized degradation rates, topographic features, and other functions provided by their composition have been incorporated into novel nerve conduits (NCs). Many of these allow for the delivery of drugs, neurotrophic factors, and whole cells locally to nerve repair sites, mitigating adverse effects that limit their systemic use. The electrical stimulation of repaired nerves in the perioperative period has shown benefits to healing and recovery in human trials, and novel biomaterials to enhance these effects show promise in preclinical models. The use of acellular nerve allografts (ANAs) circumvents the morbidity of donor nerve harvest necessitated by the use of autografts, and improvements in tissue-processing techniques may allow for more readily available and cost-effective options. Each of these interventions aid in neural regeneration after repair when applied independently, and their differing forms, benefits, and methods of application present ample opportunity for synergistic effects when applied in combination.
Collapse
Affiliation(s)
- Jordan R. Crabtree
- Division of Plastic Surgery, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Chilando M. Mulenga
- Division of Plastic Surgery, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Khoa Tran
- Division of Plastic Surgery, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Konstantin Feinberg
- Division of Plastic Surgery, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - J. Paul Santerre
- Institute of Biomedical Engineering, University of Toronto, 164 College St Room 407, Toronto, ON M5S 3G9, Canada
| | - Gregory H. Borschel
- Division of Plastic Surgery, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
13
|
Sharifi M, Kamalabadi-Farahani M, Salehi M, Ebrahimi-Brough S, Alizadeh M. Recent perspectives on the synergy of mesenchymal stem cells with micro/nano strategies in peripheral nerve regeneration-a review. Front Bioeng Biotechnol 2024; 12:1401512. [PMID: 39050683 PMCID: PMC11266111 DOI: 10.3389/fbioe.2024.1401512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 06/19/2024] [Indexed: 07/27/2024] Open
Abstract
Despite the intrinsic repair of peripheral nerve injury (PNI), it is important to carefully monitor the process of peripheral nerve repair, as peripheral nerve regeneration is slow and incomplete in large traumatic lesions. Hence, mesenchymal stem cells (MSCs) with protective and regenerative functions are utilized in synergy with innovative micro/nano technologies to enhance the regeneration process of peripheral nerves. Nonetheless, as MSCs are assessed using standard regenerative criteria including sensory-motor indices, structural features, and morphology, it is challenging to differentiate between the protective and regenerative impacts of MSCs on neural tissue. This study aims to analyze the process of nerve regeneration, particularly the performance of MSCs with and without synergistic approaches. It also focuses on the paracrine secretions of MSCs and their conversion into neurons with functional properties that influence nerve regeneration after PNI. Furthermore, the study explores new ideas for nerve regeneration after PNI by considering the synergistic effect of MSCs and therapeutic compounds, neuronal cell derivatives, biological or polymeric conduits, organic/inorganic nanoparticles, and electrical stimulation. Finally, the study highlights the main obstacles to developing synergy in nerve regeneration after PNI and aims to open new windows based on recent advances in neural tissue regeneration.
Collapse
Affiliation(s)
- Majid Sharifi
- Student Research Committee, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
- Tissue Engineering and Stem Cells Research Center, Shahroud University of Medical Sciences, Shahroud, Iran
- Department of Tissue Engineering, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Mohammad Kamalabadi-Farahani
- Tissue Engineering and Stem Cells Research Center, Shahroud University of Medical Sciences, Shahroud, Iran
- Department of Tissue Engineering, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Majid Salehi
- Tissue Engineering and Stem Cells Research Center, Shahroud University of Medical Sciences, Shahroud, Iran
- Department of Tissue Engineering, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
- Health Technology Incubator Center, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Somayeh Ebrahimi-Brough
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Morteza Alizadeh
- Department of Tissue Engineering and Biomaterials, School of Advanced Medical Sciences and Technologies, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
14
|
Hu C, Liu B, Huang X, Wang Z, Qin K, Sun L, Fan Z. Sea Cucumber-Inspired Microneedle Nerve Guidance Conduit for Synergistically Inhibiting Muscle Atrophy and Promoting Nerve Regeneration. ACS NANO 2024; 18:14427-14440. [PMID: 38776414 DOI: 10.1021/acsnano.4c00794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
Muscle atrophy resulting from peripheral nerve injury (PNI) poses a threat to a patient's mobility and sensitivity. However, an effective method to inhibit muscle atrophy following PNI remains elusive. Drawing inspiration from the sea cucumber, we have integrated microneedles (MNs) and microchannel technology into nerve guidance conduits (NGCs) to develop bionic microneedle NGCs (MNGCs) that emulate the structure and piezoelectric function of sea cucumbers. Morphologically, MNGCs feature an outer surface with outward-pointing needle tips capable of applying electrical stimulation to denervated muscles. Simultaneously, the interior contains microchannels designed to guide the migration of Schwann cells (SCs). Physiologically, the incorporation of conductive reduced graphene oxide and piezoelectric zinc oxide nanoparticles into the polycaprolactone scaffold enhances conductivity and piezoelectric properties, facilitating SCs' migration, myelin regeneration, axon growth, and the restoration of neuromuscular function. These combined effects ultimately lead to the inhibition of muscle atrophy and the restoration of nerve function. Consequently, the concept of the synergistic effect of inhibiting muscle atrophy and promoting nerve regeneration has the capacity to transform the traditional approach to PNI repair and find broad applications in PNI repair.
Collapse
Affiliation(s)
- Cewen Hu
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing of Gansu Province, School of Stomatology, Lanzhou University, Lanzhou 730000, People's Republic of China
| | - Bin Liu
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing of Gansu Province, School of Stomatology, Lanzhou University, Lanzhou 730000, People's Republic of China
| | - Xinyue Huang
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing of Gansu Province, School of Stomatology, Lanzhou University, Lanzhou 730000, People's Republic of China
| | - Zhilong Wang
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing of Gansu Province, School of Stomatology, Lanzhou University, Lanzhou 730000, People's Republic of China
| | - Kaiqi Qin
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing of Gansu Province, School of Stomatology, Lanzhou University, Lanzhou 730000, People's Republic of China
| | - Luyi Sun
- Polymer Program, Institute of Materials Science and Department of Chemical & Biomolecular Engineering, University of Connecticut, Storrs, Connecticut 06269, United States
| | - Zengjie Fan
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing of Gansu Province, School of Stomatology, Lanzhou University, Lanzhou 730000, People's Republic of China
| |
Collapse
|
15
|
Sun R, Lang Y, Chang MW, Zhao M, Li C, Liu S, Wang B. Leveraging Oriented Lateral Walls of Nerve Guidance Conduit with Core-Shell MWCNTs Fibers for Peripheral Nerve Regeneration. Adv Healthc Mater 2024; 13:e2303867. [PMID: 38258406 DOI: 10.1002/adhm.202303867] [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: 12/03/2023] [Indexed: 01/24/2024]
Abstract
Peripheral nerve regeneration and functional recovery rely on the chemical, physical, and structural properties of nerve guidance conduits (NGCs). However, the limited support for long-distance nerve regeneration and axonal guidance has hindered the widespread use of NGCs. This study introduces a novel nerve guidance conduit with oriented lateral walls, incorporating multi-walled carbon nanotubes (MWCNTs) within core-shell fibers prepared in a single step using a modified electrohydrodynamic (EHD) printing technique to promote peripheral nerve regeneration. The structured conduit demonstrated exceptional stability, mechanical properties, and biocompatibility, significantly enhancing the functionality of NGCs. In vitro cell studies revealed that RSC96 cells adhered and proliferated effectively along the oriented fibers, demonstrating a favorable response to the distinctive architectures and properties. Subsequently, a rat sciatic nerve injury model demonstrated effective efficacy in promoting peripheral nerve regeneration and functional recovery. Tissue analysis and functional testing highlighted the significant impact of MWCNT concentration in enhancing peripheral nerve regeneration and confirming well-matured aligned axonal growth, muscle recovery, and higher densities of myelinated axons. These findings demonstrate the potential of oriented lateral architectures with coaxial MWCNT fibers as a promising approach to support long-distance regeneration and encourage directional nerve growth for peripheral nerve repair in clinical applications.
Collapse
Affiliation(s)
- Renyuan Sun
- State Key Laboratory of Reliability and Intelligence of Electrical Equipment, Tianjin Key Laboratory of Bio-Electromagnetic and Neural Engineering, Hebei Key Laboratory of Bioelectromagnetics and Neuroengineering, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin, 300132, China
| | - Yuna Lang
- State Key Laboratory of Reliability and Intelligence of Electrical Equipment, Tianjin Key Laboratory of Bio-Electromagnetic and Neural Engineering, Hebei Key Laboratory of Bioelectromagnetics and Neuroengineering, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin, 300132, China
| | - Ming-Wei Chang
- Nanotechnology and Integrated Bioengineering Centre, University of Ulster, Belfast, BT15 1AP, UK
| | - Mingkang Zhao
- State Key Laboratory of Reliability and Intelligence of Electrical Equipment, Tianjin Key Laboratory of Bio-Electromagnetic and Neural Engineering, Hebei Key Laboratory of Bioelectromagnetics and Neuroengineering, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin, 300132, China
| | - Chao Li
- State Key Laboratory of Reliability and Intelligence of Electrical Equipment, Tianjin Key Laboratory of Bio-Electromagnetic and Neural Engineering, Hebei Key Laboratory of Bioelectromagnetics and Neuroengineering, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin, 300132, China
| | - Shiheng Liu
- State Key Laboratory of Reliability and Intelligence of Electrical Equipment, Tianjin Key Laboratory of Bio-Electromagnetic and Neural Engineering, Hebei Key Laboratory of Bioelectromagnetics and Neuroengineering, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin, 300132, China
| | - Baolin Wang
- State Key Laboratory of Reliability and Intelligence of Electrical Equipment, Tianjin Key Laboratory of Bio-Electromagnetic and Neural Engineering, Hebei Key Laboratory of Bioelectromagnetics and Neuroengineering, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin, 300132, China
| |
Collapse
|
16
|
Jafarisavari Z, Ai J, Abbas Mirzaei S, Soleimannejad M, Asadpour S. Development of new nanofibrous nerve conduits by PCL-Chitosan-Hyaluronic acid containing Piracetam-Vitamin B12 for sciatic nerve: A rat model. Int J Pharm 2024; 655:123978. [PMID: 38458406 DOI: 10.1016/j.ijpharm.2024.123978] [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: 12/28/2023] [Revised: 03/04/2024] [Accepted: 03/05/2024] [Indexed: 03/10/2024]
Abstract
Peripheral nerve injury is a critical condition that can disrupt nerve functions. Despite the progress in engineering artificial nerve guidance conduits (NGCs), nerve regeneration remains challenging. Here, we developed new nanofibrous NGCs using polycaprolactone (PCL) and chitosan (CH) containing piracetam (PIR)/vitamin B12(VITB12) with an electrospinning method. The lumen of NGCs was coated by hyaluronic acid (HA) to promote regeneration in sciatic nerve injury. The NGCs were characterized via Scanning Electron Microscopy (SEM), Fourier transform infrared (FTIR), tensile, swelling, contact angle, degradation, and drug release tests. Neuronal precursor cell line (PCL12 cell) and rat mesenchymal stem cells derived from bone marrow (MSCs) were seeded on the nanofibrous conduits. After that, the biocompatibility of the NGCs was evaluated by the 2,5-diphenyl-2H-tetrazolium bromide (MTT) assay, 4',6-diamidino-2-phenylindole (DAPI) staining, and SEM images. The SEM demonstrated that PCL/CH/PIR/VITB12 NGCs had nonaligned, interconnected, smooth fibers. The mechanical properties of these NGCs were similar to rat sciatic nerve. These conduits had an appropriate swelling and degradation rate. The In Vitro studies exhibited favorable biocompatibility of the PCL/CH/PIR/VITB12 NGCs towards PC12 cells and MSCs. The in vitro studies exhibited favorable biocompatibility of the PCL/CH/PIR/VIT B12 NGCs towards MSCs and PC12 cells. To analyze functional efficacy, NGCs were implanted into a 10 mm Wistar rat sciatic nerve gap and bridged the proximal and distal stump of the defect. After three months, the results of sciatic functional index (55.3 ± 1.8), hot plate latency test (5.6 ± 0.5 s), gastrocnemius muscle wet weight-loss (38.57 ± 1.6 %) and histopathological examination using hematoxylin-eosin (H&E) /toluidine blue/ Anti-Neurofilament (NF200) staining demonstrated that the produced conduit recovered motor and sensory functions and had comparable nerve regeneration compared to the autograft that can be as the gold standard to bridge the nerve gaps.
Collapse
Affiliation(s)
- Zahra Jafarisavari
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Jafar Ai
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Abbas Mirzaei
- Department of Medical Biotechnology, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Mostafa Soleimannejad
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Shiva Asadpour
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran; Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran.
| |
Collapse
|
17
|
Zhou W, Rahman MSU, Sun C, Li S, Zhang N, Chen H, Han CC, Xu S, Liu Y. Perspectives on the Novel Multifunctional Nerve Guidance Conduits: From Specific Regenerative Procedures to Motor Function Rebuilding. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2307805. [PMID: 37750196 DOI: 10.1002/adma.202307805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 09/19/2023] [Indexed: 09/27/2023]
Abstract
Peripheral nerve injury potentially destroys the quality of life by inducing functional movement disorders and sensory capacity loss, which results in severe disability and substantial psychological, social, and financial burdens. Autologous nerve grafting has been commonly used as treatment in the clinic; however, its rare donor availability limits its application. A series of artificial nerve guidance conduits (NGCs) with advanced architectures are also proposed to promote injured peripheral nerve regeneration, which is a complicated process from axon sprouting to targeted muscle reinnervation. Therefore, exploring the interactions between sophisticated NGC complexes and versatile cells during each process including axon sprouting, Schwann cell dedifferentiation, nerve myelination, and muscle reinnervation is necessary. This review highlights the contribution of functional NGCs and the influence of microscale biomaterial architecture on biological processes of nerve repair. Progressive NGCs with chemical molecule induction, heterogenous topographical morphology, electroactive, anisotropic assembly microstructure, and self-powered electroactive and magnetic-sensitive NGCs are also collected, and they are expected to be pioneering features in future multifunctional and effective NGCs.
Collapse
Affiliation(s)
- Weixian Zhou
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Muhammad Saif Ur Rahman
- Institute for Advanced Study, Shenzhen University, Shenzhen, 518060, P. R. China
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education Guangdong province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen, 518060, P. R. China
| | - Chengmei Sun
- Institute for Advanced Study, Shenzhen University, Shenzhen, 518060, P. R. China
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education Guangdong province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen, 518060, P. R. China
| | - Shilin Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Nuozi Zhang
- Institute for Advanced Study, Shenzhen University, Shenzhen, 518060, P. R. China
| | - Hao Chen
- Institute for Advanced Study, Shenzhen University, Shenzhen, 518060, P. R. China
| | - Charles C Han
- Institute for Advanced Study, Shenzhen University, Shenzhen, 518060, P. R. China
- Materials Science and Engineering, University of Maryland, College Park, MD, 20742, USA
| | - Shanshan Xu
- Institute for Advanced Study, Shenzhen University, Shenzhen, 518060, P. R. China
- Materials Science and Engineering, University of Maryland, College Park, MD, 20742, USA
| | - Ying Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| |
Collapse
|
18
|
Shlapakova LE, Surmeneva MA, Kholkin AL, Surmenev RA. Revealing an important role of piezoelectric polymers in nervous-tissue regeneration: A review. Mater Today Bio 2024; 25:100950. [PMID: 38318479 PMCID: PMC10840125 DOI: 10.1016/j.mtbio.2024.100950] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 12/12/2023] [Accepted: 01/08/2024] [Indexed: 02/07/2024] Open
Abstract
Nerve injuries pose a drastic threat to nerve mobility and sensitivity and lead to permanent dysfunction due to low regenerative capacity of mature neurons. The electrical stimuli that can be provided by electroactive materials are some of the most effective tools for the formation of soft tissues, including nerves. Electric output can provide a distinctly favorable bioelectrical microenvironment, which is especially relevant for the nervous system. Piezoelectric biomaterials have attracted attention in the field of neural tissue engineering owing to their biocompatibility and ability to generate piezoelectric surface charges. In this review, an outlook of the most recent achievements in the field of piezoelectric biomaterials is described with an emphasis on piezoelectric polymers for neural tissue engineering. First, general recommendations for the design of an optimal nerve scaffold are discussed. Then, specific mechanisms determining nerve regeneration via piezoelectric stimulation are considered. Activation of piezoelectric responses via natural body movements, ultrasound, and magnetic fillers is also examined. The use of magnetoelectric materials in combination with alternating magnetic fields is thought to be the most promising due to controllable reproducible cyclic deformations and deep tissue permeation by magnetic fields without tissue heating. In vitro and in vivo applications of nerve guidance scaffolds and conduits made of various piezopolymers are reviewed too. Finally, challenges and prospective research directions regarding piezoelectric biomaterials promoting nerve regeneration are discussed. Thus, the most relevant scientific findings and strategies in neural tissue engineering are described here, and this review may serve as a guideline both for researchers and clinicians.
Collapse
Affiliation(s)
- Lada E. Shlapakova
- Physical Materials Science and Composite Materials Center, Research School of Chemistry & Applied Biomedical Sciences, National Research Tomsk Polytechnic University, Tomsk, 634050, Russia
| | - Maria A. Surmeneva
- Physical Materials Science and Composite Materials Center, Research School of Chemistry & Applied Biomedical Sciences, National Research Tomsk Polytechnic University, Tomsk, 634050, Russia
- Piezo- and Magnetoelectric Materials Research & Development Centre, Research School of Chemistry & Applied Biomedical Sciences, National Research Tomsk Polytechnic University, 634050, Tomsk, Russia
| | - Andrei L. Kholkin
- Piezo- and Magnetoelectric Materials Research & Development Centre, Research School of Chemistry & Applied Biomedical Sciences, National Research Tomsk Polytechnic University, 634050, Tomsk, Russia
- Department of Physics & CICECO - Aveiro Institute of Materials, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Roman A. Surmenev
- Physical Materials Science and Composite Materials Center, Research School of Chemistry & Applied Biomedical Sciences, National Research Tomsk Polytechnic University, Tomsk, 634050, Russia
- Piezo- and Magnetoelectric Materials Research & Development Centre, Research School of Chemistry & Applied Biomedical Sciences, National Research Tomsk Polytechnic University, 634050, Tomsk, Russia
| |
Collapse
|
19
|
Liu B, Alimi OA, Wang Y, Kong Y, Kuss M, Krishnan MA, Hu G, Xiao Y, Dong J, DiMaio DJ, Duan B. Differentiated mesenchymal stem cells-derived exosomes immobilized in decellularized sciatic nerve hydrogels for peripheral nerve repair. J Control Release 2024; 368:24-41. [PMID: 38367864 PMCID: PMC11411504 DOI: 10.1016/j.jconrel.2024.02.019] [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: 11/23/2023] [Revised: 01/31/2024] [Accepted: 02/12/2024] [Indexed: 02/19/2024]
Abstract
Peripheral nerve injury (PNI) and the limitations of current treatments often result in incomplete sensory and motor function recovery, which significantly impact the patient's quality of life. While exosomes (Exo) derived from stem cells and Schwann cells have shown promise on promoting PNI repair following systemic administration or intraneural injection, achieving effective local and sustained Exo delivery holds promise to treat local PNI and remains challenging. In this study, we developed Exo-loaded decellularized porcine nerve hydrogels (DNH) for PNI repair. We successfully isolated Exo from differentiated human adipose-derived mesenchymal stem cells (hADMSC) with a Schwann cell-like phenotype (denoted as dExo). These dExo were further combined with polyethylenimine (PEI), and DNH to create polyplex hydrogels (dExo-loaded pDNH). At a PEI content of 0.1%, pDNH showed cytocompatibility for hADMSCs and supported neurite outgrowth of dorsal root ganglions. The sustained release of dExos from dExo-loaded pDNH persisted for at least 21 days both in vitro and in vivo. When applied around injured nerves in a mouse sciatic nerve crush injury model, the dExo-loaded pDNH group significantly improved sensory and motor function recovery and enhanced remyelination compared to dExo and pDNH only groups, highlighting the synergistic regenerative effects. Interestingly, we observed a negative correlation between the number of colony-stimulating factor-1 receptor (CSF-1R) positive cells and the extent of PNI regeneration at the 21-day post-surgery stage. Subsequent in vitro experiments demonstrated the potential involvement of the CSF-1/CSF-1R axis in Schwann cells and macrophage interaction, with dExo effectively downregulating CSF-1/CSF-1R signaling.
Collapse
Affiliation(s)
- Bo Liu
- Mary & Dick Holland Regenerative Medicine Program and Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA; Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Olawale A Alimi
- Mary & Dick Holland Regenerative Medicine Program and Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA; Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Yanfei Wang
- Mary & Dick Holland Regenerative Medicine Program and Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA; Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA; College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Erie, PA 16509, USA
| | - Yunfan Kong
- Mary & Dick Holland Regenerative Medicine Program and Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA; Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Mitchell Kuss
- Mary & Dick Holland Regenerative Medicine Program and Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA; Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Mena Asha Krishnan
- Mary & Dick Holland Regenerative Medicine Program and Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA; Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Guoku Hu
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Yi Xiao
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Jixin Dong
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Dominick J DiMaio
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Bin Duan
- Mary & Dick Holland Regenerative Medicine Program and Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA; Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA; Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE 68588, USA.
| |
Collapse
|
20
|
Mayrhofer-Schmid M, Aman M, Panayi AC, Raasveld FV, Kneser U, Eberlin KR, Harhaus L, Böcker A. Fibrin Glue Coating Limits Scar Tissue Formation around Peripheral Nerves. Int J Mol Sci 2024; 25:3687. [PMID: 38612497 PMCID: PMC11011750 DOI: 10.3390/ijms25073687] [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: 02/25/2024] [Revised: 03/20/2024] [Accepted: 03/25/2024] [Indexed: 04/14/2024] Open
Abstract
Scar tissue formation presents a significant barrier to peripheral nerve recovery in clinical practice. While different experimental methods have been described, there is no clinically available gold standard for its prevention. This study aims to determine the potential of fibrin glue (FG) to limit scarring around peripheral nerves. Thirty rats were divided into three groups: glutaraldehyde-induced sciatic nerve injury treated with FG (GA + FG), sciatic nerve injury with no treatment (GA), and no sciatic nerve injury (Sham). Neural regeneration was assessed with weekly measurements of the visual static sciatic index as a parameter for sciatic nerve function across a 12-week period. After 12 weeks, qualitative and quantitative histological analysis of scar tissue formation was performed. Furthermore, histomorphometric analysis and wet muscle weight analysis were performed after the postoperative observation period. The GA + FG group showed a faster functional recovery (6 versus 9 weeks) compared to the GA group. The FG-treated group showed significantly lower perineural scar tissue formation and significantly higher fiber density, myelin thickness, axon thickness, and myelinated fiber thickness than the GA group. A significantly higher wet muscle weight ratio of the tibialis anterior muscle was found in the GA + FG group compared to the GA group. Our results suggest that applying FG to injured nerves is a promising scar tissue prevention strategy associated with improved regeneration both at the microscopic and at the functional level. Our results can serve as a platform for innovation in the field of perineural regeneration with immense clinical potential.
Collapse
Affiliation(s)
- Maximilian Mayrhofer-Schmid
- Department of Hand-, Plastic and Reconstructive Surgery, Burn Center, BG Trauma Center Ludwigshafen, Department of Hand- and Plastic Surgery, University of Heidelberg, 69120 Heidelberg, Germany
- Hand and Arm Center, Department of Orthopaedic Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02215, USA
| | - Martin Aman
- Department of Hand-, Plastic and Reconstructive Surgery, Burn Center, BG Trauma Center Ludwigshafen, Department of Hand- and Plastic Surgery, University of Heidelberg, 69120 Heidelberg, Germany
| | - Adriana C. Panayi
- Department of Hand-, Plastic and Reconstructive Surgery, Burn Center, BG Trauma Center Ludwigshafen, Department of Hand- and Plastic Surgery, University of Heidelberg, 69120 Heidelberg, Germany
| | - Floris V. Raasveld
- Hand and Arm Center, Department of Orthopaedic Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02215, USA
- Department of Plastic, Reconstructive and Hand Surgery, Erasmus Medical Center, Erasmus University, 3015 GD Rotterdam, The Netherlands
| | - Ulrich Kneser
- Department of Hand-, Plastic and Reconstructive Surgery, Burn Center, BG Trauma Center Ludwigshafen, Department of Hand- and Plastic Surgery, University of Heidelberg, 69120 Heidelberg, Germany
| | - Kyle R. Eberlin
- Division of Plastic and Reconstructive Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02215, USA
| | - Leila Harhaus
- Department of Hand-, Plastic and Reconstructive Surgery, Burn Center, BG Trauma Center Ludwigshafen, Department of Hand- and Plastic Surgery, University of Heidelberg, 69120 Heidelberg, Germany
- Department of Hand Surgery, Peripheral Nerve Surgery and Rehabilitation, BG Trauma Center Ludwigshafen, 67071 Ludwigshafen, Germany
| | - Arne Böcker
- Department of Hand-, Plastic and Reconstructive Surgery, Burn Center, BG Trauma Center Ludwigshafen, Department of Hand- and Plastic Surgery, University of Heidelberg, 69120 Heidelberg, Germany
- Department of Hand Surgery, Peripheral Nerve Surgery and Rehabilitation, BG Trauma Center Ludwigshafen, 67071 Ludwigshafen, Germany
| |
Collapse
|
21
|
Castro VO, Livi S, Sperling LE, Dos Santos MG, Merlini C. Biodegradable Electrospun Conduit with Aligned Fibers Based on Poly(lactic- co-glycolic Acid) (PLGA)/Carbon Nanotubes and Choline Bitartrate Ionic Liquid. ACS APPLIED BIO MATERIALS 2024; 7:1536-1546. [PMID: 38346264 DOI: 10.1021/acsabm.3c00980] [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] [Indexed: 03/19/2024]
Abstract
Functionally active aligned fibers are a promising approach to enhance neuro adhesion and guide the extension of neurons for peripheral nerve regeneration. Therefore, the present study developed poly(lactic-co-glycolic acid) (PLGA)-aligned electrospun mats and investigated the synergic effect with carbon nanotubes (CNTs) and Choline Bitartrate ionic liquid (Bio-IL) on PLGA fibers. Morphology, thermal, and mechanical performances were determined as well as the hydrolytic degradation and the cytotoxicity. Results revealed that electrospun mats are composed of highly aligned fibers, and CNTs were aligned and homogeneously distributed into the fibers. Bio-IL changed thermal transition behavior, reduced glass transition temperature (Tg), and favored crystal phase formation. The mechanical properties increased in the presence of CNTs and slightly decreased in the presence of the Bio-IL. The results demonstrated a decrease in the degradation rate in the presence of CNTs, whereas the use of Bio-IL led to an increase in the degradation rate. Cytotoxicity results showed that all the electrospun mats display metabolic activity above 70%, which demonstrates that they are biocompatible. Moreover, superior biocompatibility was observed for the electrospun containing Bio-IL combined with higher amounts of CNTs, showing a high potential to be used in nerve tissue engineering.
Collapse
Affiliation(s)
- Vanessa Oliveira Castro
- Mechanical Engineering Department, Universidade Federal de Santa Catarina (UFSC), Florianópolis, Santa Catarina 88040-535, Brazil
- Université de Lyon, CNRS, Université Claude Bernard Lyon 1, INSA Lyon, Université Jean Monnet, UMR 5223, Ingénierie des Matériaux Polymères, Villeurbanne F-69621 Cédex, France
| | - Sébastien Livi
- Université de Lyon, CNRS, Université Claude Bernard Lyon 1, INSA Lyon, Université Jean Monnet, UMR 5223, Ingénierie des Matériaux Polymères, Villeurbanne F-69621 Cédex, France
| | - Laura Elena Sperling
- Hematology and Stem Cell Laboratory, Faculty of Pharmacy, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul 90610-000, Brazil
| | - Marcelo Garrido Dos Santos
- Hematology and Stem Cell Laboratory, Faculty of Pharmacy, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul 90610-000, Brazil
| | - Claudia Merlini
- Materials Engineering Special Coordination, Universidade Federal de Santa Catarina (UFSC), Blumenau, Santa Catarina 89036-002, Brazil
| |
Collapse
|
22
|
Wang Y, Yang B, Huang Z, Yang Z, Wang J, Ao Q, Yin G, Li Y. Progress and mechanism of graphene oxide-composited materials in application of peripheral nerve repair. Colloids Surf B Biointerfaces 2024; 234:113672. [PMID: 38071946 DOI: 10.1016/j.colsurfb.2023.113672] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 11/21/2023] [Accepted: 11/23/2023] [Indexed: 02/09/2024]
Abstract
Peripheral nerve injuries (PNI) are one of the most common nerve injuries, and graphene oxide (GO) has demonstrated significant potential in the treatment of PNI. GO could enhance the proliferation, adhesion, migration, and differentiation of neuronal cells by upregulating the expression of relevant proteins, and regulate the angiogenesis process and immune response. Therefore, GO is a suitable additional component for fabricating artificial nerve scaffolds (ANS), in which the slight addition of GO could improve the physicochemical performance of the matrix materials, through hydrogen bonds and electrostatic attraction. GO-composited ANS can increase the expression of nerve regeneration-associated genes and factors, promoting angiogenesis by activating the RAS/MAPK and AKT-eNOS-VEGF signaling pathway, respectively. Moreover, GO could be metabolized and excreted from the body through the pathway of peroxidase degradation in vivo. Consequently, the application of GO in PNI regeneration exhibits significant potential for transitioning from laboratory research to clinical use.
Collapse
Affiliation(s)
- Yulin Wang
- College of Biomedical Engineering, Sichuan University, China; Institute of Regulatory Science for Medical Devices, Sichuan University, China
| | - Bing Yang
- College of Biomedical Engineering, Sichuan University, China; Precision Medical Center of Southwest China Hospital, Sichuan University, China
| | - Zhongbing Huang
- College of Biomedical Engineering, Sichuan University, China.
| | - Zhaopu Yang
- Center for Drug Inspection, Guizhou Medical Products Administration, China
| | - Juan Wang
- College of Biomedical Engineering, Sichuan University, China
| | - Qiang Ao
- College of Biomedical Engineering, Sichuan University, China; Institute of Regulatory Science for Medical Devices, Sichuan University, China
| | - Guangfu Yin
- College of Biomedical Engineering, Sichuan University, China
| | - Ya Li
- College of Biomedical Engineering, Sichuan University, China; Institute of Regulatory Science for Medical Devices, Sichuan University, China
| |
Collapse
|
23
|
Yang CY, Hou Z, Hu P, Li C, Li Z, Cheng Z, Yang S, Ma P, Meng Z, Wu H, Pan Y, Cao Z, Wang X. Multi-needle blow-spinning technique for fabricating collagen nanofibrous nerve guidance conduit with scalable productivity and high performance. Mater Today Bio 2024; 24:100942. [PMID: 38283983 PMCID: PMC10819744 DOI: 10.1016/j.mtbio.2024.100942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 12/18/2023] [Accepted: 01/02/2024] [Indexed: 01/30/2024] Open
Abstract
Nerve guidance conduits (NGCs) have been widely accepted as a promising strategy for peripheral nerve regeneration. Fabricating ideal NGCs with good biocompatibility, biodegradability, permeability, appropriate mechanical properties (space maintenance, suturing performance, etc.), and oriented topographic cues is still current research focus. From the perspective of translation, the technique stability and scalability are also an important consideration for industrial production. Recently, blow-spinning technique shows great potentials in nanofibrous scaffolds fabrication, possessing high quality, high fiber production rates, low cost, ease of maintenance, and high reliability. In this study, we proposed for the first time the preparation of a novel NGC via blow-spinning technique to obtain optimized performances and high productivity. A new collagen nanofibrous neuro-tube with the bilayered design was developed, incorporating inner oriented and outer random topographical cues. The bilayer structure enhances the mechanical properties of the conduit in dry and wet, displaying good radial support and suturing performance. The porous nature of the blow-spun collagen membrane enables good nutrient delivery and metabolism. The in vitro and in vivo evaluations indicated the bilayer-structure conduit could promoted Schwann cells growth, neurotrophic factors secretion, and axonal regeneration and motor functional recovery in rat.
Collapse
Affiliation(s)
- Chun-Yi Yang
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, PR China
| | - Zhaohui Hou
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, PR China
| | - Peilun Hu
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, PR China
- Department of Orthopaedics Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 102218, PR China
| | - Chengli Li
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, PR China
- Department of Orthopaedics Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 102218, PR China
| | - Zifan Li
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, PR China
| | - Zekun Cheng
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, PR China
| | - Shuhui Yang
- School of Materials Science and Engineering, Zhejiang-Mauritius Joint Research Center for Biomaterials and Tissue Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, PR China
| | - Pengchao Ma
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, PR China
| | - Zhe Meng
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, PR China
| | - Hui Wu
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, PR China
| | - Yongwei Pan
- Department of Orthopaedics Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 102218, PR China
| | - Zheng Cao
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, PR China
- Center for Biomaterials and Regenerative Medicine, Wuzhen Laboratory, Tongxiang, 314500, PR China
| | - Xiumei Wang
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, PR China
| |
Collapse
|
24
|
Tanaka H, Kurimoto S, Hirata H. Efficacy of collagen conduit wrapping with collagen fibers on nerve regeneration in sciatic nerve injury with partial transection: An experimental study in the rat model. J Biomed Mater Res B Appl Biomater 2024; 112:e35369. [PMID: 38247253 DOI: 10.1002/jbm.b.35369] [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: 06/07/2023] [Revised: 10/09/2023] [Accepted: 12/02/2023] [Indexed: 01/23/2024]
Abstract
Peripheral nerve injuries (PNIs) include complete and partial transection, crushing, and chronic compression injuries. Hollow absorbable conduits are used to treat complete transection with short defects, while wrapping the injured part with an absorbent material promotes nerve recovery by inhibiting inflammatory cell infiltration and scar tissue formation in crush injuries. For treatment of partially transected nerve injuries (PTNIs), such as injection-related iatrogenic PNI, whether wrapping the entire nerve, including the injury site, or bridging the transected fascicle with an artificial nerve conduit (ANC) is beneficial remains to be verified. The purpose of this study was to investigate whether wrapping the injured nerve and placing collagen fibers as scaffolds at the nerve defect site contribute to neural recovery in PTNI. A unilateral 5-mm partial nerve defect was created at the mid-thigh level in a rat sciatic nerve injury model. Fifty-four Sprague-Dawley (SD) rats (150-250 g) were divided into three groups (n = 9 each): group 1, collagen fibers were placed in the nerve defect and the sciatic nerve was wrapped with collagen conduit; group 2, the sciatic nerve was wrapped by collagen conduit without collagen fibers; and group 3, nerve defect was reconstructed with collagen-filled conduit. Nerve regeneration was evaluated by analyses of gait, electrophysiology, wet muscle weight, and axon numbers with immunohistochemistry at 12 and 24 weeks. Dorsiflexion angles among all groups improved significantly from 12 to 24 weeks postoperatively. At 24 weeks postoperatively, compound muscle action potential amplitudes (CMAPs) of tibialis anterior were 5.26 ± 4.64, 1.31 ± 1.17, and 0.14 ± 0.24 mV (p < .05), CMAPs of gastrocnemius were 21.3 ± 5.98, 15.4 ± 5.46, and 13.11 ± 3.91 mV in groups 1, 2, and 3, respectively; and the value of group 1 was significantly higher than that of group 3 (p < .05). Axon numbers were 2194 ± 629; 1106 ± 645; and 805 ± 907 in groups 1, 2, and 3, respectively (p < .05). For PTNI reconstruction, artificial nerve wrap (ANW) was superior to ANC. Providing collagen scaffold at the nerve defect site enhanced nerve recovery during reconstruction with ANW.
Collapse
Affiliation(s)
- Hiromasa Tanaka
- Department of Hand Surgery, Graduate School of Medicine, Nagoya University, Nagoya City, Japan
| | - Shigeru Kurimoto
- Department of Hand Surgery, Graduate School of Medicine, Nagoya University, Nagoya City, Japan
| | - Hitoshi Hirata
- Department of Hand Surgery, Graduate School of Medicine, Nagoya University, Nagoya City, Japan
| |
Collapse
|
25
|
Rein S, Schober R, Poetschke J, Kremer T. Non degradation of chitosan and initial degradation of collagen nerve conduits used for protection of nerve coaptations. Microsurgery 2024; 44:e31093. [PMID: 37477338 DOI: 10.1002/micr.31093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 07/03/2023] [Accepted: 07/06/2023] [Indexed: 07/22/2023]
Abstract
BACKGROUND Nerve conduits are either used to bridge nerve gaps of up to 3 cm or to protect nerve coaptations. Biodegradable nerve conduits, which are currently commercially available, include Chitosan or collagen-based ones. As histological aspects of their degradation are highly relevant for the progress of neuronal regeneration, the aim of this study was to report the histopathological signs of such nerve conduits, which were removed during revision surgery. MATERIALS AND METHODS Either Chitosan (n = 2) or collagen (n = 2) nerve conduits were implanted after neuroma resection and nerve grafting (n = 2) or traumatic nerve lesion after cut (n = 1) or crush injury (n = 1) in two females and two men, aged between 17 and 57 years. Revision surgery with removal of the nerve conduits was indicated due to persisting neuropathic pain and sensorimotor deficits, limited joint motion, or neurolysis with hardware removal at a median time of 17 months (range: 5.5-48 months). Histopathological analyses of all removed nerve conduits were performed. RESULTS A scar neuroma was diagnosed in one out of four patients. Mechanical complication occurred in one patient after nerve conduit implantation bridged over finger joints. Intraoperatively no or only initial signs of degradation of the nerve conduits were observed. Chitosan conduits revealed largely unchanged shape and structure of chitosan, and coating of the conduit by a vascularized fibrous membrane. The latter contained deposits taken up by macrophages, most likely representing dissolved chitosan. Characteristic histopathologic features of the degradation of collagen conduits were a disintegration of the compact collagen into separate fine circular strands, No foreign body reaction was observed in all removed nerve conduits. CONCLUSIONS Both Chitosan nerve conduits have not been degraded. The collagen nerve conduits showed a beginning degradation process. Furthermore, wrapping the repaired nerve with a nerve conduit did neither prevent adhesions nor improved nerve gliding. Therefore, biodegradation in time should be particularly addressed in further developments of nerve conduits.
Collapse
Affiliation(s)
- Susanne Rein
- Department of Plastic and Handsurgery, Burn Unit, Klinikum St. Georg gGmbH, Leipzig, Germany
- Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Ralf Schober
- Institute for Pathology and Tumour Diagnostics, Klinikum St. Georg gGmbH, Leipzig, Germany
| | - Julian Poetschke
- Department of Plastic and Handsurgery, Burn Unit, Klinikum St. Georg gGmbH, Leipzig, Germany
| | - Thomas Kremer
- Department of Plastic and Handsurgery, Burn Unit, Klinikum St. Georg gGmbH, Leipzig, Germany
| |
Collapse
|
26
|
Kunisaki A, Kodama A, Ishikawa M, Ueda T, Lima MD, Kondo T, Adachi N. Oxidation-treated carbon nanotube yarns accelerate neurite outgrowth and induce axonal regeneration in peripheral nerve defect. Sci Rep 2023; 13:21799. [PMID: 38066058 PMCID: PMC10709329 DOI: 10.1038/s41598-023-48534-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 11/28/2023] [Indexed: 12/18/2023] Open
Abstract
Carbon nanotubes (CNTs) have the potential to promote peripheral nerve regeneration, although with limited capacity and foreign body reaction. This study investigated whether CNTs hydrophilized by oxidation can improve peripheral nerve regeneration and reduce foreign body reactions and inflammation. Three different artificial nerve conduit models were created using CNTs treated with ozone (O group), strong acid (SA group), and untreated (P group). They were implanted into a rat sciatic nerve defect model and evaluated after 8 and 16 weeks. At 16 weeks, the SA group showed significant recovery in functional and electrophysiological evaluations compared with the others. At 8 weeks, histological examination revealed a significant increase in the density of regenerated neurofilament and decreased foreign body giant cells in the SA group compared with the others. Oxidation-treated CNTs improved biocompatibility, induced nerve regeneration, and inhibited foreign-body reactions.
Collapse
Affiliation(s)
- Atsushi Kunisaki
- Department of Orthopaedic Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Akira Kodama
- Department of Orthopaedic Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan.
| | - Masakazu Ishikawa
- Department of Orthopaedic Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Takahiro Ueda
- Nano-Science and Technology Center, LINTEC OF AMERICA, INC., Richardson, USA
| | - Marcio D Lima
- Nano-Science and Technology Center, LINTEC OF AMERICA, INC., Richardson, USA
| | - Takeshi Kondo
- Nano-Science and Technology Center, LINTEC OF AMERICA, INC., Richardson, USA
| | - Nobuo Adachi
- Department of Orthopaedic Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
27
|
Ling J, He C, Zhang S, Zhao Y, Zhu M, Tang X, Li Q, Xu L, Yang Y. Progress in methods for evaluating Schwann cell myelination and axonal growth in peripheral nerve regeneration via scaffolds. Front Bioeng Biotechnol 2023; 11:1308761. [PMID: 38162183 PMCID: PMC10755477 DOI: 10.3389/fbioe.2023.1308761] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 11/20/2023] [Indexed: 01/03/2024] Open
Abstract
Peripheral nerve injury (PNI) is a neurological disorder caused by trauma that is frequently induced by accidents, war, and surgical complications, which is of global significance. The severity of the injury determines the potential for lifelong disability in patients. Artificial nerve scaffolds have been investigated as a powerful tool for promoting optimal regeneration of nerve defects. Over the past few decades, bionic scaffolds have been successfully developed to provide guidance and biological cues to facilitate Schwann cell myelination and orientated axonal growth. Numerous assessment techniques have been employed to investigate the therapeutic efficacy of nerve scaffolds in promoting the growth of Schwann cells and axons upon the bioactivities of distinct scaffolds, which have encouraged a greater understanding of the biological mechanisms involved in peripheral nerve development and regeneration. However, it is still difficult to compare the results from different labs due to the diversity of protocols and the availability of innovative technologies when evaluating the effectiveness of novel artificial scaffolds. Meanwhile, due to the complicated process of peripheral nerve regeneration, several evaluation methods are usually combined in studies on peripheral nerve repair. Herein, we have provided an overview of the evaluation methods used to study the outcomes of scaffold-based therapies for PNI in experimental animal models and especially focus on Schwann cell functions and axonal growth within the regenerated nerve.
Collapse
Affiliation(s)
- Jue Ling
- Key Laboratory of Neuroregeneration, Ministry of Education and Jiangsu Province, Co-Innovation Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, China
| | - Chang He
- Key Laboratory of Neuroregeneration, Ministry of Education and Jiangsu Province, Co-Innovation Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, China
| | - Shuxuan Zhang
- Key Laboratory of Neuroregeneration, Ministry of Education and Jiangsu Province, Co-Innovation Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, China
| | - Yahong Zhao
- Key Laboratory of Neuroregeneration, Ministry of Education and Jiangsu Province, Co-Innovation Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, China
| | - Meifeng Zhu
- College of Life Sciences, Nankai University, Tianjin, China
| | - Xiaoxuan Tang
- Key Laboratory of Neuroregeneration, Ministry of Education and Jiangsu Province, Co-Innovation Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, China
| | - Qiaoyuan Li
- Key Laboratory of Neuroregeneration, Ministry of Education and Jiangsu Province, Co-Innovation Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, China
| | - Liming Xu
- Institute of Medical Device Control, National Institutes for Food and Drug Control, Beijing, China
| | - Yumin Yang
- Key Laboratory of Neuroregeneration, Ministry of Education and Jiangsu Province, Co-Innovation Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, China
| |
Collapse
|
28
|
Joshi A, Choudhury S, Asthana S, Homer-Vanniasinkam S, Nambiar U, Chatterjee K. Emerging 4D fabrication of next-generation nerve guiding conduits: a critical perspective. Biomater Sci 2023; 11:7703-7708. [PMID: 37981830 DOI: 10.1039/d3bm01299a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2023]
Abstract
The latest advancements in the field of manufacturing for biomedicine, digital health, targeted therapy, and personalized medicine have fuelled the fabrication of smart medical devices. Four-dimensional (4D) fabrication strategies, which combine the manufacturing of three-dimensional (3D) parts with smart materials and/or design, have proved beneficial in creating customized and self-fitting structures that change their properties on demand with time. These frontier techniques that yield dynamic implants can indeed alleviate various drawbacks of current clinical practices, such as the use of sutures and complex microsurgeries and associated inflammation, among others. Among various clinical applications, 4D fabrication has lately made remarkable progress in the development of next-generation nerve-guiding conduits for treating peripheral nerve injuries (PNIs) by improving the end-to-end co-aptation of transected nerve endings. The current perspective highlights the relevance of 4D fabrication in developing state-of-the-art technologies for the treatment of PNIs. Various 4D fabrication/bio-fabrication techniques for PNI treatment are summarized while identifying the challenges and opportunities for the future. Such advancements hold immense promise for improving the quality of life of patients suffering from nerve damage and the potential for extending the treatment of many other disorders. Although the techniques are being described for PNIs, they will lend themselves suitably to certain cases of cranial nerve injuries as well.
Collapse
Affiliation(s)
- Akshat Joshi
- Department of Bioengineering, Indian Institute of Science, C. V. Raman Avenue, Bangalore 560012, India.
| | - Saswat Choudhury
- Department of Bioengineering, Indian Institute of Science, C. V. Raman Avenue, Bangalore 560012, India.
| | - Sonal Asthana
- Department of Materials Engineering, Indian Institute of Science, C. V. Raman Avenue, Bangalore 560012, India
- Department of Hepatobiliary and Multi-Organ Transplantation Surgery, Aster CMI Hospital, Bangalore 560024, India
| | - Shervanthi Homer-Vanniasinkam
- Department of Materials Engineering, Indian Institute of Science, C. V. Raman Avenue, Bangalore 560012, India
- Department of Mechanical Engineering and Division of Surgery, University College London, WC1E 7JE, UK
| | - Uma Nambiar
- Bagchi-Parthasarathy Hospital, Indian Institute of Science, C. V. Raman Avenue, Bangalore 560012, India
| | - Kaushik Chatterjee
- Department of Bioengineering, Indian Institute of Science, C. V. Raman Avenue, Bangalore 560012, India.
- Department of Materials Engineering, Indian Institute of Science, C. V. Raman Avenue, Bangalore 560012, India
| |
Collapse
|
29
|
Rahman M, Mahady Dip T, Padhye R, Houshyar S. Review on electrically conductive smart nerve guide conduit for peripheral nerve regeneration. J Biomed Mater Res A 2023; 111:1916-1950. [PMID: 37555548 DOI: 10.1002/jbm.a.37595] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 01/29/2023] [Accepted: 07/26/2023] [Indexed: 08/10/2023]
Abstract
At present, peripheral nerve injuries (PNIs) are one of the leading causes of substantial impairment around the globe. Complete recovery of nerve function after an injury is challenging. Currently, autologous nerve grafts are being used as a treatment; however, this has several downsides, for example, donor site morbidity, shortage of donor sites, loss of sensation, inflammation, and neuroma development. The most promising alternative is the development of a nerve guide conduit (NGC) to direct the restoration and renewal of neuronal axons from the proximal to the distal end to facilitate nerve regeneration and maximize sensory and functional recovery. Alternatively, the response of nerve cells to electrical stimulation (ES) has a substantial regenerative effect. The incorporation of electrically conductive biomaterials in the fabrication of smart NGCs facilitates the function of ES throughout the active proliferation state. This article overviews the potency of the various categories of electroactive smart biomaterials, including conductive and piezoelectric nanomaterials, piezoelectric polymers, and organic conductive polymers that researchers have employed latterly to fabricate smart NGCs and their potentiality in future clinical application. It also summarizes a comprehensive analysis of the recent research and advancements in the application of ES in the field of NGC.
Collapse
Affiliation(s)
- Mustafijur Rahman
- Center for Materials Innovation and Future Fashion (CMIFF), School of Fashion and Textiles, RMIT University, Brunswick, Australia
- Department of Dyes and Chemical Engineering, Bangladesh University of Textiles, Dhaka, Bangladesh
| | - Tanvir Mahady Dip
- Department of Materials, University of Manchester, Manchester, UK
- Department of Yarn Engineering, Bangladesh University of Textiles, Dhaka, Bangladesh
| | - Rajiv Padhye
- Center for Materials Innovation and Future Fashion (CMIFF), School of Fashion and Textiles, RMIT University, Brunswick, Australia
| | - Shadi Houshyar
- School of Engineering, RMIT University, Melbourne, Victoria, Australia
| |
Collapse
|
30
|
Meng Q, Burrell JC, Zhang Q, Le AD. Potential Application of Orofacial MSCs in Tissue Engineering Nerve Guidance for Peripheral Nerve Injury Repair. Stem Cell Rev Rep 2023; 19:2612-2631. [PMID: 37642899 DOI: 10.1007/s12015-023-10609-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/15/2023] [Indexed: 08/31/2023]
Abstract
Injury to the peripheral nerve causes potential loss of sensory and motor functions, and peripheral nerve repair (PNR) remains a challenging endeavor. The current clinical methods of nerve repair, such as direct suture, autografts, and acellular nerve grafts (ANGs), exhibit their respective disadvantages like nerve tension, donor site morbidity, size mismatch, and immunogenicity. Even though commercially available nerve guidance conduits (NGCs) have demonstrated some clinical successes, the overall clinical outcome is still suboptimal, especially for nerve injuries with a large gap (≥ 3 cm) due to the lack of biologics. In the last two decades, the combination of advanced tissue engineering technologies, stem cell biology, and biomaterial science has significantly advanced the generation of a new generation of NGCs incorporated with biological factors or supportive cells, including mesenchymal stem cells (MSCs), which hold great promise to enhance peripheral nerve repair/regeneration (PNR). Orofacial MSCs are emerging as a unique source of MSCs for PNR due to their neural crest-origin and easy accessibility. In this narrative review, we have provided an update on the pathophysiology of peripheral nerve injury and the properties and biological functions of orofacial MSCs. Then we have highlighted the application of orofacial MSCs in tissue engineering nerve guidance for PNR in various preclinical models and the potential challenges and future directions in this field.
Collapse
Affiliation(s)
- Qingyu Meng
- Department of Oral & Maxillofacial Surgery & Pharmacology, University of Pennsylvania School of Dental Medicine, 240 South 40Th Street, Philadelphia, PA, 19104, USA
| | - Justin C Burrell
- Department of Oral & Maxillofacial Surgery & Pharmacology, University of Pennsylvania School of Dental Medicine, 240 South 40Th Street, Philadelphia, PA, 19104, USA
| | - Qunzhou Zhang
- Department of Oral & Maxillofacial Surgery & Pharmacology, University of Pennsylvania School of Dental Medicine, 240 South 40Th Street, Philadelphia, PA, 19104, USA.
| | - Anh D Le
- Department of Oral & Maxillofacial Surgery & Pharmacology, University of Pennsylvania School of Dental Medicine, 240 South 40Th Street, Philadelphia, PA, 19104, USA.
- Department of Oral & Maxillofacial Surgery, Penn Medicine Hospital of the University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA, 19104, USA.
| |
Collapse
|
31
|
Litowczenko J, Wychowaniec JK, Załęski K, Marczak Ł, Edwards-Gayle CJC, Tadyszak K, Maciejewska BM. Micro/nano-patterns for enhancing differentiation of human neural stem cells and fabrication of nerve conduits via soft lithography and 3D printing. BIOMATERIALS ADVANCES 2023; 154:213653. [PMID: 37862812 DOI: 10.1016/j.bioadv.2023.213653] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 10/02/2023] [Accepted: 10/06/2023] [Indexed: 10/22/2023]
Abstract
Topographical cues on materials can manipulate cellular fate, particularly for neural cells that respond well to such cues. Utilizing biomaterial surfaces with topographical features can effectively influence neuronal differentiation and promote neurite outgrowth. This is crucial for improving the regeneration of damaged neural tissue after injury. Here, we utilized groove patterns to create neural conduits that promote neural differentiation and axonal growth. We investigated the differentiation of human neural stem cells (NSCs) on silicon dioxide groove patterns with varying height-to-width/spacing ratios. We hypothesize that NSCs can sense the microgrooves with nanoscale depth on different aspect ratio substrates and exhibit different morphologies and differentiation fate. A comprehensive approach was employed, analyzing cell morphology, neurite length, and cell-specific markers. These aspects provided insights into the behavior of the investigated NSCs and their response to the topographical cues. Three groove-pattern models were designed with varying height-to-width/spacing ratios of 80, 42, and 30 for groove pattern widths of 1 μm, 5 μm, and 10 μm and nanoheights of 80 nm, 210 nm, and 280 nm. Smaller groove patterns led to longer neurites and more effective differentiation towards neurons, whereas larger patterns promoted multidimensional differentiation towards both neurons and glia. We transferred these cues onto patterned polycaprolactone (PCL) and PCL-graphene oxide (PCL-GO) composite 'stamps' using simple soft lithography and reproducible extrusion 3D printing methods. The patterned scaffolds elicited a response from NSCs comparable to that of silicon dioxide groove patterns. The smallest pattern stimulated the highest neurite outgrowth, while the middle-sized grooves of PCL-GO induced effective synaptogenesis. We demonstrated the potential for such structures to be wrapped into tubes and used as grafts for peripheral nerve regeneration. Grooved PCL and PCL-GO conduits could be a promising alternative to nerve grafting.
Collapse
Affiliation(s)
- Jagoda Litowczenko
- NanoBioMedical Centre, Adam Mickiewicz University, Wszechnicy Piastowskiej 3, PL61614 Poznań, Poland.
| | - Jacek K Wychowaniec
- NanoBioMedical Centre, Adam Mickiewicz University, Wszechnicy Piastowskiej 3, PL61614 Poznań, Poland; AO Research Institute Davos, Clavadelerstrasse 8, 7270 Davos, Switzerland
| | - Karol Załęski
- NanoBioMedical Centre, Adam Mickiewicz University, Wszechnicy Piastowskiej 3, PL61614 Poznań, Poland
| | - Łukasz Marczak
- European Centre for Bioinformatics and Genomics, Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego 12/14, 61-704 Poznań, Poland
| | | | - Krzysztof Tadyszak
- Institute of Macromolecular Chemistry, CAS, Heyrovského nám. 2, 162 06 Prague 6, Czech Republic
| | - Barbara M Maciejewska
- NanoBioMedical Centre, Adam Mickiewicz University, Wszechnicy Piastowskiej 3, PL61614 Poznań, Poland
| |
Collapse
|
32
|
Wu S, Shen W, Ge X, Ao F, Zheng Y, Wang Y, Jia X, Mao Y, Luo Y. Advances in Large Gap Peripheral Nerve Injury Repair and Regeneration with Bridging Nerve Guidance Conduits. Macromol Biosci 2023; 23:e2300078. [PMID: 37235853 DOI: 10.1002/mabi.202300078] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 05/10/2023] [Indexed: 05/28/2023]
Abstract
Peripheral nerve injury is a common complication of accidents and diseases. The traditional autologous nerve graft approach remains the gold standard for the treatment of nerve injuries. While sources of autologous nerve grafts are very limited and difficult to obtain. Nerve guidance conduits are widely used in the treatment of peripheral nerve injuries as an alternative to nerve autografts and allografts. However, the development of nerve conduits does not meet the needs of large gap peripheral nerve injury. Functional nerve conduits can provide a good microenvironment for axon elongation and myelin regeneration. Herein, the manufacturing methods and different design types of functional bridging nerve conduits for nerve conduits combined with electrical or magnetic stimulation and loaded with Schwann cells, etc., are summarized. It summarizes the literature and finds that the technical solutions of functional nerve conduits with electrical stimulation, magnetic stimulation and nerve conduits combined with Schwann cells can be used as effective strategies for bridging large gap nerve injury and provide an effective way for the study of large gap nerve injury repair. In addition, functional nerve conduits provide a new way to construct delivery systems for drugs and growth factors in vivo.
Collapse
Affiliation(s)
- Shang Wu
- School of Biological and Pharmaceutical Sciences, Shaanxi University of Science and Technology, Xi'an, 710021, P. R. China
| | - Wen Shen
- School of Biological and Pharmaceutical Sciences, Shaanxi University of Science and Technology, Xi'an, 710021, P. R. China
| | - Xuemei Ge
- College of Light Industry and Food Engineering, Nanjing Forestry University, Nanjing, 210037, P. R. China
| | - Fen Ao
- College of Bioresources Chemical and Materials Engineering, Shaanxi University of Science and Technology, Xi'an, 710021, P. R. China
| | - Yan Zheng
- School of Biological and Pharmaceutical Sciences, Shaanxi University of Science and Technology, Xi'an, 710021, P. R. China
| | - Yigang Wang
- Department of Pharmacy, No. 215 Hospital of Shaanxi Nuclear Industry, Xianyang, Shaanxi, 712000, P. R. China
| | - Xiaoni Jia
- Central Laboratory, Xi'an Mental Health Center, Xi'an, 710061, P. R. China
| | - Yueyang Mao
- School of Biological and Pharmaceutical Sciences, Shaanxi University of Science and Technology, Xi'an, 710021, P. R. China
| | - Yali Luo
- College of Light Industry and Food Engineering, Nanjing Forestry University, Nanjing, 210037, P. R. China
| |
Collapse
|
33
|
Shoman N. Nerve guide conduits, nerve transfers, and local and free muscle transfer in facial nerve palsy. Curr Opin Otolaryngol Head Neck Surg 2023; 31:306-312. [PMID: 37581264 DOI: 10.1097/moo.0000000000000914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/16/2023]
Abstract
PURPOSE OF REVIEW To highlight the recent literature on reinnervation options in the management of facial nerve paralysis using nerve conduits, and nerve and muscle transfers. RECENT FINDINGS Engineering of natural and synthetic nerve conduits has progressed and many of these products are now available on the market. The use of the masseter nerve has become more popular recently as a choice in nerve transfer procedures due to various unique advantages. Various authors have recently described mimetic muscle reinnervation using more than one nerve transfer, as well as dual and triple innervation of free muscle transfer. SUMMARY The ideal nerve conduit continues to be elusive, however significant progress has been made with many natural and synthetic materials and designs tested and introduced on the market. Many authors have modified the classic approaches in motor nerve transfer, as well as local and free muscle transfer, and described new ones, that aim to combine their advantages, particularly the simplification to a single stage and use of multiple reinnervation to the mimetic muscles. These advances are valuable to the reconstructive surgeon as powerful tools that can be tailored to the unique challenges of patients with facial nerve palsy looking for dynamic reanimation options.
Collapse
Affiliation(s)
- Nael Shoman
- Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| |
Collapse
|
34
|
Harley-Troxell ME, Steiner R, Advincula RC, Anderson DE, Dhar M. Interactions of Cells and Biomaterials for Nerve Tissue Engineering: Polymers and Fabrication. Polymers (Basel) 2023; 15:3685. [PMID: 37765540 PMCID: PMC10536046 DOI: 10.3390/polym15183685] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 08/31/2023] [Accepted: 09/01/2023] [Indexed: 09/29/2023] Open
Abstract
Neural injuries affect millions globally, significantly impacting their quality of life. The inability of these injuries to heal, limited ability to regenerate, and the lack of available treatments make regenerative medicine and tissue engineering a promising field of research for developing methods for nerve repair. This review evaluates the use of natural and synthetic polymers, and the fabrication methods applied that influence a cell's behavior. Methods include cross-linking hydrogels, incorporation of nanoparticles, and 3D printing with and without live cells. The endogenous cells within the injured area and any exogenous cells seeded on the polymer construct play a vital role in regulating healthy neural activity. This review evaluates the body's local and systemic reactions to the implanted materials. Although numerous variables are involved, many of these materials and methods have exhibited the potential to provide a biomaterial environment that promotes biocompatibility and the regeneration of a physical and functional nerve. Future studies may evaluate advanced methods for modifying material properties and characterizing the tissue-biomaterial interface for clinical applications.
Collapse
Affiliation(s)
- Meaghan E. Harley-Troxell
- Tissue Engineering and Regenerative Medicine, Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN 37996, USA; (M.E.H.-T.); (R.S.); (D.E.A.)
| | - Richard Steiner
- Tissue Engineering and Regenerative Medicine, Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN 37996, USA; (M.E.H.-T.); (R.S.); (D.E.A.)
| | - Rigoberto C. Advincula
- Department of Chemical and Biomolecular Engineering, University of Tennessee, Knoxville, TN 37996, USA;
- Oak Ridge National Laboratory, Center for Nanophase Materials Sciences, Oak Ridge, TN 37831, USA
| | - David E. Anderson
- Tissue Engineering and Regenerative Medicine, Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN 37996, USA; (M.E.H.-T.); (R.S.); (D.E.A.)
| | - Madhu Dhar
- Tissue Engineering and Regenerative Medicine, Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN 37996, USA; (M.E.H.-T.); (R.S.); (D.E.A.)
| |
Collapse
|
35
|
Chen L, Song X, Yao Z, Zhou C, Yang J, Yang Q, Chen J, Wu J, Sun Z, Gu L, Ma Y, Lee SJ, Zhang C, Mao HQ, Sun L. Gelatin nanofiber-reinforced decellularized amniotic membrane promotes axon regeneration and functional recovery in the surgical treatment of peripheral nerve injury. Biomaterials 2023; 300:122207. [PMID: 37352606 DOI: 10.1016/j.biomaterials.2023.122207] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 06/08/2023] [Accepted: 06/12/2023] [Indexed: 06/25/2023]
Abstract
Effective recovery of peripheral nerve injury (PNI) after surgical treatment relies on promoting axon regeneration and minimizing the fibrotic response. Decellularized amniotic membrane (dAM) has unique features as a natural matrix for promoting PNI repair due to its pro-regenerative extracellular matrix (ECM) structure and anti-inflammatory properties. However, the fragile nature and rapid degradation rate of dAM limit its widespread use in PNI surgery. Here we report an engineered composite membrane for PNI repair by combining dAM with gelatin (Gel) nanofiber membrane to construct a Gel nanofiber-dAM composite membrane (Gel-dAM) through interfacial bonding. The Gel-dAM showed enhanced mechanical properties and reduced degradation rate, while retaining maximal bioactivity and biocompatibility of dAM. These factors led to improved axon regeneration, reduced fibrotic response, and better functional recovery in PNI repair. As a fully natural materials-derived off-the-shelf matrix, Gel-dAM exhibits superior clinical translational potential for the surgical treatment of PNI.
Collapse
Affiliation(s)
- Long Chen
- Department of Orthopedics, Guizhou Provincial People's Hospital, Guiyang, Guizhou, 550000, China; The Lab of Tissue Engineering and Translational Medicine, College of Medicine, Guizhou University, Guiyang, Guizhou, 550000, China
| | - Xiongbo Song
- Department of Orthopedics, Guizhou Provincial People's Hospital, Guiyang, Guizhou, 550000, China; The Lab of Tissue Engineering and Translational Medicine, College of Medicine, Guizhou University, Guiyang, Guizhou, 550000, China
| | - Zhicheng Yao
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, 21218, USA; Department of Materials Science and Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA; Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Conglai Zhou
- Department of Orthopedics, Guizhou Provincial People's Hospital, Guiyang, Guizhou, 550000, China; The Lab of Tissue Engineering and Translational Medicine, College of Medicine, Guizhou University, Guiyang, Guizhou, 550000, China
| | - Junjun Yang
- The Lab of Tissue Engineering and Translational Medicine, College of Medicine, Guizhou University, Guiyang, Guizhou, 550000, China
| | - Qiming Yang
- Department of Orthopedics, Guizhou Provincial People's Hospital, Guiyang, Guizhou, 550000, China; The Lab of Tissue Engineering and Translational Medicine, College of Medicine, Guizhou University, Guiyang, Guizhou, 550000, China
| | - Junrong Chen
- Department of Orthopedics, Guizhou Provincial People's Hospital, Guiyang, Guizhou, 550000, China; The Lab of Tissue Engineering and Translational Medicine, College of Medicine, Guizhou University, Guiyang, Guizhou, 550000, China
| | - Jiarui Wu
- Department of Orthopedics, Guizhou Provincial People's Hospital, Guiyang, Guizhou, 550000, China; The Lab of Tissue Engineering and Translational Medicine, College of Medicine, Guizhou University, Guiyang, Guizhou, 550000, China
| | - Zeyu Sun
- Department of Orthopedics, Guizhou Provincial People's Hospital, Guiyang, Guizhou, 550000, China
| | - Liling Gu
- Department of Orthopedics, Guizhou Provincial People's Hospital, Guiyang, Guizhou, 550000, China
| | - Yi Ma
- The Lab of Tissue Engineering and Translational Medicine, College of Medicine, Guizhou University, Guiyang, Guizhou, 550000, China
| | - Shin-Jae Lee
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, 21218, USA; Department of Materials Science and Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Chi Zhang
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, 21218, USA; Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
| | - Hai-Quan Mao
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, 21218, USA; Department of Materials Science and Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA; Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA; Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| | - Li Sun
- Department of Orthopedics, Guizhou Provincial People's Hospital, Guiyang, Guizhou, 550000, China; The Lab of Tissue Engineering and Translational Medicine, College of Medicine, Guizhou University, Guiyang, Guizhou, 550000, China.
| |
Collapse
|
36
|
Yang X, Li X, Wu Z, Cao L. Photocrosslinked methacrylated natural macromolecular hydrogels for tissue engineering: A review. Int J Biol Macromol 2023; 246:125570. [PMID: 37369259 DOI: 10.1016/j.ijbiomac.2023.125570] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 06/14/2023] [Accepted: 06/24/2023] [Indexed: 06/29/2023]
Abstract
A hydrogel is a three-dimensional (3D) network structure formed through polymer crosslinking, and these have emerged as a popular research topic in recent years. Hydrogel crosslinking can be classified as physical, chemical, or enzymatic, and photocrosslinking is a branch of chemical crosslinking. Compared with other methods, photocrosslinking can control the hydrogel crosslinking initiation, crosslinking time, and crosslinking strength using light. Owing to these properties, photocrosslinked hydrogels have important research prospects in tissue engineering, in situ gel formation, 3D bioprinting, and drug delivery. Methacrylic anhydride modification is a common method for imparting photocrosslinking properties to polymers, and graft-substituted polymers can be photocrosslinked under UV irradiation. In this review, we first introduce the characteristics of common natural polysaccharide- and protein-based hydrogels and the processes used for methacrylate group modification. Next, we discuss the applications of methacrylated natural hydrogels in tissue engineering. Finally, we summarize and discuss existing methacrylated natural hydrogels in terms of limitations and future developments. We expect that this review will help researchers in this field to better understand the synthesis of methacrylate-modified natural hydrogels and their applications in tissue engineering.
Collapse
Affiliation(s)
- Xiaoli Yang
- Department of Histology and Embryology, Fuzhou Medical College of Nanchang University, Fuzhou 344000, PR China
| | - Xiaojing Li
- Department of Histology and Embryology, Fuzhou Medical College of Nanchang University, Fuzhou 344000, PR China
| | - Zhaoping Wu
- Jiujiang City Key Laboratory of Cell Therapy, The First Hospital of Jiujiang City, Jiujiang 332000, PR China
| | - Lingling Cao
- Jiujiang City Key Laboratory of Cell Therapy, The First Hospital of Jiujiang City, Jiujiang 332000, PR China.
| |
Collapse
|
37
|
Tagandurdyyeva NA, Trube MA, Shemyakin IO, Solomitskiy DN, Medvedev GV, Dresvyanina EN, Nashchekina YA, Ivan’kova EM, Dobrovol’skaya IP, Kamalov AM, Sukhorukova EG, Moskalyuk OA, Yudin VE. Properties of Resorbable Conduits Based on Poly(L-Lactide) Nanofibers and Chitosan Fibers for Peripheral Nerve Regeneration. Polymers (Basel) 2023; 15:3323. [PMID: 37571217 PMCID: PMC10422266 DOI: 10.3390/polym15153323] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 07/28/2023] [Accepted: 08/02/2023] [Indexed: 08/13/2023] Open
Abstract
New tubular conduits have been developed for the regeneration of peripheral nerves and the repair of defects that are larger than 3 cm. The conduits consist of a combination of poly(L-lactide) nanofibers and chitosan composite fibers with chitin nanofibrils. In vitro studies were conducted to assess the biocompatibility of the conduits using human embryonic bone marrow stromal cells (FetMSCs). The studies revealed good adhesion and differentiation of the cells on the conduits just one day after cultivation. Furthermore, an in vivo study was carried out to evaluate motor-coordination disorders using the sciatic nerve functional index (SFI) assessment. The presence of chitosan monofibers and chitosan composite fibers with chitin nanofibrils in the conduit design increased the regeneration rate of the sciatic nerve, with an SFI value ranging from 76 to 83. The degree of recovery of nerve conduction was measured by the amplitude of M-response, which showed a 46% improvement. The conduit design imitates the oriented architecture of the nerve, facilitates electrical communication between the damaged nerve's ends, and promotes the direction of nerve growth, thereby increasing the regeneration rate.
Collapse
Affiliation(s)
- Nurjemal A. Tagandurdyyeva
- Institute of Biomedical Systems and Biotechnology, Peter the Great St. Petersburg Polytechnic University, Polytekhnicheskaya Str., 29, Saint Petersburg 195251, Russia;
| | - Maxim A. Trube
- Institute of Medicine, RUDN University, Miklukho-Maklaya Str., 6, Moscow 117198, Russia;
| | - Igor’ O. Shemyakin
- Scientific Research Center, Pavlov First Saint-Petersburg State Medical University, L’va Tolstogo Str., 6-8, Saint Petersburg 197022, Russia; (I.O.S.); (D.N.S.); (E.G.S.)
| | - Denis N. Solomitskiy
- Scientific Research Center, Pavlov First Saint-Petersburg State Medical University, L’va Tolstogo Str., 6-8, Saint Petersburg 197022, Russia; (I.O.S.); (D.N.S.); (E.G.S.)
| | - German V. Medvedev
- Medsi Clinic, Department of Plastic Surgery, Marata Str., 6A, Saint Petersburg 191025, Russia;
| | - Elena N. Dresvyanina
- Institute of Textile and Fashion, Saint Petersburg State University of Industrial Technologies and Design, B. Morskaya Str., 18, Saint Petersburg 191186, Russia;
| | - Yulia A. Nashchekina
- Cell Technologies Center, Institute of Cytology Russian Academy of Sciences, Tikhoretsky Ave., 4, Saint Petersburg 194064, Russia;
| | - Elena M. Ivan’kova
- Laboratory of Mechanics of Polymers and Composites, Institute of Macromolecular Compounds Russian Academy of Science, Bol’shoi Prospect V.O. 31, Saint Petersburg 199004, Russia; (E.M.I.); (I.P.D.); (V.E.Y.)
| | - Irina P. Dobrovol’skaya
- Laboratory of Mechanics of Polymers and Composites, Institute of Macromolecular Compounds Russian Academy of Science, Bol’shoi Prospect V.O. 31, Saint Petersburg 199004, Russia; (E.M.I.); (I.P.D.); (V.E.Y.)
| | - Almaz M. Kamalov
- Institute of Biomedical Systems and Biotechnology, Peter the Great St. Petersburg Polytechnic University, Polytekhnicheskaya Str., 29, Saint Petersburg 195251, Russia;
| | - Elena G. Sukhorukova
- Scientific Research Center, Pavlov First Saint-Petersburg State Medical University, L’va Tolstogo Str., 6-8, Saint Petersburg 197022, Russia; (I.O.S.); (D.N.S.); (E.G.S.)
| | - Olga A. Moskalyuk
- Laboratory of Polymer and Composite Materials–SmartTextiles, IRC–X-ray Coherent Optics, Immanuel Kant Baltic Federal University, A. Nevskogo Str., 14, Kaliningrad 236041, Russia
| | - Vladimir E. Yudin
- Laboratory of Mechanics of Polymers and Composites, Institute of Macromolecular Compounds Russian Academy of Science, Bol’shoi Prospect V.O. 31, Saint Petersburg 199004, Russia; (E.M.I.); (I.P.D.); (V.E.Y.)
| |
Collapse
|
38
|
Naghilou A, Peter K, Millesi F, Stadlmayr S, Wolf S, Rad A, Semmler L, Supper P, Ploszczanski L, Liu J, Burghammer M, Riekel C, Bismarck A, Backus EHG, Lichtenegger H, Radtke C. Insights into the material properties of dragline spider silk affecting Schwann cell migration. Int J Biol Macromol 2023:125398. [PMID: 37330085 DOI: 10.1016/j.ijbiomac.2023.125398] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 06/09/2023] [Accepted: 06/13/2023] [Indexed: 06/19/2023]
Abstract
Dragline silk of Trichonephila spiders has attracted attention in various applications. One of the most fascinating uses of dragline silk is in nerve regeneration as a luminal filling for nerve guidance conduits. In fact, conduits filled with spider silk can measure up to autologous nerve transplantation, but the reasons behind the success of silk fibers are not yet understood. In this study dragline fibers of Trichonephila edulis were sterilized with ethanol, UV radiation, and autoclaving and the resulting material properties were characterized with regard to the silk's suitability for nerve regeneration. Rat Schwann cells (rSCs) were seeded on these silks in vitro and their migration and proliferation were investigated as an indication for the fiber's ability to support the growth of nerves. It was found that rSCs migrate faster on ethanol treated fibers. To elucidate the reasons behind this behavior, the fiber's morphology, surface chemistry, secondary protein structure, crystallinity, and mechanical properties were studied. The results demonstrate that the synergy of dragline silk's stiffness and its composition has a crucial effect on the migration of rSCs. These findings pave the way towards understanding the response of SCs to silk fibers as well as the targeted production of synthetic alternatives for regenerative medicine applications.
Collapse
Affiliation(s)
- Aida Naghilou
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Vienna, Spitalgasse 23, 1090 Vienna, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria.
| | - Karolina Peter
- University of Natural Resources and Life Sciences, Department of Material Sciences and Process Engineering, Institute of Physics and Materials Science, Peter-Jordan-Strasse 82, 1190 Vienna, Austria
| | - Flavia Millesi
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Vienna, Spitalgasse 23, 1090 Vienna, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Sarah Stadlmayr
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Vienna, Spitalgasse 23, 1090 Vienna, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Sonja Wolf
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Vienna, Spitalgasse 23, 1090 Vienna, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Anda Rad
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Vienna, Spitalgasse 23, 1090 Vienna, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Lorenz Semmler
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Vienna, Spitalgasse 23, 1090 Vienna, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Paul Supper
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Vienna, Spitalgasse 23, 1090 Vienna, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Leon Ploszczanski
- University of Natural Resources and Life Sciences, Department of Material Sciences and Process Engineering, Institute of Physics and Materials Science, Peter-Jordan-Strasse 82, 1190 Vienna, Austria
| | - Jiliang Liu
- European Synchrotron Radiation Facility, 71 avenue des Martyrs, 38000 Grenoble, France
| | - Manfred Burghammer
- European Synchrotron Radiation Facility, 71 avenue des Martyrs, 38000 Grenoble, France
| | - Christian Riekel
- European Synchrotron Radiation Facility, 71 avenue des Martyrs, 38000 Grenoble, France
| | - Alexander Bismarck
- University of Vienna, Faculty of Chemistry, Institute of Materials Chemistry & Research, Währingerstraße 42, 1090 Vienna, Austria
| | - Ellen H G Backus
- University of Vienna, Faculty of Chemistry, Institute of Physical Chemistry, Währingerstraße 42, 1090 Vienna, Austria
| | - Helga Lichtenegger
- University of Natural Resources and Life Sciences, Department of Material Sciences and Process Engineering, Institute of Physics and Materials Science, Peter-Jordan-Strasse 82, 1190 Vienna, Austria
| | - Christine Radtke
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Vienna, Spitalgasse 23, 1090 Vienna, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria
| |
Collapse
|
39
|
Barros Araújo CB, da Silva Soares IL, da Silva Lima DP, Barros RM, de Lima Damasceno BPG, Oshiro-Junior JA. Polyvinyl Alcohol Nanofibers Blends as Drug Delivery System in Tissue Regeneration. Curr Pharm Des 2023; 29:1149-1162. [PMID: 37157221 DOI: 10.2174/1381612829666230508144912] [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: 09/26/2022] [Revised: 01/08/2023] [Accepted: 01/23/2023] [Indexed: 05/10/2023]
Abstract
Nanofibers have shown promising clinical results in the process of tissue regeneration since they provide a similar structure to the extracellular matrix of different tissues, high surface-to-volume ratio and porosity, flexibility, and gas permeation, offering topographical features that stimulate cell adhesion and proliferation. Electrospinning is one of the most used techniques for manufacturing nanomaterials due to its simplicity and low cost. In this review, we highlight the use of nanofibers produced with polyvinyl alcohol and polymeric associations (PVA/blends) as a matrix for release capable of modifying the pharmacokinetic profile of different active ingredients in the regeneration of connective, epithelial, muscular, and nervous tissues. Articles were selected by three independent reviewers by analyzing the databases, such as Web of Science, PubMed, Science Direct, and Google Scholar (last 10 years). Descriptors used were "nanofibers", "poly (vinyl alcohol)", "muscle tissue", "connective tissue", "epithelial tissue", and "neural tissue engineering". The guiding question was: How do different compositions of polyvinyl alcohol polymeric nanofibers modify the pharmacokinetics of active ingredients in different tissue regeneration processes? The results demonstrated the versatility of the production of PVA nanofibers by solution blow technique with different actives (lipo/hydrophilic) and with pore sizes varying between 60 and 450 nm depending on the polymers used in the mixture, which influences the drug release that can be controlled for hours or days. The tissue regeneration showed better cellular organization and greater cell proliferation compared to the treatment with the control group, regardless of the tissue analyzed. We highlight that, among all blends, the combinations PVA/PCL and PVA/CS showed good compatibility and slow degradation, indicating their use in prolonged times of biodegradation, thus benefiting tissue regeneration in bone and cartilage connective tissues, acting as a physical barrier that results in guided regeneration, and preventing the invasion of cells from other tissues with increased proliferation rate.
Collapse
Affiliation(s)
- Camila Beatriz Barros Araújo
- Pharmaceutical Sciences Postgraduate Center for Biological and Health Sciences, State University of Paraíba, Av. Juvêncio Arruda, S/N, Campina Grande, 58429-600, Paraíba, Brazil
| | - Ingrid Larissa da Silva Soares
- Pharmaceutical Sciences Postgraduate Center for Biological and Health Sciences, State University of Paraíba, Av. Juvêncio Arruda, S/N, Campina Grande, 58429-600, Paraíba, Brazil
- Research Center in Pharmaceutical Sciences, UNIFACISA University Center, Manoel Cardoso Palhano, Campina Grande, 58408-326, Paraíba, Brazil
| | - Diego Paulo da Silva Lima
- Pharmaceutical Sciences Postgraduate Center for Biological and Health Sciences, State University of Paraíba, Av. Juvêncio Arruda, S/N, Campina Grande, 58429-600, Paraíba, Brazil
| | - Rafaella Moreno Barros
- Pharmaceutical Sciences Postgraduate Center for Biological and Health Sciences, State University of Paraíba, Av. Juvêncio Arruda, S/N, Campina Grande, 58429-600, Paraíba, Brazil
| | - Bolívar Ponciano Goulart de Lima Damasceno
- Pharmaceutical Sciences Postgraduate Center for Biological and Health Sciences, State University of Paraíba, Av. Juvêncio Arruda, S/N, Campina Grande, 58429-600, Paraíba, Brazil
| | - João Augusto Oshiro-Junior
- Pharmaceutical Sciences Postgraduate Center for Biological and Health Sciences, State University of Paraíba, Av. Juvêncio Arruda, S/N, Campina Grande, 58429-600, Paraíba, Brazil
- Research Center in Pharmaceutical Sciences, UNIFACISA University Center, Manoel Cardoso Palhano, Campina Grande, 58408-326, Paraíba, Brazil
| |
Collapse
|
40
|
Zennifer A, Thangadurai M, Sundaramurthi D, Sethuraman S. Additive manufacturing of peripheral nerve conduits - Fabrication methods, design considerations and clinical challenges. SLAS Technol 2023; 28:102-126. [PMID: 37028493 DOI: 10.1016/j.slast.2023.03.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 03/20/2023] [Accepted: 03/28/2023] [Indexed: 04/08/2023]
Abstract
Tissue-engineered nerve guidance conduits (NGCs) are a viable clinical alternative to autografts and allografts and have been widely used to treat peripheral nerve injuries (PNIs). Although these NGCs are successful to some extent, they cannot aid in native regeneration by improving native-equivalent neural innervation or regrowth. Further, NGCs exhibit longer recovery period and high cost limiting their clinical applications. Additive manufacturing (AM) could be an alternative to the existing drawbacks of the conventional NGCs fabrication methods. The emergence of the AM technique has offered ease for developing personalized three-dimensional (3D) neural constructs with intricate features and higher accuracy on a larger scale, replicating the native feature of nerve tissue. This review introduces the structural organization of peripheral nerves, the classification of PNI, and limitations in clinical and conventional nerve scaffold fabrication strategies. The principles and advantages of AM-based techniques, including the combinatorial approaches utilized for manufacturing 3D nerve conduits, are briefly summarized. This review also outlines the crucial parameters, such as the choice of printable biomaterials, 3D microstructural design/model, conductivity, permeability, degradation, mechanical property, and sterilization required to fabricate large-scale additive-manufactured NGCs successfully. Finally, the challenges and future directions toward fabricating the 3D-printed/bioprinted NGCs for clinical translation are also discussed.
Collapse
Affiliation(s)
- Allen Zennifer
- Tissue Engineering & Additive Manufacturing (TEAM) Lab, Centre for Nanotechnology & Advanced Biomaterials, ABCDE Innovation Centre, School of Chemical & Biotechnology, SASTRA Deemed University, India
| | - Madhumithra Thangadurai
- Tissue Engineering & Additive Manufacturing (TEAM) Lab, Centre for Nanotechnology & Advanced Biomaterials, ABCDE Innovation Centre, School of Chemical & Biotechnology, SASTRA Deemed University, India
| | - Dhakshinamoorthy Sundaramurthi
- Tissue Engineering & Additive Manufacturing (TEAM) Lab, Centre for Nanotechnology & Advanced Biomaterials, ABCDE Innovation Centre, School of Chemical & Biotechnology, SASTRA Deemed University, India
| | - Swaminathan Sethuraman
- Tissue Engineering & Additive Manufacturing (TEAM) Lab, Centre for Nanotechnology & Advanced Biomaterials, ABCDE Innovation Centre, School of Chemical & Biotechnology, SASTRA Deemed University, India.
| |
Collapse
|
41
|
Liu Y, Zhang X, Xiao C, Liu B. Engineered hydrogels for peripheral nerve repair. Mater Today Bio 2023; 20:100668. [PMID: 37273791 PMCID: PMC10232914 DOI: 10.1016/j.mtbio.2023.100668] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 05/06/2023] [Accepted: 05/16/2023] [Indexed: 06/06/2023] Open
Abstract
Peripheral nerve injury (PNI) is a complex disease that often appears in young adults. It is characterized by a high incidence, limited treatment options, and poor clinical outcomes. This disease not only causes dysfunction and psychological disorders in patients but also brings a heavy burden to the society. Currently, autologous nerve grafting is the gold standard in clinical treatment, but complications, such as the limited source of donor tissue and scar tissue formation, often further limit the therapeutic effect. Recently, a growing number of studies have used tissue-engineered materials to create a natural microenvironment similar to the nervous system and thus promote the regeneration of neural tissue and the recovery of impaired neural function with promising results. Hydrogels are often used as materials for the culture and differentiation of neurogenic cells due to their unique physical and chemical properties. Hydrogels can provide three-dimensional hydration networks that can be integrated into a variety of sizes and shapes to suit the morphology of neural tissues. In this review, we discuss the recent advances of engineered hydrogels for peripheral nerve repair and analyze the role of several different therapeutic strategies of hydrogels in PNI through the application characteristics of hydrogels in nerve tissue engineering (NTE). Furthermore, the prospects and challenges of the application of hydrogels in the treatment of PNI are also discussed.
Collapse
Affiliation(s)
- Yao Liu
- Hand and Foot Surgery Department, First Hospital of Jilin University, Xinmin Street, Changchun, 130061, PR China
| | - Xiaonong Zhang
- Key Laboratory of Polymer Ecomaterials, Jilin Biomedical Polymers Engineering Laboratory, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China
| | - Chunsheng Xiao
- Key Laboratory of Polymer Ecomaterials, Jilin Biomedical Polymers Engineering Laboratory, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China
| | - Bin Liu
- Hand and Foot Surgery Department, First Hospital of Jilin University, Xinmin Street, Changchun, 130061, PR China
| |
Collapse
|
42
|
Stocco E, Barbon S, Emmi A, Tiengo C, Macchi V, De Caro R, Porzionato A. Bridging Gaps in Peripheral Nerves: From Current Strategies to Future Perspectives in Conduit Design. Int J Mol Sci 2023; 24:ijms24119170. [PMID: 37298122 DOI: 10.3390/ijms24119170] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 05/17/2023] [Accepted: 05/22/2023] [Indexed: 06/12/2023] Open
Abstract
In peripheral nerve injuries (PNI) with substance loss, where tensionless end-to-end suture is not achievable, the positioning of a graft is required. Available options include autografts (e.g., sural nerve, medial and lateral antebrachial cutaneous nerves, superficial branch of the radial nerve), allografts (Avance®; human origin), and hollow nerve conduits. There are eleven commercial hollow conduits approved for clinical, and they consist of devices made of a non-biodegradable synthetic polymer (polyvinyl alcohol), biodegradable synthetic polymers (poly(DL-lactide-ε-caprolactone); polyglycolic acid), and biodegradable natural polymers (collagen type I with/without glycosaminoglycan; chitosan; porcine small intestinal submucosa); different resorption times are available for resorbable guides, ranging from three months to four years. Unfortunately, anatomical/functional nerve regeneration requirements are not satisfied by any of the possible alternatives; to date, focusing on wall and/or inner lumen organization/functionalization seems to be the most promising strategy for next-generation device fabrication. Porous or grooved walls as well as multichannel lumens and luminal fillers are the most intriguing options, eventually also including the addition of cells (Schwann cells, bone marrow-derived, and adipose tissue derived stem cells) to support nerve regeneration. This review aims to describe common alternatives for severe PNI recovery with a highlight of future conduits.
Collapse
Affiliation(s)
- Elena Stocco
- Section of Human Anatomy, Department of Neuroscience, University of Padova, 35121 Padova, Italy
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, Padova University Hospital, Via Giustiniani, 2, 35128 Padova, Italy
- L.i.f.e.L.a.b. Program, Consorzio per la Ricerca Sanitaria (CORIS), Veneto Region, 35128 Padova, Italy
- Foundation for Biology and Regenerative Medicine, Tissue Engineering and Signaling-TES, Onlus, 35030 Padova, Italy
| | - Silvia Barbon
- Section of Human Anatomy, Department of Neuroscience, University of Padova, 35121 Padova, Italy
- L.i.f.e.L.a.b. Program, Consorzio per la Ricerca Sanitaria (CORIS), Veneto Region, 35128 Padova, Italy
- Foundation for Biology and Regenerative Medicine, Tissue Engineering and Signaling-TES, Onlus, 35030 Padova, Italy
| | - Aron Emmi
- Section of Human Anatomy, Department of Neuroscience, University of Padova, 35121 Padova, Italy
| | - Cesare Tiengo
- Plastic Surgery Unit, Department of Neuroscience, University of Padova, 35121 Padova, Italy
| | - Veronica Macchi
- Section of Human Anatomy, Department of Neuroscience, University of Padova, 35121 Padova, Italy
- L.i.f.e.L.a.b. Program, Consorzio per la Ricerca Sanitaria (CORIS), Veneto Region, 35128 Padova, Italy
| | - Raffaele De Caro
- Section of Human Anatomy, Department of Neuroscience, University of Padova, 35121 Padova, Italy
- L.i.f.e.L.a.b. Program, Consorzio per la Ricerca Sanitaria (CORIS), Veneto Region, 35128 Padova, Italy
| | - Andrea Porzionato
- Section of Human Anatomy, Department of Neuroscience, University of Padova, 35121 Padova, Italy
- L.i.f.e.L.a.b. Program, Consorzio per la Ricerca Sanitaria (CORIS), Veneto Region, 35128 Padova, Italy
| |
Collapse
|
43
|
Dong Q, Yang X, Liang X, Liu J, Wang B, Zhao Y, Huselstein C, Feng X, Tong Z, Chen Y. Composite Hydrogel Conduit Incorporated with Platelet-Rich Plasma Improved the Regenerative Microenvironment for Peripheral Nerve Repair. ACS APPLIED MATERIALS & INTERFACES 2023; 15:24120-24133. [PMID: 37162458 DOI: 10.1021/acsami.3c02548] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Peripheral nerve regeneration and functional recovery remain major challenges in clinical practice. Nerve guidance conduits (NGCs) which can regulate the regenerative microenvironment are beneficial for peripheral nerve repair. Platelet-rich plasma (PRP) can secrete multiple growth factors to regulate the regenerative microenvironment. However, current administration methods of PRP are rapidly activated followed by the burst release of growth factors, causing low therapeutic efficiency in vivo. To overcome these disadvantages, a composite nerve conduit was fabricated by incorporating PRP into a gelatin methacrylate (GelMA) and sodium alginate (SA) hydrogel. The GelMA/SA-3/PRP-20 NGCs possess optimal mechanical properties, degradation rate, and superior biological performance. Importantly, GelMA/SA-3/PRP-20 NGCs achieved the sustained release of two major growth factors (VEGF-A, PDGF-BB) from PRP. Moreover, the GelMA/SA-3/PRP-20 NGCs significantly promoted the migration of Schwann cells and the neovascularization of endothelial cells in vitro. While bridging 10 mm rat sciatic nerve defects, the GelMA/SA-3/PRP-20 NGCs promoted axonal regeneration and functional recovery of peripheral nerves. Therefore, the GelMA/SA-3/PRP-20 NGCs could regulate the regenerative microenvironment by sustained release of growth factors from PRP and shed new light on the clinical application of PRP in peripheral nerve repair.
Collapse
Affiliation(s)
- Qi Dong
- Department of Biomedical Engineering and Hubei Province Key Laboratory of Allergy and Immune Related Disease, TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan 430071, China
| | - Xindi Yang
- Department of Biomedical Engineering and Hubei Province Key Laboratory of Allergy and Immune Related Disease, TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan 430071, China
| | - Xiao Liang
- Department of Biomedical Engineering and Hubei Province Key Laboratory of Allergy and Immune Related Disease, TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan 430071, China
| | - Jing Liu
- Department of Biomedical Engineering and Hubei Province Key Laboratory of Allergy and Immune Related Disease, TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan 430071, China
| | - Binyi Wang
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan 430071, China
| | - Yanteng Zhao
- Department of Blood Transfusion, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Céline Huselstein
- UMR 7365 CNRS, Medical School, University of Lorraine, 54505 Nancy, France
| | | | - Zan Tong
- Department of Biomedical Engineering and Hubei Province Key Laboratory of Allergy and Immune Related Disease, TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan 430071, China
| | - Yun Chen
- Department of Biomedical Engineering and Hubei Province Key Laboratory of Allergy and Immune Related Disease, TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan 430071, China
| |
Collapse
|
44
|
Valentino C, Vigani B, Zucca G, Ruggeri M, Marrubini G, Boselli C, Icaro Cornaglia A, Sandri G, Rossi S. Design of Novel Mechanically Resistant and Biodegradable Multichannel Platforms for the Treatment of Peripheral Nerve Injuries. Biomacromolecules 2023; 24:1731-1743. [PMID: 36922716 PMCID: PMC10091422 DOI: 10.1021/acs.biomac.2c01498] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 03/06/2023] [Indexed: 03/18/2023]
Abstract
Peripheral nerve injury is one of the most debilitating pathologies that severely impair patients' life. Although many efforts have been made to advance in the treatment of such a complex disorder, successful strategies to ensure full recovery are still scarce. The aim of the present work was to develop flexible and mechanically resistant platforms intended to act as a support and guide for neural cells during the regeneration process of peripheral nerve injury. For this purpose, poly(lactic-co-glycolic acid) (PLGA)/poly(d,l-lactic acid) (PDLLA)/poly(ethylene glycol) 400 (PEG)-multichannel-based scaffolds (MCs) were prepared through a multistep process involving electrospun microfibers coated with a polymer blend solution and used as a sacrificial mold. In particular, scaffolds characterized by random (MCR) and aligned (MCA) multichannel were obtained. A design of experiments approach (DoE) was employed to identify a scaffold-optimized composition. MCs were characterized for morphological and mechanical properties, suturability, degradability, cell colonization, and in vivo safety. A new biodegradable, biocompatible, and safe microscale multichannel scaffold was developed as the result of an easy multistep procedure.
Collapse
Affiliation(s)
- Caterina Valentino
- Department
of Drug Sciences, University of Pavia, Viale Taramelli, 12, 27100 Pavia, Italy
| | - Barbara Vigani
- Department
of Drug Sciences, University of Pavia, Viale Taramelli, 12, 27100 Pavia, Italy
| | - Gaia Zucca
- Department
of Drug Sciences, University of Pavia, Viale Taramelli, 12, 27100 Pavia, Italy
| | - Marco Ruggeri
- Department
of Drug Sciences, University of Pavia, Viale Taramelli, 12, 27100 Pavia, Italy
| | - Giorgio Marrubini
- Department
of Drug Sciences, University of Pavia, Viale Taramelli, 12, 27100 Pavia, Italy
| | - Cinzia Boselli
- Department
of Drug Sciences, University of Pavia, Viale Taramelli, 12, 27100 Pavia, Italy
| | - Antonia Icaro Cornaglia
- Department
of Public Health, Experimental, and Forensic Medicine, University of Pavia, Via Forlanini 2, 27100 Pavia, Italy
| | - Giuseppina Sandri
- Department
of Drug Sciences, University of Pavia, Viale Taramelli, 12, 27100 Pavia, Italy
| | - Silvia Rossi
- Department
of Drug Sciences, University of Pavia, Viale Taramelli, 12, 27100 Pavia, Italy
| |
Collapse
|
45
|
Fang Y, Wang C, Liu Z, Ko J, Chen L, Zhang T, Xiong Z, Zhang L, Sun W. 3D Printed Conductive Multiscale Nerve Guidance Conduit with Hierarchical Fibers for Peripheral Nerve Regeneration. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2205744. [PMID: 36808712 PMCID: PMC10131803 DOI: 10.1002/advs.202205744] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 01/27/2023] [Indexed: 06/18/2023]
Abstract
Nerve guidance conduits (NGCs) have become a promising alternative for peripheral nerve regeneration; however, the outcome of nerve regeneration and functional recovery is greatly affected by the physical, chemical, and electrical properties of NGCs. In this study, a conductive multiscale filled NGC (MF-NGC) consisting of electrospun poly(lactide-co-caprolactone) (PCL)/collagen nanofibers as the sheath, reduced graphene oxide /PCL microfibers as the backbone, and PCL microfibers as the internal structure for peripheral nerve regeneration is developed. The printed MF-NGCs presented good permeability, mechanical stability, and electrical conductivity, which further promoted the elongation and growth of Schwann cells and neurite outgrowth of PC12 neuronal cells. Animal studies using a rat sciatic nerve injury model reveal that the MF-NGCs promote neovascularization and M2 transition through the rapid recruitment of vascular cells and macrophages. Histological and functional assessments of the regenerated nerves confirm that the conductive MF-NGCs significantly enhance peripheral nerve regeneration, as indicated by improved axon myelination, muscle weight increase, and sciatic nerve function index. This study demonstrates the feasibility of using 3D-printed conductive MF-NGCs with hierarchically oriented fibers as functional conduits that can significantly enhance peripheral nerve regeneration.
Collapse
Affiliation(s)
- Yongcong Fang
- Biomanufacturing CenterDepartment of Mechanical EngineeringTsinghua UniversityBeijing100084P. R. China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of BeijingBeijing100084P. R. China
- “Biomanufacturing and Engineering Living Systems” Innovation International Talents Base (111 Base)Beijing100084P. R. China
| | - Chengjin Wang
- Biomanufacturing CenterDepartment of Mechanical EngineeringTsinghua UniversityBeijing100084P. R. China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of BeijingBeijing100084P. R. China
- “Biomanufacturing and Engineering Living Systems” Innovation International Talents Base (111 Base)Beijing100084P. R. China
| | - Zibo Liu
- Biomanufacturing CenterDepartment of Mechanical EngineeringTsinghua UniversityBeijing100084P. R. China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of BeijingBeijing100084P. R. China
- “Biomanufacturing and Engineering Living Systems” Innovation International Talents Base (111 Base)Beijing100084P. R. China
| | - Jeonghoon Ko
- Biomanufacturing CenterDepartment of Mechanical EngineeringTsinghua UniversityBeijing100084P. R. China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of BeijingBeijing100084P. R. China
- “Biomanufacturing and Engineering Living Systems” Innovation International Talents Base (111 Base)Beijing100084P. R. China
| | - Li Chen
- Biomanufacturing CenterDepartment of Mechanical EngineeringTsinghua UniversityBeijing100084P. R. China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of BeijingBeijing100084P. R. China
- “Biomanufacturing and Engineering Living Systems” Innovation International Talents Base (111 Base)Beijing100084P. R. China
| | - Ting Zhang
- Biomanufacturing CenterDepartment of Mechanical EngineeringTsinghua UniversityBeijing100084P. R. China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of BeijingBeijing100084P. R. China
- “Biomanufacturing and Engineering Living Systems” Innovation International Talents Base (111 Base)Beijing100084P. R. China
| | - Zhuo Xiong
- Biomanufacturing CenterDepartment of Mechanical EngineeringTsinghua UniversityBeijing100084P. R. China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of BeijingBeijing100084P. R. China
- “Biomanufacturing and Engineering Living Systems” Innovation International Talents Base (111 Base)Beijing100084P. R. China
| | - Lei Zhang
- Biomanufacturing CenterDepartment of Mechanical EngineeringTsinghua UniversityBeijing100084P. R. China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of BeijingBeijing100084P. R. China
- “Biomanufacturing and Engineering Living Systems” Innovation International Talents Base (111 Base)Beijing100084P. R. China
| | - Wei Sun
- Biomanufacturing CenterDepartment of Mechanical EngineeringTsinghua UniversityBeijing100084P. R. China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of BeijingBeijing100084P. R. China
- “Biomanufacturing and Engineering Living Systems” Innovation International Talents Base (111 Base)Beijing100084P. R. China
- Department of Mechanical EngineeringDrexel UniversityPhiladelphiaPA19104USA
| |
Collapse
|
46
|
Wei L, Wang S, Shan M, Li Y, Wang Y, Wang F, Wang L, Mao J. Conductive fibers for biomedical applications. Bioact Mater 2023; 22:343-364. [PMID: 36311045 PMCID: PMC9588989 DOI: 10.1016/j.bioactmat.2022.10.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 09/12/2022] [Accepted: 10/07/2022] [Indexed: 11/26/2022] Open
Abstract
Bioelectricity has been stated as a key factor in regulating cell activity and tissue function in electroactive tissues. Thus, various biomedical electronic constructs have been developed to interfere with cell behaviors to promote tissue regeneration, or to interface with cells or tissue/organ surfaces to acquire physiological status via electrical signals. Benefiting from the outstanding advantages of flexibility, structural diversity, customizable mechanical properties, and tunable distribution of conductive components, conductive fibers are able to avoid the damage-inducing mechanical mismatch between the construct and the biological environment, in return to ensure stable functioning of such constructs during physiological deformation. Herein, this review starts by presenting current fabrication technologies of conductive fibers including wet spinning, microfluidic spinning, electrospinning and 3D printing as well as surface modification on fibers and fiber assemblies. To provide an update on the biomedical applications of conductive fibers and fiber assemblies, we further elaborate conductive fibrous constructs utilized in tissue engineering and regeneration, implantable healthcare bioelectronics, and wearable healthcare bioelectronics. To conclude, current challenges and future perspectives of biomedical electronic constructs built by conductive fibers are discussed.
Collapse
Affiliation(s)
- Leqian Wei
- Key Laboratory of Textile Science & Technology, Ministry of Education, College of Textiles, Donghua University, Shanghai, 201620, China
- Key Laboratory of Textile Industry for Biomedical Textile Materials and Technology, Donghua University, Shanghai, 201620, China
| | - Shasha Wang
- Key Laboratory of Textile Science & Technology, Ministry of Education, College of Textiles, Donghua University, Shanghai, 201620, China
- Key Laboratory of Textile Industry for Biomedical Textile Materials and Technology, Donghua University, Shanghai, 201620, China
| | - Mengqi Shan
- Key Laboratory of Textile Science & Technology, Ministry of Education, College of Textiles, Donghua University, Shanghai, 201620, China
- Key Laboratory of Textile Industry for Biomedical Textile Materials and Technology, Donghua University, Shanghai, 201620, China
| | - Yimeng Li
- Key Laboratory of Textile Science & Technology, Ministry of Education, College of Textiles, Donghua University, Shanghai, 201620, China
- Key Laboratory of Textile Industry for Biomedical Textile Materials and Technology, Donghua University, Shanghai, 201620, China
| | - Yongliang Wang
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao City, Shandong Province, 266071, China
| | - Fujun Wang
- Key Laboratory of Textile Science & Technology, Ministry of Education, College of Textiles, Donghua University, Shanghai, 201620, China
- Key Laboratory of Textile Industry for Biomedical Textile Materials and Technology, Donghua University, Shanghai, 201620, China
| | - Lu Wang
- Key Laboratory of Textile Science & Technology, Ministry of Education, College of Textiles, Donghua University, Shanghai, 201620, China
- Key Laboratory of Textile Industry for Biomedical Textile Materials and Technology, Donghua University, Shanghai, 201620, China
| | - Jifu Mao
- Key Laboratory of Textile Science & Technology, Ministry of Education, College of Textiles, Donghua University, Shanghai, 201620, China
- Key Laboratory of Textile Industry for Biomedical Textile Materials and Technology, Donghua University, Shanghai, 201620, China
| |
Collapse
|
47
|
Sumam P, Parameswaran R. Neuronal cell response on aligned fibroporous electrospun mat generated from silver ion complexed ethylene vinyl alcohol copolymer. J Biomed Mater Res B Appl Biomater 2023; 111:782-794. [PMID: 36333924 DOI: 10.1002/jbm.b.35189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 09/12/2022] [Accepted: 10/10/2022] [Indexed: 11/07/2022]
Abstract
Generating electrospun mats with aligned fibers and obtaining neurite extension in the aligned fiber direction could provide hope for fabricating nerve guidance conduits or wraps through an easy method. The growing interest in generating electrospun mats with aligned fibers for tissue engineering is looking for simple methods to generate the same. Here, in this study, ethylene vinyl alcohol copolymer (EVAL) chains were complexed with silver ions (Ag+ ) to generate aligned fibers during the electrospinning process. The fibers thus produced were subjected to physico-chemical characterization and biological studies to ensure their properties and to examine whether suitable for neuronal cell attachment and neurite extension that may be useful in making nerve guidance conduits or wraps. The presence of silver ions and its complex formation with -OH of EVAL has been confirmed with EDX and XPS analysis respectively. The alignment of fibers was visualized from SEM analysis and confirmed using directionality analysis using Fiji-ImageJ software. Mechanical properties done with dumbbells punched out in longitudinal and transverse directions also substantiated the alignment of fibers. The results obtained from direct contact, MTT, and live/dead assay showed the cells are viable on the material. From the actin staining and immunostaining assays, it was evident that the PC12 cells could attach and extend their neurites in an aligned manner on the fibers. The maximum neurite extension was up to 200 μm in length. These properties of electrospun EVAL-Ag mat with aligned fibers indicated that it could be developed as a biocompatible nerve guidance conduit or wrap.
Collapse
Affiliation(s)
- Prima Sumam
- Division of Polymeric Medical Devices, Department of Medical Devices Engineering, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, India
| | - Ramesh Parameswaran
- Division of Polymeric Medical Devices, Department of Medical Devices Engineering, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, India
| |
Collapse
|
48
|
García-García ÓD, El Soury M, Campos F, Sánchez-Porras D, Geuna S, Alaminos M, Gambarotta G, Chato-Astrain J, Raimondo S, Carriel V. Comprehensive ex vivo and in vivo preclinical evaluation of novel chemo enzymatic decellularized peripheral nerve allografts. Front Bioeng Biotechnol 2023; 11:1162684. [PMID: 37082209 PMCID: PMC10111265 DOI: 10.3389/fbioe.2023.1162684] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 03/22/2023] [Indexed: 04/07/2023] Open
Abstract
As a reliable alternative to autografts, decellularized peripheral nerve allografts (DPNAs) should mimic the complex microstructure of native nerves and be immunogenically compatible. Nevertheless, there is a current lack of decellularization methods able to remove peripheral nerve cells without significantly altering the nerve extracellular matrix (ECM). The aims of this study are firstly to characterize ex vivo, in a histological, biochemical, biomechanical and ultrastructural way, three novel chemical-enzymatic decellularization protocols (P1, P2 and P3) in rat sciatic nerves and compared with the Sondell classic decellularization method and then, to select the most promising DPNAs to be tested in vivo. All the DPNAs generated present an efficient removal of the cellular material and myelin, while preserving the laminin and collagen network of the ECM (except P3) and were free from any significant alterations in the biomechanical parameters and biocompatibility properties. Then, P1 and P2 were selected to evaluate their regenerative effectivity and were compared with Sondell and autograft techniques in an in vivo model of sciatic defect with a 10-mm gap, after 15 weeks of follow-up. All study groups showed a partial motor and sensory recovery that were in correlation with the histological, histomorphometrical and ultrastructural analyses of nerve regeneration, being P2 the protocol showing the most similar results to the autograft control group.
Collapse
Affiliation(s)
- Óscar Darío García-García
- Tissue Engineering Group, Department of Histology, University of Granada and Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain
- Doctoral Program in Biomedicine, University of Granada, Granada, Spain
- Department of Clinical and Biological Sciences and Neuroscience Institute Cavalieri Ottolenghi (NICO), University of Torino, Orbassano, Italy
| | - Marwa El Soury
- Tissue Engineering Group, Department of Histology, University of Granada and Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain
- Department of Clinical and Biological Sciences and Neuroscience Institute Cavalieri Ottolenghi (NICO), University of Torino, Orbassano, Italy
| | - Fernando Campos
- Tissue Engineering Group, Department of Histology, University of Granada and Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain
| | - David Sánchez-Porras
- Tissue Engineering Group, Department of Histology, University of Granada and Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain
| | - Stefano Geuna
- Department of Clinical and Biological Sciences and Neuroscience Institute Cavalieri Ottolenghi (NICO), University of Torino, Orbassano, Italy
| | - Miguel Alaminos
- Tissue Engineering Group, Department of Histology, University of Granada and Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain
| | - Giovanna Gambarotta
- Department of Clinical and Biological Sciences and Neuroscience Institute Cavalieri Ottolenghi (NICO), University of Torino, Orbassano, Italy
| | - Jesús Chato-Astrain
- Tissue Engineering Group, Department of Histology, University of Granada and Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain
- *Correspondence: Jesús Chato-Astrain, ; Víctor Carriel,
| | - Stefania Raimondo
- Department of Clinical and Biological Sciences and Neuroscience Institute Cavalieri Ottolenghi (NICO), University of Torino, Orbassano, Italy
| | - Víctor Carriel
- Tissue Engineering Group, Department of Histology, University of Granada and Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain
- *Correspondence: Jesús Chato-Astrain, ; Víctor Carriel,
| |
Collapse
|
49
|
Perrelle JM, Boreland AJ, Gamboa JM, Gowda P, Murthy NS. Biomimetic Strategies for Peripheral Nerve Injury Repair: An Exploration of Microarchitecture and Cellularization. BIOMEDICAL MATERIALS & DEVICES (NEW YORK, N.Y.) 2023; 1:21-37. [PMID: 38343513 PMCID: PMC10857769 DOI: 10.1007/s44174-022-00039-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 09/14/2022] [Indexed: 02/15/2024]
Abstract
Injuries to the nervous system present formidable challenges to scientists, clinicians, and patients. While regeneration within the central nervous system is minimal, peripheral nerves can regenerate, albeit with limitations. The regenerative mechanisms of the peripheral nervous system thus provide fertile ground for clinical and scientific advancement, and opportunities to learn fundamental lessons regarding nerve behavior in the context of regeneration, particularly the relationship of axons to their support cells and the extracellular matrix environment. However, few current interventions adequately address peripheral nerve injuries. This article aims to elucidate areas in which progress might be made toward developing better interventions, particularly using synthetic nerve grafts. The article first provides a thorough review of peripheral nerve anatomy, physiology, and the regenerative mechanisms that occur in response to injury. This is followed by a discussion of currently available interventions for peripheral nerve injuries. Promising biomaterial fabrication techniques which aim to recapitulate nerve architecture, along with approaches to enhancing these biomaterial scaffolds with growth factors and cellular components, are then described. The final section elucidates specific considerations when developing nerve grafts, including utilizing induced pluripotent stem cells, Schwann cells, nerve growth factors, and multilayered structures that mimic the architectures of the natural nerve.
Collapse
Affiliation(s)
- Jeremy M. Perrelle
- Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Andrew J. Boreland
- Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
- Graduate Program in Molecular Biosciences, Rutgers University, Piscataway, NJ, USA
| | - Jasmine M. Gamboa
- Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Prarthana Gowda
- Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - N. Sanjeeva Murthy
- Laboratory for Biomaterials Research, Department of Chemistry and Chemical Biology, Rutgers University, Piscataway, NJ 08854, USA
| |
Collapse
|
50
|
Jin B, Yu Y, Chen X, Yang Y, Xiong Y, Im YJ, Zhao Y, Xiao J. Microtubes with gradient decellularized porcine sciatic nerve matrix from microfluidics for sciatic nerve regeneration. Bioact Mater 2023; 21:511-519. [PMID: 36185737 PMCID: PMC9508151 DOI: 10.1016/j.bioactmat.2022.08.027] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 07/28/2022] [Accepted: 08/16/2022] [Indexed: 11/21/2022] Open
Abstract
Long-range peripheral nerve defect is a severe and worldwide disease. With the increasing development of tissue engineering, the excellent ability of nerve extracellular matrix (ECM) in peripheral nerve injury (PNI) has been widely studied and verified. Here, we present a novel microtube that contains gradient decellularized porcine sciatic nerve ECM hydrogel (pDScNM-gel) from microfluidics for sciatic nerve regeneration. The pDScNM is confirmed to enhance cell proliferation and migration, and improve the axon growth of primary dorsal root ganglions (DRGs) in a concentration-related manner. These behaviors were also achieved when cells were co-cultured in a gradient pDScNM microtube. The in vivo sciatic nerve regeneration and functional recovery were also demonstrated by assembling the gradient pDScNM microtubes with a medical silicon tube. These results indicated that the microtubes with gradient pDScNM could act as a promising alternative for repairing peripheral nerve defects and showed great potential in clinical use.
Collapse
Affiliation(s)
- Binghui Jin
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang Province, 325035, China
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
- College of Pharmacy, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Yunru Yu
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325001, China
| | - Xiangxiang Chen
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang Province, 325035, China
| | - Yanhong Yang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang Province, 325035, China
| | - Yushan Xiong
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang Province, 325035, China
| | - Young Jun Im
- College of Pharmacy, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Yuanjin Zhao
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325001, China
| | - Jian Xiao
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang Province, 325035, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325001, China
| |
Collapse
|