1
|
Yergeshov A, Zoughaib M, Dayob K, Kamalov M, Luong D, Zakirova A, Mullin R, Salakhieva D, Abdullin TI. Newly Designed PCL-Wrapped Cryogel-Based Conduit Activated with IKVAV Peptide Derivative for Peripheral Nerve Repair. Pharmaceutics 2024; 16:1569. [PMID: 39771548 PMCID: PMC11677967 DOI: 10.3390/pharmaceutics16121569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 11/08/2024] [Accepted: 12/05/2024] [Indexed: 01/11/2025] Open
Abstract
Background: The combination of macroporous cryogels with synthetic peptide factors represents a promising but poorly explored strategy for the development of extracellular matrix (ECM)-mimicking scaffolds for peripheral nerve (PN) repair. Methods: In this study, IKVAV peptide was functionalized with terminal lysine residues to allow its in situ cross-linking with gelatin macromer, resulting in the formation of IKVAV-containing proteinaceous cryogels. The controllable inclusion and distribution of the peptide molecules within the scaffold was verified using a fluorescently labelled peptide counterpart. The optimized cryogel scaffold was combined with polycaprolactone (PCL)-based shell tube to form a suturable nerve conduit (NC) to be implanted into sciatic nerve diastasis in rats. Results: The NC constituents did not impair the viability of primary skin fibroblasts. Concentration-dependent effects of the peptide component on interrelated viscoelastic and swelling properties of the cryogels as well as on proliferation and morphological differentiation of neurogenic PC-12 cells were established, also indicating the existence of an optimal-density range of the introduced peptide. The in vivo implanted NC sustained the connection of the nerve stumps with partial degradation of the PCL tube over eight weeks, whereas the core-filling cryogel profoundly improved local electromyographic recovery and morphological repair of the nerve tissues, confirming the regenerative activity of the developed scaffold. Conclusions: These results provide proof-of-concept for the development of a newly designed PN conduit prototype based on IKVAV-activated cryogel, and they can be exploited to create other ECM-mimicking scaffolds.
Collapse
Affiliation(s)
- Abdulla Yergeshov
- Scientific and Educational Center of Pharmaceutics, Kazan (Volga Region) Federal University, 18 Kremlyovskaya St., 420008 Kazan, Russia (M.Z.); (K.D.)
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, 18 Kremlyovskaya St., 420008 Kazan, Russia
| | - Mohamed Zoughaib
- Scientific and Educational Center of Pharmaceutics, Kazan (Volga Region) Federal University, 18 Kremlyovskaya St., 420008 Kazan, Russia (M.Z.); (K.D.)
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, 18 Kremlyovskaya St., 420008 Kazan, Russia
| | - Kenana Dayob
- Scientific and Educational Center of Pharmaceutics, Kazan (Volga Region) Federal University, 18 Kremlyovskaya St., 420008 Kazan, Russia (M.Z.); (K.D.)
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, 18 Kremlyovskaya St., 420008 Kazan, Russia
| | - Marat Kamalov
- Scientific and Educational Center of Pharmaceutics, Kazan (Volga Region) Federal University, 18 Kremlyovskaya St., 420008 Kazan, Russia (M.Z.); (K.D.)
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, 18 Kremlyovskaya St., 420008 Kazan, Russia
| | - Duong Luong
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072, Australia
| | - Albina Zakirova
- Academy of Postgraduate Education under FSBU FSCC of FMBA of Russia, Department of Oncology and Plastic Surgery, 91 Volokolamsk Highway, 125371 Moscow, Russia
| | - Ruslan Mullin
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, 18 Kremlyovskaya St., 420008 Kazan, Russia
- State Autonomous Healthcare Institution Republican Clinical Hospital of the Ministry of Health of the Republic of Tatarstan, 138 Orenburg Highway, 420064 Kazan, Russia
| | - Diana Salakhieva
- Scientific and Educational Center of Pharmaceutics, Kazan (Volga Region) Federal University, 18 Kremlyovskaya St., 420008 Kazan, Russia (M.Z.); (K.D.)
| | - Timur I. Abdullin
- Scientific and Educational Center of Pharmaceutics, Kazan (Volga Region) Federal University, 18 Kremlyovskaya St., 420008 Kazan, Russia (M.Z.); (K.D.)
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, 18 Kremlyovskaya St., 420008 Kazan, Russia
| |
Collapse
|
2
|
QingNing S, Mohd Ismail ZI, Ab Patar MNA, Mat Lazim N, Hadie SNH, Mohd Noor NF. The limelight of adipose-derived stem cells in the landscape of neural tissue engineering for peripheral nerve injury. Tissue Cell 2024; 91:102556. [PMID: 39293138 DOI: 10.1016/j.tice.2024.102556] [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: 03/25/2024] [Revised: 09/04/2024] [Accepted: 09/05/2024] [Indexed: 09/20/2024]
Abstract
BACKGROUND AND AIMS Challenges in treating peripheral nerve injury include prolonged repair time and insufficient functional recovery. Stem cell therapy coupled with neural tissue engineering has been shown to induce nerve regeneration following peripheral nerve injury. Among these stem cells, adipose-derived stem cells (ADSCs) are preferred due to their accessibility, expansion, multidirectional differentiation, and production of essential nutrient factors for nerve growth. In recent years, ADSC-laden nerve guide conduit has been utilized to enhance the therapeutic effects of tissue-engineered nerve grafts. This review explores existing research that recognizes the roles played by ADSCs in inducing peripheral nerve regeneration following injury and summarizes the different methods of application of ADSC-laden nerve conduit in neural tissue engineering.
Collapse
Affiliation(s)
- Sun QingNing
- Department of Anatomy, School of Medical Sciences, Universiti Sains Malaysia Health Campus, Kubang Kerian, Kelantan 16150, Malaysia; Department of Rehabilitation, School of Special Education, Zhengzhou Normal University, Zhengzhou 450044, China.
| | - Zul Izhar Mohd Ismail
- Department of Anatomy, School of Medical Sciences, Universiti Sains Malaysia Health Campus, Kubang Kerian, Kelantan 16150, Malaysia.
| | - Mohd Nor Azim Ab Patar
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia Health Campus, Kubang Kerian, Kelantan 16150, Malaysia.
| | - Norhafiza Mat Lazim
- Department of Otorhinolaryngology-Head & Neck Surgery, School of Medical Sciences, Universiti Sains Malaysia Health Campus, Kubang Kerian, Kelantan 16150, Malaysia.
| | - Siti Nurma Hanim Hadie
- Department of Anatomy, School of Medical Sciences, Universiti Sains Malaysia Health Campus, Kubang Kerian, Kelantan 16150, Malaysia.
| | - Nor Farid Mohd Noor
- Faculty of Medicine, Universiti Sultan Zainal Abidin Medical Campus, Kuala Terengganu, Terengganu 20400, Malaysia.
| |
Collapse
|
3
|
Wen Y, Li Y, Yang R, Chen Y, Shen Y, Liu Y, Liu X, Zhang B, Li H. Biofunctional coatings and drug-coated stents for restenosis therapy. Mater Today Bio 2024; 29:101259. [PMID: 39391793 PMCID: PMC11465131 DOI: 10.1016/j.mtbio.2024.101259] [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: 07/08/2024] [Revised: 09/07/2024] [Accepted: 09/17/2024] [Indexed: 10/12/2024] Open
Abstract
Palliative therapy utilizing interventional stents, such as vascular stents, biliary stents, esophageal stents, and other stents, has been a prevalent clinical strategy for treating duct narrowing and partial blockage. However, stent restenosis after implantation usually significantly compromises therapeutic efficacy and patient safety. Clinically, vascular stent restenosis is primarily attributed to endothelial hyperplasia and coagulation, while the risk of biliary stent occlusion is heightened by bacterial adhesion and bile sludge accumulation. Similarly, granulation tissue hyperplasia leads to tracheal stent restenosis. To address these issues, surface modifications of stents are extensively adopted as effective strategies to reduce the probability of restenosis and extend their functional lifespan. Applying coatings is one of the technical routes involving a complex selection of materials, drug loading capacities, release rates, and other factors. This paper provides an extensive overview of state of the art drug-coated stents, addressing both challenges and future prospects in this domain. We aim to contribute positively to the ongoing development and potential clinical applications of drug-coated stents in interventional therapy.
Collapse
Affiliation(s)
- Yanghui Wen
- Departments of General Surgery, Ningbo No.2 Hospital, Ningbo, 315010, China
| | - Yihuan Li
- Zhejiang Engineering Research Center for Biomedical Materials, Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
- Zhejiang-Japan Joint Laboratory for Antibacterial and Antifouling Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
| | - Rui Yang
- Zhejiang Engineering Research Center for Biomedical Materials, Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
- Zhejiang-Japan Joint Laboratory for Antibacterial and Antifouling Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
| | - Yunjie Chen
- Departments of General Surgery, Ningbo No.2 Hospital, Ningbo, 315010, China
| | - Yan Shen
- Zhejiang Engineering Research Center for Biomedical Materials, Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
- Zhejiang-Japan Joint Laboratory for Antibacterial and Antifouling Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
| | - Yi Liu
- Zhejiang Engineering Research Center for Biomedical Materials, Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
- Zhejiang-Japan Joint Laboratory for Antibacterial and Antifouling Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
| | - Xiaomei Liu
- Zhejiang Engineering Research Center for Biomedical Materials, Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
- Zhejiang-Japan Joint Laboratory for Antibacterial and Antifouling Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
| | - Botao Zhang
- Zhejiang Engineering Research Center for Biomedical Materials, Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
- Zhejiang-Japan Joint Laboratory for Antibacterial and Antifouling Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
| | - Hua Li
- Zhejiang Engineering Research Center for Biomedical Materials, Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
- Zhejiang-Japan Joint Laboratory for Antibacterial and Antifouling Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
| |
Collapse
|
4
|
Ramesh PA, Sethuraman S, Subramanian A. Fabrication of Anatomically Equivalent Pectin-Based Multifilament Nerve Conduits. ACS APPLIED BIO MATERIALS 2024; 7:6706-6719. [PMID: 39349393 DOI: 10.1021/acsabm.4c00872] [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: 10/02/2024]
Abstract
Reuniting denuded nerve ends after a long segmental peripheral nerve defect is challenging due to delayed axonal regeneration and incomplete, nonspecific reinnervation, as conventional hollow nerve guides fail to ensure proper fascicular complementation and obstruct axonal guidance across the defects. This study focuses on fabricating multifilament conduits using a plant-derived anionic polysaccharide, pectin, where the abundant availability of carboxylate (COO-) functional groups in pectin facilitates instantaneous sol-gel transition upon interaction with divalent cations. Despite their advantages, pectin hydrogels encounter structural instability under physiological conditions. Hence, pectin is conjugated with light-sensitive methacrylate residues (49.8% methacrylation) to overcome these issues, enabling the fabrication of dual cross-linked multifilament nerve conduits through an ionic interaction-driven, template-free 3D wet writing process, followed by photo-cross-linking at 525 nm. The anatomical equivalence including peri-, epi-, and endoneurium structures of the customized multifilament conduits was confirmed through scanning electron micrographs and micro-CT analysis of rat and goat sciatic nerve tissues. Furthermore, the fabricated multifilament nerve conduits demonstrated cytocompatibility and promoted the expression of neuron-specific intermediate filament protein (NF-200) in PC12 cells and neurite outgrowth of 16.90 ± 1.82 μm on day 14. Micro-CT imaging of an anastomosed native goat sciatic nerve with an 8-filament conduit demonstrated precise fascicular complementation in an ex vivo interpositional goat model. This approach not only eliminates the need for a suture-intensive ligation process but also highlights the customizability of multifilament conduits to meet patient- and injury-specific needs.
Collapse
Affiliation(s)
- Preethy Amruthavarshini Ramesh
- Centre for Nanotechnology and Advanced Biomaterials, ABCDE Innovation Centre, School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur 613 401, India
| | - Swaminathan Sethuraman
- Centre for Nanotechnology and Advanced Biomaterials, ABCDE Innovation Centre, School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur 613 401, India
| | - Anuradha Subramanian
- Centre for Nanotechnology and Advanced Biomaterials, ABCDE Innovation Centre, School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur 613 401, India
| |
Collapse
|
5
|
Harley-Troxell ME, Steiner R, Newby SD, Bow AJ, Masi TJ, Millis N, Matavosian AA, Crouch D, Stephenson S, Anderson DE, Dhar M. Electrospun PCL Nerve Wrap Coated with Graphene Oxide Supports Axonal Growth in a Rat Sciatic Nerve Injury Model. Pharmaceutics 2024; 16:1254. [PMID: 39458586 PMCID: PMC11510652 DOI: 10.3390/pharmaceutics16101254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 08/29/2024] [Accepted: 09/23/2024] [Indexed: 10/28/2024] Open
Abstract
Background/Objectives: Peripheral nerve injuries (PNIs) are a debilitating problem, resulting in diminished quality of life due to the continued presence of both chronic and acute pain. The current standard of practice for the repair of PNIs larger than 10 mm is the use of autologous nerve grafts. Autologous nerve grafts have limitations that often result in outcomes that are not sufficient to remove motor and sensory impairments. Bio-mimetic nanocomposite scaffolds combined with mesenchymal stem cells (MSCs) represent a promising approach for PNIs. In this study, we investigated the potential of an electrospun wrap of polycaprolactone (PCL) + graphene oxide (GO), with and without xenogeneic human adipose tissue-derived MSCs (hADMSCs) to use as a platform for neural tissue engineering. Methods: We evaluated, in vitro and in vivo, the potential of the nerve wrap in providing support for axonal growth. To establish the rat sciatic nerve defect model, a 10 mm long limiting defect was created in the rat sciatic nerve of 18 Lewis rats. Rats treated with the nanocomposites were compared with autograft-treated defects. Gait, histological, and muscle analyses were performed after sacrifice at 12 weeks post-surgery. Results: Our findings demonstrate that hADMSCs had the potential to transdifferentiate into neural lineage and that the nanocomposite successfully delivered hADMSCs to the injury site. Histologically, we show that the PCL + GO nanocomposite with hADMSCs is comparable to the autologous nerve graft, to support and guide axonal growth. Conclusions: The novel PCL + GO nerve wrap and hADMSCs used in this study provide a foundation on which to build upon and generate future strategies for PNI repair.
Collapse
Affiliation(s)
- Meaghan E. Harley-Troxell
- Laboratory of Tissue Engineering and Regenerative Medicine, Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN 37996, USA; (M.E.H.-T.); (R.S.); (S.D.N.); (A.J.B.); (N.M.); (D.E.A.)
| | - Richard Steiner
- Laboratory of Tissue Engineering and Regenerative Medicine, Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN 37996, USA; (M.E.H.-T.); (R.S.); (S.D.N.); (A.J.B.); (N.M.); (D.E.A.)
| | - Steven D. Newby
- Laboratory of Tissue Engineering and Regenerative Medicine, Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN 37996, USA; (M.E.H.-T.); (R.S.); (S.D.N.); (A.J.B.); (N.M.); (D.E.A.)
| | - Austin J. Bow
- Laboratory of Tissue Engineering and Regenerative Medicine, Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN 37996, USA; (M.E.H.-T.); (R.S.); (S.D.N.); (A.J.B.); (N.M.); (D.E.A.)
| | - Thomas J. Masi
- Department of Surgery, University of Tennessee Graduate School of Medicine, Knoxville, TN 37996, USA;
| | - Nicholas Millis
- Laboratory of Tissue Engineering and Regenerative Medicine, Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN 37996, USA; (M.E.H.-T.); (R.S.); (S.D.N.); (A.J.B.); (N.M.); (D.E.A.)
| | - Alicia Adina Matavosian
- Department of Mechanical, Aerospace, and Biomedical Engineering, University of Tennessee, Knoxville, TN 37996, USA; (A.A.M.); (D.C.)
| | - Dustin Crouch
- Department of Mechanical, Aerospace, and Biomedical Engineering, University of Tennessee, Knoxville, TN 37996, USA; (A.A.M.); (D.C.)
| | - Stacy Stephenson
- Department of Plastic and Reconstructive Surgery, University of Tennessee Medical Center, Knoxville, TN 37920, USA;
| | - David E. Anderson
- Laboratory of Tissue Engineering and Regenerative Medicine, Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN 37996, USA; (M.E.H.-T.); (R.S.); (S.D.N.); (A.J.B.); (N.M.); (D.E.A.)
| | - Madhu Dhar
- Laboratory of Tissue Engineering and Regenerative Medicine, Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN 37996, USA; (M.E.H.-T.); (R.S.); (S.D.N.); (A.J.B.); (N.M.); (D.E.A.)
| |
Collapse
|
6
|
Dogny C, André-Lévigne D, Kalbermatten DF, Madduri S. Therapeutic Potential and Challenges of Mesenchymal Stem Cell-Derived Exosomes for Peripheral Nerve Regeneration: A Systematic Review. Int J Mol Sci 2024; 25:6489. [PMID: 38928194 PMCID: PMC11203969 DOI: 10.3390/ijms25126489] [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: 05/07/2024] [Revised: 06/03/2024] [Accepted: 06/06/2024] [Indexed: 06/28/2024] Open
Abstract
Gap injuries to the peripheral nervous system result in pain and loss of function, without any particularly effective therapeutic options. Within this context, mesenchymal stem cell (MSC)-derived exosomes have emerged as a potential therapeutic option. Thus, the focus of this study was to review currently available data on MSC-derived exosome-mounted scaffolds in peripheral nerve regeneration in order to identify the most promising scaffolds and exosome sources currently in the field of peripheral nerve regeneration. We conducted a systematic review following PRISMA 2020 guidelines. Exosome origins varied (adipose-derived MSCs, bone marrow MSCs, gingival MSC, induced pluripotent stem cells and a purified exosome product) similarly to the materials (Matrigel, alginate and silicone, acellular nerve graft [ANG], chitosan, chitin, hydrogel and fibrin glue). The compound muscle action potential (CMAP), sciatic functional index (SFI), gastrocnemius wet weight and histological analyses were used as main outcome measures. Overall, exosome-mounted scaffolds showed better regeneration than scaffolds alone. Functionally, both exosome-enriched chitin and ANG showed a significant improvement over time in the sciatica functional index, CMAP and wet weight. The best histological outcomes were found in the exosome-enriched ANG scaffold with a high increase in the axonal diameter and muscle cross-section area. Further studies are needed to confirm the efficacy of exosome-mounted scaffolds in peripheral nerve regeneration.
Collapse
Affiliation(s)
- Clelia Dogny
- Department of Plastic, Reconstructive and Aesthetic Surgery, Geneva University Hospitals, 1205 Geneva, Switzerland
| | - Dominik André-Lévigne
- Department of Plastic, Reconstructive and Aesthetic Surgery, Geneva University Hospitals, 1205 Geneva, Switzerland
| | - Daniel F. Kalbermatten
- Department of Plastic, Reconstructive and Aesthetic Surgery, Geneva University Hospitals, 1205 Geneva, Switzerland
- Bioengineering and Neuroregeneration Laboratory, Department of Surgery, University of Geneva, 1211 Geneva, Switzerland
| | - Srinivas Madduri
- Department of Plastic, Reconstructive and Aesthetic Surgery, Geneva University Hospitals, 1205 Geneva, Switzerland
- Bioengineering and Neuroregeneration Laboratory, Department of Surgery, University of Geneva, 1211 Geneva, Switzerland
| |
Collapse
|
7
|
Huang Y, Ye K, He A, Wan S, Wu M, Hu D, Xu K, Wei P, Yin J. Dual-layer conduit containing VEGF-A - Transfected Schwann cells promotes peripheral nerve regeneration via angiogenesis. Acta Biomater 2024; 180:323-336. [PMID: 38561075 DOI: 10.1016/j.actbio.2024.03.029] [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: 11/20/2023] [Revised: 03/22/2024] [Accepted: 03/27/2024] [Indexed: 04/04/2024]
Abstract
Peripheral nerve injuries (PNIs) can cause neuropathies and significantly affect the patient's quality of life. Autograft transplantation is the gold standard for conventional treatment; however, its application is limited by nerve unavailability, size mismatch, and local tissue adhesion. Tissue engineering, such as nerve guidance conduits, is an alternative and promising strategy to guide nerve regeneration for peripheral nerve repair; however, only a few conduits could reach the high repair efficiency of autografts. The healing process of PNI is frequently accompanied by not only axonal and myelination regeneration but also angiogenesis, which initializes nerve regeneration through vascular endothelial growth factor A (VEGF-A). In this study, a composite nerve conduit with a poly (lactic-co-glycolic acid) (PLGA) hollow tube as the outer layer and gelatin methacryloyl (GelMA) encapsulated with VEGF-A transfected Schwann cells (SCs) as the inner layer was established to evaluate its promising ability for peripheral nerve repair. A rat model of peripheral nerve defect was used to examine the efficiency of PLGA/GelMA-SC (VA) conduits, whereas autograft, PLGA, PLGA/GelMA, and PLGA/GelMA-SC (NC) were used as controls. VEGF-A-transfected SCs can provide a stable source for VEGF-A secretion. Furthermore, encapsulation in GelMA cannot only promote proliferation and tube formation of human umbilical vein endothelial cells but also enhance dorsal root ganglia and neuronal cell extension. Previous animal studies have demonstrated that the regenerative effects of PLGA/GelMA-SC (VA) nerve conduit were similar to those of autografts and much better than those of other conduits. These findings indicate that combination of VEGF-A-overexpressing SCs and PLGA/GelMA conduit-guided peripheral nerve repair provides a promising method that enhances angiogenesis and regeneration during nerve repair. STATEMENT OF SIGNIFICANCE: Nerve guidance conduits shows promise for peripheral nerve repair, while achieving the repair efficiency of autografts remains a challenge. In this study, a composite nerve conduit with a PLGA hollow tube as the outer layer and gelatin methacryloyl (GelMA) encapsulated with vascular endothelial growth factor A (VEGF-A)-transfected Schwann cells (SCs) as the inner layer was established to evaluate its potential ability for peripheral nerve repair. This approach preserves growth factor bioactivity and enhances material properties. GelMA insertion promotes Schwann cell proliferation and morphology extension. Moreover, transfected SCs serve as a stable VEGF-A source and fostering angiogenesis. This study offers a method preserving growth factor efficacy and safeguarding SCs, providing a comprehensive solution for enhanced angiogenesis and nerve regeneration.
Collapse
Affiliation(s)
- Yuye Huang
- Department of Plastic and Reconstructive Surgery, The First Affiliated Hospital of Ningbo University, Ningbo 315010, China; Center for Medical and Engineering Innovation, Central Laboratory, The First Affiliated Hospital of Ningbo University, Ningbo 315010, China
| | - Kai Ye
- Department of Plastic and Reconstructive Surgery, The First Affiliated Hospital of Ningbo University, Ningbo 315010, China
| | - Andong He
- Center for Medical and Engineering Innovation, Central Laboratory, The First Affiliated Hospital of Ningbo University, Ningbo 315010, China; Department of Respiratory and Critical Care Medicine, Key Laboratory of Respiratory Disease of Ningbo, The First Affiliated Hospital of Ningbo University, Ningbo 315010, China
| | - Shaobo Wan
- Yuyao Traditional Chinese Medicine Hospital, Ningbo 315010, China
| | - Miaoben Wu
- Department of Plastic and Reconstructive Surgery, The First Affiliated Hospital of Ningbo University, Ningbo 315010, China
| | - Donghao Hu
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Kailei Xu
- Department of Plastic and Reconstructive Surgery, The First Affiliated Hospital of Ningbo University, Ningbo 315010, China; Center for Medical and Engineering Innovation, Central Laboratory, The First Affiliated Hospital of Ningbo University, Ningbo 315010, China; Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang Province, Ningbo 315010, China.
| | - Peng Wei
- Department of Plastic and Reconstructive Surgery, The First Affiliated Hospital of Ningbo University, Ningbo 315010, China.
| | - Jun Yin
- The State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou 310027, China; Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, School of Mechanical Engineering, Zhejiang University, Hangzhou 310027, China.
| |
Collapse
|
8
|
Zhou X, Tang A, Xiong C, Zhang G, Huang L, Xu F. Oriented Graphene Oxide Scaffold Promotes Nerve Regeneration in vitro and in vivo. Int J Nanomedicine 2024; 19:2573-2589. [PMID: 38505172 PMCID: PMC10949378 DOI: 10.2147/ijn.s439656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 02/14/2024] [Indexed: 03/21/2024] Open
Abstract
Background Treating peripheral nerve injuries (PNI) with defects remains challenging in clinical practice. The commercial conduits have shown suboptimal nerve regeneration and functional recovery due to their basic tubular design without electroactive and oriented topographical cues. Purpose To develop a new scaffold with oriented microstructure and electroactive Graphene oxide (GO) and investigate its' therapeutic effect on nerve regeneration in vitro and in vivo. Methods This study employed a straightforward approach to co-spin PCL and GO, yielding an oriented hybrid nanofibrous scaffold known as the O-GO/PCL scaffold. The physical and chemical properties of nanofibrous scaffold were tested by scanning electron microscopy (SEM), transmission electron microscope (TEM), tensile test and so on. Primary Schwann cells (SCs) and dorsal root ganglia (DRG) were used to investigate the impact of the newly developed scaffolds on the biological behavior of neural cells in vitro. Transcriptome sequencing (mRNA-seq) was employed to probe the underlying mechanisms of the synergistic effect of electroactive GO and longitudinal topographic guidance on nerve regeneration. Furthermore, the developed O-GO/PCL scaffold was utilized to bridge a 10-mm sciatic nerve defect in rat, aiming to investigate its therapeutic potential for peripheral nerve regeneration in vivo. Results and discussion The SEM and TEM revealed that the newly developed O-GO/PCL scaffold showed longitudinally oriented microstructure and GO particles were homogenously and uniformly distributed inside the nanofibers. Primary SCs were utilized to assess the biocompatibility of the GO-based scaffold, revealing that negligible cytotoxicity when GO concentration does not exceed 0.5%. In vitro analysis of nerve regeneration demonstrated that axons in the O-GO/PCL group exhibited an average length of 1054.88 ± 161.32 µm, significant longer than those in the other groups (P < 0.05). Moreover, mRNA sequencing results suggested that the O-GO/PCL scaffold could enhance nerve regeneration by upregulating genes associated with neural regeneration, encompassing ion transport, axon guidance and cell-cell interactions. Most importantly, we employed the O-GO/PCL scaffold to repair a 10-mm sciatic nerve defect in rat, resulting in augmented nerve regeneration, myelination, and functional recovery. Conclusion The O-GO/PCL scaffold with oriented microstructure and electroactive GO represents a promising heral nerve reconstruction.
Collapse
Affiliation(s)
- Xu Zhou
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, People’s Republic of China
- Department of Orthopaedics, General Hospital of Central Theater Command, Wuhan, 430070, People’s Republic of China
| | - Aolin Tang
- Department of Orthopaedics, General Hospital of Central Theater Command, Wuhan, 430070, People’s Republic of China
- Department of Orthopaedics, Minda Hospital of Hubei Minzu University, Enshi, 445000, People’s Republic of China
| | - Chengjie Xiong
- Department of Orthopaedics, General Hospital of Central Theater Command, Wuhan, 430070, People’s Republic of China
| | - Guoquan Zhang
- Department of Orthopaedics, General Hospital of Central Theater Command, Wuhan, 430070, People’s Republic of China
| | - Liangliang Huang
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, People’s Republic of China
- Department of Orthopaedics, General Hospital of Central Theater Command, Wuhan, 430070, People’s Republic of China
| | - Feng Xu
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, People’s Republic of China
- Department of Orthopaedics, General Hospital of Central Theater Command, Wuhan, 430070, People’s Republic of China
| |
Collapse
|
9
|
Wu J, Yun Z, Song W, Yu T, Xue W, Liu Q, Sun X. Highly oriented hydrogels for tissue regeneration: design strategies, cellular mechanisms, and biomedical applications. Theranostics 2024; 14:1982-2035. [PMID: 38505623 PMCID: PMC10945336 DOI: 10.7150/thno.89493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 01/19/2024] [Indexed: 03/21/2024] Open
Abstract
Many human tissues exhibit a highly oriented architecture that confers them with distinct mechanical properties, enabling adaptation to diverse and challenging environments. Hydrogels, with their water-rich "soft and wet" structure, have emerged as promising biomimetic materials in tissue engineering for repairing and replacing damaged tissues and organs. Highly oriented hydrogels can especially emulate the structural orientation found in human tissue, exhibiting unique physiological functions and properties absent in traditional homogeneous isotropic hydrogels. The design and preparation of highly oriented hydrogels involve strategies like including hydrogels with highly oriented nanofillers, polymer-chain networks, void channels, and microfabricated structures. Understanding the specific mechanism of action of how these highly oriented hydrogels affect cell behavior and their biological applications for repairing highly oriented tissues such as the cornea, skin, skeletal muscle, tendon, ligament, cartilage, bone, blood vessels, heart, etc., requires further exploration and generalization. Therefore, this review aims to fill that gap by focusing on the design strategy of highly oriented hydrogels and their application in the field of tissue engineering. Furthermore, we provide a detailed discussion on the application of highly oriented hydrogels in various tissues and organs and the mechanisms through which highly oriented structures influence cell behavior.
Collapse
Affiliation(s)
- Jiuping Wu
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
- Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Zhihe Yun
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun 130041, China
| | - Wenlong Song
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130023, China
| | - Tao Yu
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun 130041, China
| | - Wu Xue
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun 130041, China
| | - Qinyi Liu
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun 130041, China
| | - Xinzhi Sun
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| |
Collapse
|
10
|
Wan T, Zhang FS, Qin MY, Jiang HR, Zhang M, Qu Y, Wang YL, Zhang PX. Growth factors: Bioactive macromolecular drugs for peripheral nerve injury treatment - Molecular mechanisms and delivery platforms. Biomed Pharmacother 2024; 170:116024. [PMID: 38113623 DOI: 10.1016/j.biopha.2023.116024] [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: 10/31/2023] [Revised: 12/05/2023] [Accepted: 12/14/2023] [Indexed: 12/21/2023] Open
Abstract
Bioactive macromolecular drugs known as Growth Factors (GFs), approved by the Food and Drug Administration (FDA), have found successful application in clinical practice. They hold significant promise for addressing peripheral nerve injuries (PNIs). Peripheral nerve guidance conduits (NGCs) loaded with GFs, in the context of tissue engineering, can ensure sustained and efficient release of these bioactive compounds. This, in turn, maintains a stable, long-term, and effective GF concentration essential for treating damaged peripheral nerves. Peripheral nerve regeneration is a complex process that entails the secretion of various GFs. Following PNI, GFs play a pivotal role in promoting nerve cell growth and survival, axon and myelin sheath regeneration, cell differentiation, and angiogenesis. They also regulate the regenerative microenvironment, stimulate plasticity changes post-nerve injury, and, consequently, expedite nerve structure and function repair. Both exogenous and endogenous GFs, including NGF, BDNF, NT-3, GDNF, IGF-1, bFGF, and VEGF, have been successfully loaded onto NGCs using techniques like physical adsorption, blend doping, chemical covalent binding, and engineered transfection. These approaches have effectively promoted the repair of peripheral nerves. Numerous studies have demonstrated similar tissue functional therapeutic outcomes compared to autologous nerve transplantation. This evidence underscores the substantial clinical application potential of GFs in the domain of peripheral nerve repair. In this article, we provide an overview of GFs in the context of peripheral nerve regeneration and drug delivery systems utilizing NGCs. Looking ahead, commercial materials for peripheral nerve repair hold the potential to facilitate the effective regeneration of damaged peripheral nerves and maintain the functionality of distant target organs through the sustained release of GFs.
Collapse
Affiliation(s)
- Teng Wan
- Department of Orthopedics and Trauma, Peking University People's Hospital, Beijing 100044, China; Key Laboratory of Trauma and Neural Regeneration, Peking University, Beijing 100044, China; National Centre for Trauma Medicine, Beijing 100044, China
| | - Feng-Shi Zhang
- Department of Orthopedics and Trauma, Peking University People's Hospital, Beijing 100044, China; Key Laboratory of Trauma and Neural Regeneration, Peking University, Beijing 100044, China; National Centre for Trauma Medicine, Beijing 100044, China
| | - Ming-Yu Qin
- Suzhou Medical College, Soochow University, Suzhou 215026, China
| | - Hao-Ran Jiang
- Department of Orthopedics and Trauma, Peking University People's Hospital, Beijing 100044, China; Key Laboratory of Trauma and Neural Regeneration, Peking University, Beijing 100044, China; National Centre for Trauma Medicine, Beijing 100044, China
| | - Meng Zhang
- Department of Orthopedics and Trauma, Peking University People's Hospital, Beijing 100044, China; Key Laboratory of Trauma and Neural Regeneration, Peking University, Beijing 100044, China; National Centre for Trauma Medicine, Beijing 100044, China
| | - Yang Qu
- Department of Orthopedics and Trauma, Peking University People's Hospital, Beijing 100044, China; Key Laboratory of Trauma and Neural Regeneration, Peking University, Beijing 100044, China; National Centre for Trauma Medicine, Beijing 100044, China
| | - Yi-Lin Wang
- Department of Orthopedics and Trauma, Peking University People's Hospital, Beijing 100044, China; Key Laboratory of Trauma and Neural Regeneration, Peking University, Beijing 100044, China; National Centre for Trauma Medicine, Beijing 100044, China.
| | - Pei-Xun Zhang
- Department of Orthopedics and Trauma, Peking University People's Hospital, Beijing 100044, China; Key Laboratory of Trauma and Neural Regeneration, Peking University, Beijing 100044, China; National Centre for Trauma Medicine, Beijing 100044, China; Peking University People's Hospital Qingdao Hospital, Qingdao 266000, China.
| |
Collapse
|
11
|
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: 1] [Impact Index Per Article: 0.5] [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
|
12
|
Tan RES, Jeyaratnam S, Lim AYT. Updates in peripheral nerve surgery of the upper extremity: diagnosis and treatment options. ANNALS OF TRANSLATIONAL MEDICINE 2023; 11:391. [PMID: 37970602 PMCID: PMC10632574 DOI: 10.21037/atm-23-1500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Accepted: 08/11/2023] [Indexed: 11/17/2023]
Abstract
The loss of function resulting from peripheral nerve injuries confers a significant burden to the patient and society. The treatment of peripheral nerve injuries requires an accurate diagnosis and formulation of a functional reconstructive plan. Advances in peripheral nerve imaging complement electrodiagnostic studies, and provide us with detailed information regarding the status of nerve injury, repair, and regeneration in order to prognosticate recovery and determine the need for surgical intervention. When direct nerve repair is not possible, the methods for bridging a nerve gap are the nerve autograft, allograft and conduit. While current research supports the use of conduits and nerve allografts for shorter nerve gaps, the nerve autograft still remains the gold standard for bridging a nerve gap. When direct nerve repair or nerve grafting fails, or is anticipated to be insufficient, nerve transfers are an alternative for reconstruction. Knowledge of axonal counts, upper limb innervation patterns, location and clustering of upper limb peripheral nerves allows for the design of new nerve transfers. The options of nerve transfers for radial, ulnar and median nerve injuries are outlined, as well as their outcomes. Nerve transfers are an attractive option for restoring motor and sensory function while minimizing donor site morbidity. However, one must consider their limitations, and preserve donor sites for secondary tendon transfer options. This article presents the latest information regarding the imaging of peripheral nerves, methods to bridge a nerve gap, and nerve transfers to aid the peripheral nerve surgeon in choosing a reconstructive plan.
Collapse
Affiliation(s)
| | | | - Aymeric Y. T. Lim
- Department of Hand and Reconstructive Microsurgery, National University Health System, Singapore, Singapore
| |
Collapse
|
13
|
Mankavi F, Ibrahim R, Wang H. Advances in Biomimetic Nerve Guidance Conduits for Peripheral Nerve Regeneration. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:2528. [PMID: 37764557 PMCID: PMC10536071 DOI: 10.3390/nano13182528] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 09/04/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023]
Abstract
Injuries to the peripheral nervous system are a common clinical issue, causing dysfunctions of the motor and sensory systems. Surgical interventions such as nerve autografting are necessary to repair damaged nerves. Even with autografting, i.e., the gold standard, malfunctioning and mismatches between the injured and donor nerves often lead to unwanted failure. Thus, there is an urgent need for a new intervention in clinical practice to achieve full functional recovery. Nerve guidance conduits (NGCs), providing physicochemical cues to guide neural regeneration, have great potential for the clinical regeneration of peripheral nerves. Typically, NGCs are tubular structures with various configurations to create a microenvironment that induces the oriented and accelerated growth of axons and promotes neuron cell migration and tissue maturation within the injured tissue. Once the native neural environment is better understood, ideal NGCs should maximally recapitulate those key physiological attributes for better neural regeneration. Indeed, NGC design has evolved from solely physical guidance to biochemical stimulation. NGC fabrication requires fundamental considerations of distinct nerve structures, the associated extracellular compositions (extracellular matrices, growth factors, and cytokines), cellular components, and advanced fabrication technologies that can mimic the structure and morphology of native extracellular matrices. Thus, this review mainly summarizes the recent advances in the state-of-the-art NGCs in terms of biomaterial innovations, structural design, and advanced fabrication technologies and provides an in-depth discussion of cellular responses (adhesion, spreading, and alignment) to such biomimetic cues for neural regeneration and repair.
Collapse
Affiliation(s)
| | | | - Hongjun Wang
- Department of Biomedical Engineering, Semcer Center for Healthcare Innovation, Stevens Institute of Technology, Hoboken, NJ 07030, USA; (F.M.); (R.I.)
| |
Collapse
|
14
|
Monfette V, Choinière W, Godbout-Lavoie C, Pelletier S, Langelier È, Lauzon MA. Thermoelectric Freeze-Casting of Biopolymer Blends: Fabrication and Characterization of Large-Size Scaffolds for Nerve Tissue Engineering Applications. J Funct Biomater 2023; 14:330. [PMID: 37367294 DOI: 10.3390/jfb14060330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 06/03/2023] [Accepted: 06/16/2023] [Indexed: 06/28/2023] Open
Abstract
Peripheral nerve injuries (PNIs) are detrimental to the quality of life of affected individuals. Patients are often left with life-long ailments that affect them physically and psychologically. Autologous nerve transplant is still the gold standard treatment for PNIs despite limited donor site and partial recovery of nerve functions. Nerve guidance conduits are used as a nerve graft substitute and are efficient for the repair of small nerve gaps but require further improvement for repairs exceeding 30 mm. Freeze-casting is an interesting fabrication method for the conception of scaffolds meant for nerve tissue engineering since the microstructure obtained comprises highly aligned micro-channels. The present work focuses on the fabrication and characterization of large scaffolds (35 mm length, 5 mm diameter) made of collagen/chitosan blends by freeze-casting via thermoelectric effect instead of traditional freezing solvents. As a freeze-casting microstructure reference, scaffolds made from pure collagen were used for comparison. Scaffolds were covalently crosslinked for better performance under load and laminins were further added to enhance cell interactions. Microstructural features of lamellar pores display an average aspect ratio of 0.67 ± 0.2 for all compositions. Longitudinally aligned micro-channels are reported as well as enhanced mechanical properties in traction under physiological-like conditions (37 °C, pH = 7.4) resulting from crosslinking treatment. Cell viability assays using a rat Schwann cell line derived from sciatic nerve (S16) indicate that scaffold cytocompatibility is similar between scaffolds made from collagen only and scaffolds made from collagen/chitosan blend with high collagen content. These results confirm that freeze-casting via thermoelectric effect is a reliable manufacturing strategy for the fabrication of biopolymer scaffolds for future peripheral nerve repair applications.
Collapse
Affiliation(s)
- Vincent Monfette
- Department of Chemical Engineering and Biotechnological of Engineering, Faculty of Engineering, Université de Sherbrooke, Sherbrooke, QC J1K 2R1, Canada
| | - William Choinière
- Department of Chemical Engineering and Biotechnological of Engineering, Faculty of Engineering, Université de Sherbrooke, Sherbrooke, QC J1K 2R1, Canada
| | - Catherine Godbout-Lavoie
- Department of Chemical Engineering and Biotechnological of Engineering, Faculty of Engineering, Université de Sherbrooke, Sherbrooke, QC J1K 2R1, Canada
| | - Samuel Pelletier
- Department of Electrical Engineering and Informatics Engineering, Faculty of Engineering, Université de Sherbrooke, Sherbrooke, QC J1K 2R1, Canada
| | - Ève Langelier
- Department of Mechanical Engineering, Faculty of Engineering, Université de Sherbrooke, Sherbrooke, QC J1K 2R1, Canada
| | - Marc-Antoine Lauzon
- Department of Chemical Engineering and Biotechnological of Engineering, Faculty of Engineering, Université de Sherbrooke, Sherbrooke, QC J1K 2R1, Canada
- Research Center on Aging, CIUSSS de l'ESTRIE-CHUS, Sherbrooke, QC J1H 4C4, Canada
| |
Collapse
|
15
|
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: 11] [Impact Index Per Article: 5.5] [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
|
16
|
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: 10] [Impact Index Per Article: 5.0] [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
|
17
|
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
|
18
|
Lin Y, Yu J, Zhang Y, Hayat U, Liu C, Huang X, Lin H, Wang JY. 4D printed tri-segment nerve conduit using zein gel as the ink for repair of rat sciatic nerve large defect. BIOMATERIALS ADVANCES 2023; 151:213473. [PMID: 37245344 DOI: 10.1016/j.bioadv.2023.213473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 04/20/2023] [Accepted: 05/10/2023] [Indexed: 05/30/2023]
Abstract
Zein has enormous potential for application in biomedical field due to biodegradation and biocompatibility, we have recently prepared zein gel as a possible 3D printing ink. Our previous studies found that the pore structure in zein material can reduce early inflammation, promote the polarization of macrophages toward the M2 phenotype, and accelerate nerve regeneration. To further explore the role of zein in nerve repair, we used 4D printing technique to create nerve conduits with zein protein gel, and designed 2 types of tri-segment conduits with different degradation rates. Structural parts printed in support baths with higher water content show faster degradation rates than those printed in support baths with lower water content. The conduits that degraded quickly at both ends and slowly in the middle (CB75-CB40-CB75) and the conduits that degraded slowly at both ends and quickly in the middle (CB40-CB75-CB40) were 4D printed, respectively. Animal experiments suggest that the CB75-CB40-CB75 conduit is better for nerve repair, which may be because its degradation pattern can match to the pattern of nerve regeneration better. Our new strategy through 4D printing indicated that fine modulation in conduit degradation can affect efficacy of nerve repair significantly.
Collapse
Affiliation(s)
- Yaofa Lin
- Department of Orthopaedics, Jiading Branch of Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 800 Huangjiahuayuan Road, Shanghai 201803, China
| | - Jinwen Yu
- School of Biomedical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Yubei Zhang
- School of Biomedical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Uzma Hayat
- Jiaxing Yaojiao Medical Device Co. Ltd., 321 Jiachuang Road, Jiaxing 314032, China
| | - Chang Liu
- School of Biomedical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Xiaoyun Huang
- Department of Hand Surgery, Huashan Hospital, Fudan University, 12 Urumqi Middle Road, Shanghai 200040, China
| | - Haodong Lin
- Department of Orthopaedics, Jiading Branch of Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 800 Huangjiahuayuan Road, Shanghai 201803, China; Department of Orthopaedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 85 Wujin Road, Shanghai 200080, China.
| | - Jin-Ye Wang
- School of Biomedical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China; Jiaxing Yaojiao Medical Device Co. Ltd., 321 Jiachuang Road, Jiaxing 314032, China.
| |
Collapse
|
19
|
Caragher SP, Khouri KS, Raasveld FV, Winograd JM, Valerio IL, Gfrerer L, Eberlin KR. The Peripheral Nerve Surgeon's Role in the Management of Neuropathic Pain. PLASTIC AND RECONSTRUCTIVE SURGERY-GLOBAL OPEN 2023; 11:e5005. [PMID: 37360238 PMCID: PMC10287132 DOI: 10.1097/gox.0000000000005005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 03/29/2023] [Indexed: 06/28/2023]
Abstract
Neuropathic pain (NP) underlies significant morbidity and disability worldwide. Although pharmacologic and functional therapies attempt to address this issue, they remain incompletely effective for many patients. Peripheral nerve surgeons have a range of techniques for intervening on NP. The aim of this review is to enable practitioners to identify patients with NP who might benefit from surgical intervention. The workup for NP includes patient history and specific physical examination maneuvers, as well as imaging and diagnostic nerve blocks. Once diagnosed, there is a range of options surgeons can utilize based on specific causes of NP. These techniques include nerve decompression, nerve reconstruction, nerve ablative techniques, and implantable nerve-modulating devices. In addition, there is an emerging role for preoperative involvement of peripheral nerve surgeons for cases known to carry a high risk of inducing postoperative NP. Lastly, we describe the ongoing work that will enable surgeons to expand their armamentarium to better serve patients with NP.
Collapse
Affiliation(s)
| | - Kimberly S. Khouri
- Division of Plastic and Reconstructive Surgery, Massachusetts General Hosptial, Boston, Mass
| | - Floris V. Raasveld
- Division of Plastic and Reconstructive Surgery, Massachusetts General Hosptial, Boston, Mass
- Department of Plastic, Reconstructive and Hand Surgery, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Jonathan M. Winograd
- From the Harvard Medical School, Boston, Mass
- Division of Plastic and Reconstructive Surgery, Massachusetts General Hosptial, Boston, Mass
| | - Ian L. Valerio
- From the Harvard Medical School, Boston, Mass
- Division of Plastic and Reconstructive Surgery, Massachusetts General Hosptial, Boston, Mass
| | - Lisa Gfrerer
- Division of Plastic and Reconstructive Surgery, Weill Cornell Medicine, New York, N.Y
| | - Kyle R. Eberlin
- From the Harvard Medical School, Boston, Mass
- Division of Plastic and Reconstructive Surgery, Massachusetts General Hosptial, Boston, Mass
| |
Collapse
|
20
|
Himel MH, Sikder B, Ahmed T, Choudhury SM. Biomimicry in nanotechnology: a comprehensive review. NANOSCALE ADVANCES 2023; 5:596-614. [PMID: 36756510 PMCID: PMC9890514 DOI: 10.1039/d2na00571a] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 12/15/2022] [Indexed: 06/18/2023]
Abstract
Biomimicry has been utilized in many branches of science and engineering to develop devices for enhanced and better performance. The application of nanotechnology has made life easier in modern times. It has offered a way to manipulate matter and systems at the atomic level. As a result, the miniaturization of numerous devices has been possible. Of late, the integration of biomimicry with nanotechnology has shown promising results in the fields of medicine, robotics, sensors, photonics, etc. Biomimicry in nanotechnology has provided eco-friendly and green solutions to the energy problem and in textiles. This is a new research area that needs to be explored more thoroughly. This review illustrates the progress and innovations made in the field of nanotechnology with the integration of biomimicry.
Collapse
Affiliation(s)
- Mehedi Hasan Himel
- Department of Electrical and Electronic Engineering, Bangladesh University of Engineering and Technology Dhaka 1205 Bangladesh
- Department of Computer Science and Engineering, Brac University 66 Mohakhali Dhaka 1212 Bangladesh
| | - Bejoy Sikder
- Department of Electrical and Electronic Engineering, Bangladesh University of Engineering and Technology Dhaka 1205 Bangladesh
| | - Tanvir Ahmed
- Department of Electrical and Electronic Engineering, Bangladesh University of Engineering and Technology Dhaka 1205 Bangladesh
- Department of Computer Science and Engineering, Brac University 66 Mohakhali Dhaka 1212 Bangladesh
| | - Sajid Muhaimin Choudhury
- Department of Electrical and Electronic Engineering, Bangladesh University of Engineering and Technology Dhaka 1205 Bangladesh
| |
Collapse
|
21
|
Ke H, Yang H, Zhao Y, Li T, Xin D, Gai C, Jiang Z, Wang Z. 3D Gelatin Microsphere Scaffolds Promote Functional Recovery after Spinal Cord Hemisection in Rats. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2204528. [PMID: 36453595 PMCID: PMC9875663 DOI: 10.1002/advs.202204528] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 10/22/2022] [Indexed: 05/24/2023]
Abstract
Spinal cord injury (SCI) damages signal connections and conductions, with the result that neuronal circuits are disrupted leading to neural dysfunctions. Such injuries represent a serious and relatively common central nervous system condition and current treatments have limited success in the reconstruction of nerve connections in injured areas, especially where sizeable gaps are present. Biomaterial scaffolds have become an effective alternative to nerve transplantation in filling these gaps and provide the foundation for simulating the 3D structure of solid organs. However, there remain some limitations with the application of 3D bioprinting for preparation of biomaterial scaffolds. Here, the approach in constructing and testing mini-tissue building blocks and self-assembly, solid 3D gelatin microsphere (GM) scaffolds with multiple voids as based on the convenient preparation of gelatin microspheres by microfluidic devices is described. These 3D GM scaffolds demonstrate suitable biocompatibility, biodegradation, porosity, low preparation costs, and relative ease of production. Moreover, 3D GM scaffolds can effectively bridge injury gaps, establish nerve connections and signal transductions, mitigate inflammatory microenvironments, and reduce glial scar formation. Accordingly, these 3D GM scaffolds can serve as a novel and effective bridging method to promote nerve regeneration and reconstruction and thus recovery of nerve function after SCI.
Collapse
Affiliation(s)
- Hongfei Ke
- Department of PhysiologySchool of Basic Medical SciencesCheeloo College of MedicineShandong University44 Wenhua Xi RoadJinanShandong250012P. R. China
| | - Hongru Yang
- State Key Laboratory of Crystal MaterialsShandong University27 Shanda NanluJinanShandong250100P. R. China
| | - Yijing Zhao
- Department of PhysiologySchool of Basic Medical SciencesCheeloo College of MedicineShandong University44 Wenhua Xi RoadJinanShandong250012P. R. China
| | - Tingting Li
- Department of PhysiologySchool of Basic Medical SciencesCheeloo College of MedicineShandong University44 Wenhua Xi RoadJinanShandong250012P. R. China
| | - Danqing Xin
- Department of PhysiologySchool of Basic Medical SciencesCheeloo College of MedicineShandong University44 Wenhua Xi RoadJinanShandong250012P. R. China
| | - Chengcheng Gai
- Department of PhysiologySchool of Basic Medical SciencesCheeloo College of MedicineShandong University44 Wenhua Xi RoadJinanShandong250012P. R. China
| | - Zige Jiang
- Department of PhysiologySchool of Basic Medical SciencesCheeloo College of MedicineShandong University44 Wenhua Xi RoadJinanShandong250012P. R. China
| | - Zhen Wang
- Department of PhysiologySchool of Basic Medical SciencesCheeloo College of MedicineShandong University44 Wenhua Xi RoadJinanShandong250012P. R. China
| |
Collapse
|
22
|
Yan L, Entezari A, Zhang Z, Zhong J, Liang J, Li Q, Qi J. An experimental and numerical study of the microstructural and biomechanical properties of human peripheral nerve endoneurium for the design of tissue scaffolds. Front Bioeng Biotechnol 2022; 10:1029416. [PMID: 36545684 PMCID: PMC9762494 DOI: 10.3389/fbioe.2022.1029416] [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: 09/07/2022] [Accepted: 11/14/2022] [Indexed: 12/12/2022] Open
Abstract
Biomimetic design of scaffold architectures represents a promising strategy to enable the repair of tissue defects. Natural endoneurium extracellular matrix (eECM) exhibits a sophisticated microstructure and remarkable microenvironments conducive for guiding neurite regeneration. Therefore, the analysis of eECM is helpful to the design of bionic scaffold. Unfortunately, a fundamental lack of understanding of the microstructural characteristics and biomechanical properties of the human peripheral nerve eECM exists. In this study, we used microscopic computed tomography (micro-CT) to reconstruct a three-dimensional (3D) eECM model sourced from mixed nerves. The tensile strength and effective modulus of human fresh nerve fascicles were characterized experimentally. Permeability was calculated from a computational fluid dynamic (CFD) simulation of the 3D eECM model. Fluid flow of acellular nerve fascicles was tested experimentally to validate the permeability results obtained from CFD simulations. The key microstructural parameters, such as porosity is 35.5 ± 1.7%, tortuosity in endoneurium (X axis is 1.26 ± 0.028, Y axis is 1.26 ± 0.020 and Z axis is 1.17 ± 0.03, respectively), tortuosity in pore (X axis is 1.50 ± 0.09, Y axis is 1.44 ± 0.06 and Z axis is 1.13 ± 0.04, respectively), surface area-to-volume ratio (SAVR) is 0.165 ± 0.007 μm-1 and pore size is 11.8 ± 2.8 μm, respectively. These were characterized from the 3D eECM model and may exert different effects on the stiffness and permeability. The 3D microstructure of natural peripheral nerve eECM exhibits relatively lower permeability (3.10 m2 × 10-12 m2) than other soft tissues. These key microstructural and biomechanical parameters may play an important role in the design and fabrication of intraluminal guidance scaffolds to replace natural eECM. Our findings can aid the development of regenerative therapies and help improve scaffold design.
Collapse
Affiliation(s)
- Liwei Yan
- Department of Microsurgery, Trauma and Hand Surgery, The First Affiliated Hospital of Sun Yat‐sen University, Guangzhou, China
| | - Ali Entezari
- School of Biomedical Engineering, University of Technology Sydney, Ultimo, NSW, Australia,School of Aerospace, Mechanical and Mechatronic Engineering, University of Sydney, Sydney, NSW, Australia
| | - Zhongpu Zhang
- School of Computing, Engineering and Mathematics, Western Sydney University, Penrith, NSW, Australia
| | - Jingxiao Zhong
- School of Aerospace, Mechanical and Mechatronic Engineering, University of Sydney, Sydney, NSW, Australia
| | - Jing Liang
- Department of Microsurgery, Trauma and Hand Surgery, The First Affiliated Hospital of Sun Yat‐sen University, Guangzhou, China
| | - Qing Li
- School of Aerospace, Mechanical and Mechatronic Engineering, University of Sydney, Sydney, NSW, Australia,*Correspondence: Jian Qi, ; Qing Li,
| | - Jian Qi
- Department of Microsurgery, Trauma and Hand Surgery, The First Affiliated Hospital of Sun Yat‐sen University, Guangzhou, China,Guangdong Provincial Key Laboratory for Orthopedics and Traumatology, Guangzhou, China,*Correspondence: Jian Qi, ; Qing Li,
| |
Collapse
|
23
|
Shen Y, Cao X, Lu M, Gu H, Li M, Posner DA. Current treatments after spinal cord injury: Cell engineering, tissue engineering, and combined therapies. SMART MEDICINE 2022; 1:e20220017. [PMID: 39188731 PMCID: PMC11235943 DOI: 10.1002/smmd.20220017] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 10/20/2022] [Indexed: 08/28/2024]
Abstract
Both traumatic and non-traumatic spinal cord injuries (SCIs) can be categorized as damages done to our central nervous system (CNS). The patients' physical and mental health may suffer greatly because of traumatic SCI. With the widespread use of motor vehicles and increasingly aged population, the occurrence of SCI is more frequent than before, creating a considerable burden to global public health. The regeneration process of the spinal cord is hampered by a series of events that occur following SCI like edema, hemorrhage, formation of cystic cavities, and ischemia. An effective strategy for the treatment of SCI and functional recovery still has not been discovered; however, recent advances have been made in bioengineering fields that therapies based on cells, biomaterials, and biomolecules have proved effective in the repair of the spinal cord. In the light of worldwide importance of treatments for SCI, this article aims to provide a review of recent advances by first introducing the physiology, etiology, epidemiology, and mechanisms of SCI. We then put emphasis on the widely used clinical treatments and bioengineering strategies (cell-based, biomaterial-based, and biomolecule-based) for the functional regeneration of the spinal cord as well as challenges faced by scientists currently. This article provides scientists and clinicians with a comprehensive outlook on the recent advances of preclinical and clinical treatments of SCI, hoping to help them find keys to the functional regeneration of SCI.
Collapse
Affiliation(s)
- Yingbo Shen
- State Key Laboratory of BioelectronicsSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjingChina
| | - Xinyue Cao
- State Key Laboratory of BioelectronicsSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjingChina
| | - Minhui Lu
- State Key Laboratory of BioelectronicsSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjingChina
| | - Hongcheng Gu
- State Key Laboratory of BioelectronicsSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjingChina
| | - Minli Li
- State Key Laboratory of BioelectronicsSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjingChina
| | - David A. Posner
- Molecular Immunity UnitCambridge Institute of Therapeutic Immunology and Infectious DiseasesDepartment of MedicineUniversity of CambridgeCambridgeUK
| |
Collapse
|
24
|
Sah MK, Mukherjee S, Flora B, Malek N, Rath SN. Advancement in "Garbage In Biomaterials Out (GIBO)" concept to develop biomaterials from agricultural waste for tissue engineering and biomedical applications. JOURNAL OF ENVIRONMENTAL HEALTH SCIENCE & ENGINEERING 2022; 20:1015-1033. [PMID: 36406592 PMCID: PMC9672289 DOI: 10.1007/s40201-022-00815-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 06/27/2022] [Indexed: 06/16/2023]
Abstract
Presently on a global scale, one of the major concerns is to find effective strategies to manage the agricultural waste to protect the environment. One strategy that has been drawing attention among the researchers is the development of biocompatible materials from agricultural waste. This strategy implies successful conversion of agricultural waste products (e.g.: cellulose, eggshell etc.) into building blocks for biomaterial development. Some of these wastes contain even bioactive compounds having biomedical applications. The replacement and augmentation of human tissue with biomaterials as alternative to traditional method not only bypasses immune-rejection, donor scarcity, and maintenance; but also provides long term solution to damaged or malfunctioning organs. Biomaterials development as one of the key challenges in tissue engineering approach, resourced from natural origin imparts better biocompatibility due to closely mimicking composition with cellular microenvironment. The "Garbage In, Biomaterials Out (GIBO)" concept, not only recycles the agricultural wastes, but also adds to biomaterial raw products for further product development in tissue regeneration. This paper reviews the conversion of garbage agricultural by-products to the biocompatible materials for various biomedical applications. Graphical abstract The agro-waste biomass processed, purified, modified, and further utilized for the fabrication of biomaterials-based support system for tissue engineering applications to grow living body parts in vitro or in vivo.
Collapse
Affiliation(s)
- Mahesh Kumar Sah
- Department of Biotechnology, Dr. B. R. Ambedkar, National Institute of Technology, Jalandhar, Punjab 144011 India
| | - Sunny Mukherjee
- Department of Biotechnology, Dr. B. R. Ambedkar, National Institute of Technology, Jalandhar, Punjab 144011 India
| | - Bableen Flora
- Department of Biotechnology, Lovely Professional University, Jalandhar, Punjab India
| | - Naved Malek
- Department of Chemistry, S. V. National Institute of Technology, Surat, Gujarat India
| | - Subha Narayan Rath
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Medak, Telangana India
| |
Collapse
|
25
|
Lecoq FA, Barnouin L, Ardouin L, Hartmann D, Obert L. Inverted human umbilical artery as a 3D scaffold for sciatic nerve regeneration in rats. Cell Tissue Bank 2022; 23:909-922. [PMID: 35503142 PMCID: PMC9675695 DOI: 10.1007/s10561-022-10006-8] [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: 10/29/2021] [Accepted: 04/03/2022] [Indexed: 11/02/2022]
Abstract
Treatment of peripheral nerve injuries (PNIs) remains a challenge. Interposing a graft delivers better regenerative outcomes. Autografts present major drawbacks which have given rise to the development of alternatives such as artificial scaffolds, some of which are very promising. This study was designed to investigate the potential use of an inverted human umbilical cord artery (iHUA) as a 3D scaffold nerve chamber, for nerve regeneration after transection of the sciatic nerve (SN) in rats. Rats underwent surgical SN transection in their right hindlimb, followed by suture of the device at the resected stumps. Local tolerance, insert biodegradability and nerve reconstruction over time were thoroughly studied by histopathological and morphometric analysis, completed by functional test assessment of sensitivity and motricity recovery. We have demonstrated that nerve reconstruction in the presence of an iHUA insert is effective. The device is well tolerated and highly biodegraded. Although the regenerated nerve is still immature at the end of our study, signs of sensitivity and partial functional recovery were witnessed, confirming our histological findings. Our results support the potential clinical use of iHUA as a 3D scaffold to bridge nerve discontinuity and guide axonal regrowth in selected cases of PNIs.
Collapse
Affiliation(s)
- Flore-Anne Lecoq
- Institut de la Main Nantes Atlantique, Elsan Santé Atlantique, Saint Herblain, France
| | | | - Ludovic Ardouin
- Institut de la Main Nantes Atlantique, Elsan Santé Atlantique, Saint Herblain, France
| | | | | |
Collapse
|
26
|
Multichannel nerve conduit based on chitosan derivates for peripheral nerve regeneration and Schwann cell survival. Carbohydr Polym 2022; 301:120327. [DOI: 10.1016/j.carbpol.2022.120327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Revised: 11/05/2022] [Accepted: 11/07/2022] [Indexed: 11/13/2022]
|
27
|
Mariano A, Bovio CL, Criscuolo V, Santoro F. Bioinspired micro- and nano-structured neural interfaces. NANOTECHNOLOGY 2022; 33:492501. [PMID: 35947922 DOI: 10.1088/1361-6528/ac8881] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 08/10/2022] [Indexed: 06/15/2023]
Abstract
The development of a functional nervous system requires neurons to interact with and promptly respond to a wealth of biochemical, mechanical and topographical cues found in the neural extracellular matrix (ECM). Among these, ECM topographical cues have been found to strongly influence neuronal function and behavior. Here, we discuss how the blueprint of the architectural organization of the brain ECM has been tremendously useful as a source of inspiration to design biomimetic substrates to enhance neural interfaces and dictate neuronal behavior at the cell-material interface. In particular, we focus on different strategies to recapitulate cell-ECM and cell-cell interactions. In order to mimic cell-ECM interactions, we introduce roughness as a first approach to provide informative topographical biomimetic cues to neurons. We then examine 3D scaffolds and hydrogels, as softer 3D platforms for neural interfaces. Moreover, we will discuss how anisotropic features such as grooves and fibers, recapitulating both ECM fibrils and axonal tracts, may provide recognizable paths and tracks that neuron can follow as they develop and establish functional connections. Finally, we show how isotropic topographical cues, recapitulating shapes, and geometries of filopodia- and mushroom-like dendritic spines, have been instrumental to better reproduce neuron-neuron interactions for applications in bioelectronics and neural repair strategies. The high complexity of the brain architecture makes the quest for the fabrication of create more biologically relevant biomimetic architectures in continuous and fast development. Here, we discuss how recent advancements in two-photon polymerization and remotely reconfigurable dynamic interfaces are paving the way towards to a new class of smart biointerfaces forin vitroapplications spanning from neural tissue engineering as well as neural repair strategies.
Collapse
Affiliation(s)
- Anna Mariano
- Tissue Electronics, Istituto Italiano di Tecnologia, I-80125 Naples, Italy
| | - Claudia Latte Bovio
- Tissue Electronics, Istituto Italiano di Tecnologia, I-80125 Naples, Italy
- Dipartimento di Chimica, Materiali e Produzione Industriale, Università di Napoli Federico II, I-80125, Naples, Italy
| | - Valeria Criscuolo
- Faculty of Electrical Engineering and IT, RWTH Aachen, D-52074, Germany
| | - Francesca Santoro
- Tissue Electronics, Istituto Italiano di Tecnologia, I-80125 Naples, Italy
- Faculty of Electrical Engineering and IT, RWTH Aachen, D-52074, Germany
- Institute for Biological Information Processing-Bioelectronics, Forschungszentrum Juelich, D-52428, Germany
| |
Collapse
|
28
|
Veletić M, Apu EH, Simić M, Bergsland J, Balasingham I, Contag CH, Ashammakhi N. Implants with Sensing Capabilities. Chem Rev 2022; 122:16329-16363. [PMID: 35981266 DOI: 10.1021/acs.chemrev.2c00005] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Because of the aging human population and increased numbers of surgical procedures being performed, there is a growing number of biomedical devices being implanted each year. Although the benefits of implants are significant, there are risks to having foreign materials in the body that may lead to complications that may remain undetectable until a time at which the damage done becomes irreversible. To address this challenge, advances in implantable sensors may enable early detection of even minor changes in the implants or the surrounding tissues and provide early cues for intervention. Therefore, integrating sensors with implants will enable real-time monitoring and lead to improvements in implant function. Sensor integration has been mostly applied to cardiovascular, neural, and orthopedic implants, and advances in combined implant-sensor devices have been significant, yet there are needs still to be addressed. Sensor-integrating implants are still in their infancy; however, some have already made it to the clinic. With an interdisciplinary approach, these sensor-integrating devices will become more efficient, providing clear paths to clinical translation in the future.
Collapse
Affiliation(s)
- Mladen Veletić
- Department of Electronic Systems, Norwegian University of Science and Technology, 7491 Trondheim, Norway.,The Intervention Centre, Technology and Innovation Clinic, Oslo University Hospital, 0372 Oslo, Norway
| | - Ehsanul Hoque Apu
- Institute for Quantitative Health Science and Engineering (IQ) and Department of Biomedical Engineering (BME), Michigan State University, East Lansing, Michigan 48824, United States.,Division of Hematology and Oncology, Department of Internal Medicine, Michigan Medicine, University of Michigan, Ann Arbor, Michigan 48105, United States
| | - Mitar Simić
- Faculty of Electrical Engineering, University of Banja Luka, 78000 Banja Luka, Bosnia and Herzegovina
| | - Jacob Bergsland
- The Intervention Centre, Technology and Innovation Clinic, Oslo University Hospital, 0372 Oslo, Norway
| | - Ilangko Balasingham
- Department of Electronic Systems, Norwegian University of Science and Technology, 7491 Trondheim, Norway.,The Intervention Centre, Technology and Innovation Clinic, Oslo University Hospital, 0372 Oslo, Norway
| | - Christopher H Contag
- Institute for Quantitative Health Science and Engineering (IQ) and Department of Biomedical Engineering (BME), Michigan State University, East Lansing, Michigan 48824, United States
| | - Nureddin Ashammakhi
- Institute for Quantitative Health Science and Engineering (IQ) and Department of Biomedical Engineering (BME), Michigan State University, East Lansing, Michigan 48824, United States.,Department of Bioengineering, University of California, Los Angeles, California 90095, United States
| |
Collapse
|
29
|
Idrisova KF, Zeinalova AK, Masgutova GA, Bogov AA, Allegrucci C, Syromiatnikova VY, Salafutdinov II, Garanina EE, Andreeva DI, Kadyrov AA, Rizvanov AA, Masgutov RF. Application of neurotrophic and proangiogenic factors as therapy after peripheral nervous system injury. Neural Regen Res 2022; 17:1240-1247. [PMID: 34782557 PMCID: PMC8643040 DOI: 10.4103/1673-5374.327329] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 10/14/2020] [Accepted: 06/04/2021] [Indexed: 11/24/2022] Open
Abstract
The intrinsic ability of peripheral nerves to regenerate after injury is extremely limited, especially in case of severe injury. This often leads to poor motor function and permanent disability. Existing approaches for the treatment of injured nerves do not provide appropriate conditions to support survival and growth of nerve cells. This drawback can be compensated by the use of gene therapy and cell therapy-based drugs that locally provide an increase in the key regulators of nerve growth, including neurotrophic factors and extracellular matrix proteins. Each growth factor plays its own specific angiotrophic or neurotrophic role. Currently, growth factors are widely studied as accelerators of nerve regeneration. Particularly noteworthy is synergy between various growth factors, that is essential for both angiogenesis and neurogenesis. Fibroblast growth factor 2 and vascular endothelial growth factor are widely known for their proangiogenic effects. At the same time, fibroblast growth factor 2 and vascular endothelial growth factor stimulate neural cell growth and play an important role in neurodegenerative diseases of the peripheral nervous system. Taken together, their neurotrophic and angiogenic properties have positive effect on the regeneration process. In this review we provide an in-depth overview of the role of fibroblast growth factor 2 and vascular endothelial growth factor in the regeneration of peripheral nerves, thus demonstrating their neurotherapeutic efficacy in improving neuron survival in the peripheral nervous system.
Collapse
Affiliation(s)
| | | | | | | | - Cinzia Allegrucci
- Biodiscovery Institute, School of Veterinary Medicine and Science, University of Nottingham, Nottingham, UK
| | | | | | | | | | | | | | - Ruslan Faridovich Masgutov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
- Republican Clinical Hospital, Kazan, Russia
| |
Collapse
|
30
|
Deng P, Chen F, Zhang H, Chen Y, Zhou J. Multifunctional Double-Layer Composite Hydrogel Conduit Based on Chitosan for Peripheral Nerve Repairing. Adv Healthc Mater 2022; 11:e2200115. [PMID: 35396930 DOI: 10.1002/adhm.202200115] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 03/31/2022] [Indexed: 12/21/2022]
Abstract
Peripheral nerve regeneration and functional recovery is a major challenge in clinical practice. Nerve conduit is an effective treatment for peripheral nerve repair, but the traditional hollow nerve conduit is not satisfactory in peripheral nerve repair due to the limitation of cell migration and nutrient transport. Herein, the double cross-linked hydrogels with injectable, self-healing, and conductive properties are synthesized by the Schiff base reaction between polyaniline-modified carboxymethyl chitosan and aldehyde-modified Pluronic F-127 (F127-CHO), and the hydrophobic interaction of F127-CHO. The conductive hydrogel is injected into the cavity of chitosan conduit prepared by electrodeposition. The inner conductive hydrogel and the outer chitosan conduit are formed into a whole through the Schiff base reaction to obtain a double-layer composite hydrogel nerve conduit. The double-layer composite hydrogel neural conduit loaded with 7,8-dihydroxyflavone (DHF) has excellent degradability, biocompatibility, antioxidant activity, and Schwann cell proliferation activity. In the rat sciatic nerve defect model, the double-layer composite hydrogel nerve conduit significantly promotes sciatic nerve regeneration compared with the chitosan hollow conduit. Surprisingly, the repair ability of double-layered hydrogel nerve conduit loaded with DHF is comparable to that of autologous transplantation. Therefore, this multifunctional double-layer composite hydrogel conduit has great potential for peripheral nerve repairing.
Collapse
Affiliation(s)
- Pengpeng Deng
- Hubei Engineering Center of Natural Polymers‐based Medical Materials Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry Wuhan University Wuhan 430072 China
- Department of Biomedical Engineering Hubei Province Key Laboratory of Allergy and Immune Related Diseases School of Basic Medical Science Wuhan University Wuhan 430071 China
| | - Feixiang Chen
- Glyn O. Philips Hydrocolloid Research Centre at HUT Hubei University of Technology Wuhan 430068 China
| | - Haodong Zhang
- Hubei Engineering Center of Natural Polymers‐based Medical Materials Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry Wuhan University Wuhan 430072 China
| | - Yun Chen
- Glyn O. Philips Hydrocolloid Research Centre at HUT Hubei University of Technology Wuhan 430068 China
| | - Jinping Zhou
- Hubei Engineering Center of Natural Polymers‐based Medical Materials Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry Wuhan University Wuhan 430072 China
| |
Collapse
|
31
|
Liu K, Yan L, Li R, Song Z, Ding J, Liu B, Chen X. 3D Printed Personalized Nerve Guide Conduits for Precision Repair of Peripheral Nerve Defects. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2103875. [PMID: 35182046 PMCID: PMC9036027 DOI: 10.1002/advs.202103875] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 12/25/2021] [Indexed: 05/07/2023]
Abstract
The treatment of peripheral nerve defects has always been one of the most challenging clinical practices in neurosurgery. Currently, nerve autograft is the preferred treatment modality for peripheral nerve defects, while the therapy is constantly plagued by the limited donor, loss of donor function, formation of neuroma, nerve distortion or dislocation, and nerve diameter mismatch. To address these clinical issues, the emerged nerve guide conduits (NGCs) are expected to offer effective platforms to repair peripheral nerve defects, especially those with large or complex topological structures. Up to now, numerous technologies are developed for preparing diverse NGCs, such as solvent casting, gas foaming, phase separation, freeze-drying, melt molding, electrospinning, and three-dimensional (3D) printing. 3D printing shows great potential and advantages because it can quickly and accurately manufacture the required NGCs from various natural and synthetic materials. This review introduces the application of personalized 3D printed NGCs for the precision repair of peripheral nerve defects and predicts their future directions.
Collapse
Affiliation(s)
- Kai Liu
- Department of Hand and Foot SurgeryThe First Hospital of Jilin University1 Xinmin StreetChangchun130061P. R. China
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of Sciences5625 Renmin StreetChangchun130022P. R. China
| | - Lesan Yan
- Biomedical Materials and Engineering Research Center of Hubei ProvinceState Key Laboratory of Advanced Technology for Materials Synthesis and ProcessingWuhan University of Technology122 Luoshi RoadWuhan430070P. R. China
| | - Ruotao Li
- Department of Hand and Foot SurgeryThe First Hospital of Jilin University1 Xinmin StreetChangchun130061P. R. China
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of Sciences5625 Renmin StreetChangchun130022P. R. China
| | - Zhiming Song
- Department of Sports MedicineThe First Hospital of Jilin University1 Xinmin StreetChangchun130061P. R. China
| | - Jianxun Ding
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of Sciences5625 Renmin StreetChangchun130022P. R. China
- State Key Laboratory of Molecular Engineering of PolymersFudan University220 Handan RoadShanghai200433P. R. China
| | - Bin Liu
- Department of Hand and Foot SurgeryThe First Hospital of Jilin University1 Xinmin StreetChangchun130061P. R. China
| | - Xuesi Chen
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of Sciences5625 Renmin StreetChangchun130022P. R. China
| |
Collapse
|
32
|
Bioactive injectable hydrogels for on demand molecule/cell delivery and for tissue regeneration in the central nervous system. Acta Biomater 2022; 140:88-101. [PMID: 34852302 DOI: 10.1016/j.actbio.2021.11.038] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 11/21/2021] [Accepted: 11/23/2021] [Indexed: 12/21/2022]
Abstract
Currently there are no potential curative therapies that can improve the central nervous system (CNS) regeneration after traumatic injuries or diseases. Indeed, the regeneration of CNS is greatly impaired by limited drug penetration across the blood brain barrier (BBB), poor drug targeting, deficient progenitor neural cells and limited proliferation of mature neural cells. To overcome these limitations, bioengineered injectable hydrogels in combination with drug and cell therapy have been proposed to mimic the complexity of the CNS microenvironment and architecture. Additionally, to enhance relevant CNS regeneration, proper biophysical and biochemical cues are needed. Recently, great efforts have been devoted to tailor stimuli-responsive hydrogels as novel carrier systems which are able to guide neural tissue regeneration. This review provides an extensive overview on the most promising injectable hydrogels for neural tissue engineering. A special emphasis is made to highlight the ability of these hydrogels to deliver bioactive compounds/cells upon the exposure to internal and external stimuli. Bioactive injectable hydrogels have a broad application in central nervous system's (CNS) regeneration. This review gives an overview of the latest pioneering approaches in CNS recovery using stimuli-responsive hydrogels for several neurodegenerative disorders. STATEMENT OF SIGNIFICANCE: This review summarizes the latest innovations on bioactive injectable hydrogels, focusing on tailoring internal/external stimuli-responsive hydrogels for the new injectable systems design, able to guide neural tissue response. The purpose is to highlight the advantages and the limitations of thermo-responsive, photo responsive, magnetic responsive, electric responsive, ultrasound responsive and enzymes-triggered injectable hydrogels in developing customizable neurotherapies. We believe that this comprehensive review will help in identifying the strengths and gaps in the existing literature and to further support the use of injectable hydrogels in stimulating CNS regeneration.
Collapse
|
33
|
Augmenting Peripheral Nerve Regeneration with Adipose-Derived Stem Cells. Stem Cell Rev Rep 2022; 18:544-558. [PMID: 34417730 PMCID: PMC8858329 DOI: 10.1007/s12015-021-10236-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/04/2021] [Indexed: 02/03/2023]
Abstract
Peripheral nerve injuries (PNIs) are common and debilitating, cause significant health care costs for society, and rely predominately on autografts, which necessitate grafting a nerve section non-locally to repair the nerve injury. One possible approach to improving treatment is bolstering endogenous regenerative mechanisms or bioengineering new nervous tissue in the peripheral nervous system. In this review, we discuss critical-sized nerve gaps and nerve regeneration in rats, and summarize the roles of adipose-derived stem cells (ADSCs) in the treatment of PNIs. Several regenerative treatment modalities for PNI are described: ADSCs differentiating into Schwann cells (SCs), ADSCs secreting growth factors to promote peripheral nerve growth, ADSCs promoting myelination growth, and ADSCs treatments with scaffolds. ADSCs' roles in regenerative treatment and features are compared to mesenchymal stem cells, and the administration routes, cell dosages, and cell fates are discussed. ADSCs secrete neurotrophic factors and exosomes and can differentiate into Schwann cell-like cells (SCLCs) that share features with naturally occurring SCs, including the ability to promote nerve regeneration in the PNS. Future clinical applications are also discussed.
Collapse
|
34
|
Promotion of Adrenal Pheochromocytoma (PC-12) Cell Proliferation and Outgrowth Using Schwann Cell-Laden Gelatin Methacrylate Substrate. Gels 2022; 8:gels8020084. [PMID: 35200467 PMCID: PMC8871842 DOI: 10.3390/gels8020084] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 01/25/2022] [Accepted: 01/25/2022] [Indexed: 12/15/2022] Open
Abstract
Peripheral nerve injuries cause different degrees of nerve palsy and function loss. Due to the limitations of autografts, nerve tissue engineering (TE) scaffolds incorporated with various neurotrophic factors and cells have been investigated to promote nerve regeneration. However, the molecular mechanism is still poorly understood. In this study, we co-cultured Schwann cells (SCs) and rat adrenal pheochromocytoma (PC-12) cells on 50% degrees of methacryloyl substitution gelatin methacrylate (GelMA) scaffold. The SCs were encapsulated within the GelMA, and PC-12 cells were on the surface. A 5% GelMA was used as the co-culture scaffold since it better supports SCs proliferation, viability, and myelination and promotes higher neurotrophic factors secretion than 10% GelMA. In the co-culture, PC-12 cells demonstrated a higher cell proliferation rate and axonal extension than culturing without SCs, indicating that the secretion of neurotrophic factors from SCs can stimulate PC-12 growth and axonal outgrowth. The mRNA level for neurotrophic factors of SCs in 5% GelMA was further evaluated. We found significant upregulation when compared with a 2D culture, which suggested that this co-culture system could be a potential scaffold to investigate the mechanism of how SCs affect neuronal behaviors.
Collapse
|
35
|
Optimal Morphometric Characteristics of a Tubular Polymeric Scaffold to Promote Peripheral Nerve Regeneration: A Scoping Review. Polymers (Basel) 2022; 14:polym14030397. [PMID: 35160387 PMCID: PMC8838152 DOI: 10.3390/polym14030397] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 01/05/2022] [Accepted: 01/13/2022] [Indexed: 12/27/2022] Open
Abstract
Cellular behavior in nerve regeneration is affected by the architecture of the polymeric nerve guide conduits (NGCs); therefore, design features of polymeric NGCs are critical for neural tissue engineering. Hence, the purpose of this scoping review is to summarize the adequate quantitative/morphometric parameters of the characteristics of NGC that provide a supportive environment for nerve regeneration, enhancing the understanding of a previous study. 394 studies were found, of which 29 studies were selected. The selected studies revealed four morphometric characteristics for promoting nerve regeneration: wall thickness, fiber size, pore size, and porosity. An NGC with a wall thickness between 250–400 μm and porosity of 60–80%, with a small pore on the inner surface and a large pore on the outer surface, significantly favored nerve regeneration; resulting in an increase in nutrient permeability, retention of neurotrophic factors, and optimal mechanical properties. On the other hand, the superiority of electrospun fibers is described; however, the size of the fiber is controversial in the literature, obtaining optimal results in the range of 300 nm to 30 µm. The incorporation of these optimal morphometric characteristics will encourage nerve regeneration and help reduce the number of experimental studies as it will provide the initial morphometric parameters for the preparation of an NGC.
Collapse
|
36
|
Li Y, Fraser D, Mereness J, Van Hove A, Basu S, Newman M, Benoit DSW. Tissue Engineered Neurovascularization Strategies for Craniofacial Tissue Regeneration. ACS APPLIED BIO MATERIALS 2022; 5:20-39. [PMID: 35014834 PMCID: PMC9016342 DOI: 10.1021/acsabm.1c00979] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Craniofacial tissue injuries, diseases, and defects, including those within bone, dental, and periodontal tissues and salivary glands, impact an estimated 1 billion patients globally. Craniofacial tissue dysfunction significantly reduces quality of life, and successful repair of damaged tissues remains a significant challenge. Blood vessels and nerves are colocalized within craniofacial tissues and act synergistically during tissue regeneration. Therefore, the success of craniofacial regenerative approaches is predicated on successful recruitment, regeneration, or integration of both vascularization and innervation. Tissue engineering strategies have been widely used to encourage vascularization and, more recently, to improve innervation through host tissue recruitment or prevascularization/innervation of engineered tissues. However, current scaffold designs and cell or growth factor delivery approaches often fail to synergistically coordinate both vascularization and innervation to orchestrate successful tissue regeneration. Additionally, tissue engineering approaches are typically investigated separately for vascularization and innervation. Since both tissues act in concert to improve craniofacial tissue regeneration outcomes, a revised approach for development of engineered materials is required. This review aims to provide an overview of neurovascularization in craniofacial tissues and strategies to target either process thus far. Finally, key design principles are described for engineering approaches that will support both vascularization and innervation for successful craniofacial tissue regeneration.
Collapse
Affiliation(s)
- Yiming Li
- Department of Biomedical Engineering, University of Rochester, Rochester, New York 14627, United States.,Department of Orthopaedics and Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York 14642, United States
| | - David Fraser
- Department of Orthopaedics and Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York 14642, United States.,Eastman Institute for Oral Health, University of Rochester Medical Center, Rochester, New York 14620, United States.,Translational Biomedical Sciences Program, University of Rochester Medical Center, Rochester, New York 14642, United States
| | - Jared Mereness
- Department of Biomedical Engineering, University of Rochester, Rochester, New York 14627, United States.,Department of Orthopaedics and Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York 14642, United States.,Department of Environmental Medicine, University of Rochester Medical Center, Rochester, New York 14642, United States
| | - Amy Van Hove
- Department of Biomedical Engineering, University of Rochester, Rochester, New York 14627, United States.,Department of Orthopaedics and Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York 14642, United States
| | - Sayantani Basu
- Department of Biomedical Engineering, University of Rochester, Rochester, New York 14627, United States.,Department of Orthopaedics and Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York 14642, United States
| | - Maureen Newman
- Department of Biomedical Engineering, University of Rochester, Rochester, New York 14627, United States.,Department of Orthopaedics and Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York 14642, United States
| | - Danielle S W Benoit
- Department of Biomedical Engineering, University of Rochester, Rochester, New York 14627, United States.,Department of Orthopaedics and Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York 14642, United States.,Eastman Institute for Oral Health, University of Rochester Medical Center, Rochester, New York 14620, United States.,Translational Biomedical Sciences Program, University of Rochester Medical Center, Rochester, New York 14642, United States.,Department of Environmental Medicine, University of Rochester Medical Center, Rochester, New York 14642, United States.,Materials Science Program, University of Rochester, Rochester, New York 14627, United States.,Department of Chemical Engineering, University of Rochester, Rochester, New York 14627, United States.,Department of Biomedical Genetics and Center for Oral Biology, University of Rochester Medical Center, Rochester, New York 14642, United States
| |
Collapse
|
37
|
Cao S, Deng Y, Zhang L, Aleahmad M. Chitosan nanoparticles, as biological macromolecule-based drug delivery systems to improve the healing potential of artificial neural guidance channels: A review. Int J Biol Macromol 2022; 201:569-579. [PMID: 35031319 DOI: 10.1016/j.ijbiomac.2022.01.017] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 12/25/2021] [Accepted: 01/05/2022] [Indexed: 12/13/2022]
Abstract
The healing potential of artificial neural guidance channels (NGCs) can be improved by various approaches such as seeding them with supporting cells, the incorporation of various cues, and modification with different fabrication methods. Recently, the therapeutic appeal towards the use of drug-delivering NGCs has increased. In this framework, neuroprotective agents are incorporated into the structure of NGCs using different techniques. Among available methods, nanoparticle-based drug carriers offer numerous advantages over other formulations such as controlled drug release, targeted delivery, high encapsulation efficacy, and high surface to volume ratio. Chitosan nanoparticles have different interesting features for drug delivery applications. These nanocarriers are biocompatible, biodegradable, non-immunogenic, stable, and possess tunable properties. In the current review, applications, challenges, and future perspectives of drug-loaded chitosan nanoparticles to augment the healing potential of NGCs will be discussed.
Collapse
Affiliation(s)
- Shuang Cao
- Department of Neuroelectrophysiology, Jinan Children's Hospital, Jinan 250022, Shandong, China
| | - Yang Deng
- School of Public Health and Management, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian 271016, Shandong, China.
| | - Le Zhang
- School of Public Health and Management, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian 271016, Shandong, China.
| | - Mehdi Aleahmad
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
38
|
Zhang F, Zhang M, Liu S, Li C, Ding Z, Wan T, Zhang P. Application of Hybrid Electrically Conductive Hydrogels Promotes Peripheral Nerve Regeneration. Gels 2022; 8:41. [PMID: 35049576 PMCID: PMC8775167 DOI: 10.3390/gels8010041] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 12/18/2021] [Accepted: 01/01/2022] [Indexed: 12/12/2022] Open
Abstract
Peripheral nerve injury (PNI) occurs frequently, and the prognosis is unsatisfactory. As the gold standard of treatment, autologous nerve grafting has several disadvantages, such as lack of donors and complications. The use of functional biomaterials to simulate the natural microenvironment of the nervous system and the combination of different biomaterials are considered to be encouraging alternative methods for effective tissue regeneration and functional restoration of injured nerves. Considering the inherent presence of an electric field in the nervous system, electrically conductive biomaterials have been used to promote nerve regeneration. Due to their singular physical properties, hydrogels can provide a three-dimensional hydrated network that can be integrated into diverse sizes and shapes and stimulate the natural functions of nerve tissue. Therefore, conductive hydrogels have become the most effective biological material to simulate human nervous tissue's biological and electrical characteristics. The principal merits of conductive hydrogels include their physical properties and their electrical peculiarities sufficient to effectively transmit electrical signals to cells. This review summarizes the recent applications of conductive hydrogels to enhance peripheral nerve regeneration.
Collapse
Affiliation(s)
- Fengshi Zhang
- Department of Orthopedics and Trauma, Peking University People’s Hospital, Beijing 100044, China; (F.Z.); (M.Z.); (S.L.); (C.L.); (Z.D.); (T.W.)
- Key Laboratory of Trauma and Neural Regeneration, Ministry of Education, Beijing 100044, China
- National Center for Trauma Medicine, Beijing 100044, China
| | - Meng Zhang
- Department of Orthopedics and Trauma, Peking University People’s Hospital, Beijing 100044, China; (F.Z.); (M.Z.); (S.L.); (C.L.); (Z.D.); (T.W.)
- Key Laboratory of Trauma and Neural Regeneration, Ministry of Education, Beijing 100044, China
- National Center for Trauma Medicine, Beijing 100044, China
| | - Songyang Liu
- Department of Orthopedics and Trauma, Peking University People’s Hospital, Beijing 100044, China; (F.Z.); (M.Z.); (S.L.); (C.L.); (Z.D.); (T.W.)
- Key Laboratory of Trauma and Neural Regeneration, Ministry of Education, Beijing 100044, China
- National Center for Trauma Medicine, Beijing 100044, China
| | - Ci Li
- Department of Orthopedics and Trauma, Peking University People’s Hospital, Beijing 100044, China; (F.Z.); (M.Z.); (S.L.); (C.L.); (Z.D.); (T.W.)
- Key Laboratory of Trauma and Neural Regeneration, Ministry of Education, Beijing 100044, China
- National Center for Trauma Medicine, Beijing 100044, China
| | - Zhentao Ding
- Department of Orthopedics and Trauma, Peking University People’s Hospital, Beijing 100044, China; (F.Z.); (M.Z.); (S.L.); (C.L.); (Z.D.); (T.W.)
- Key Laboratory of Trauma and Neural Regeneration, Ministry of Education, Beijing 100044, China
- National Center for Trauma Medicine, Beijing 100044, China
| | - Teng Wan
- Department of Orthopedics and Trauma, Peking University People’s Hospital, Beijing 100044, China; (F.Z.); (M.Z.); (S.L.); (C.L.); (Z.D.); (T.W.)
- Key Laboratory of Trauma and Neural Regeneration, Ministry of Education, Beijing 100044, China
- National Center for Trauma Medicine, Beijing 100044, China
| | - Peixun Zhang
- Department of Orthopedics and Trauma, Peking University People’s Hospital, Beijing 100044, China; (F.Z.); (M.Z.); (S.L.); (C.L.); (Z.D.); (T.W.)
- Key Laboratory of Trauma and Neural Regeneration, Ministry of Education, Beijing 100044, China
- National Center for Trauma Medicine, Beijing 100044, China
| |
Collapse
|
39
|
Piras CC, Kay AG, Genever PG, Fitremann J, Smith DK. Self-assembled gel tubes, filaments and 3D-printing with in situ metal nanoparticle formation and enhanced stem cell growth. Chem Sci 2022; 13:1972-1981. [PMID: 35308847 PMCID: PMC8848986 DOI: 10.1039/d1sc06062g] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 01/16/2022] [Indexed: 12/18/2022] Open
Abstract
This paper reports simple strategies to fabricate self-assembled artificial tubular and filamentous systems from a low molecular weight gelator (LMWG). In the first strategy, tubular ‘core–shell’ gel structures based on the dibenzylidenesorbitol-based LMWG DBS-CONHNH2 were made in combination with the polymer gelator (PG) calcium alginate. In the second approach, gel filaments based on DBS-CONHNH2 alone were prepared by wet spinning at elevated concentrations using a ‘solvent-switch’ approach. The higher concentrations used in wet-spinning prevent the need for a supporting PG. Furthermore, this can be extended into a 3D-printing method, with the printed LMWG objects showing excellent stability for at least a week in water. The LMWG retains its unique ability for in situ precious metal reduction, yielding Au nanoparticles (AuNPs) within the tubes and filaments when they are exposed to AuCl3 solutions. Since the gel filaments have a higher loading of DBS-CONHNH2, they can be loaded with significantly more AuNPs. Cytotoxicity and viability studies on human mesenchymal stem cells show that the DBS-CONHNH2 and DBS-CONHNH2/alginate hybrid gels loaded with AuNPs are biocompatible, with the presence of AuNPs enhancing stem cell metabolism. Taken together, these results indicate that DBS-CONHNH2 can be shaped and 3D-printed, and has considerable potential for use in tissue engineering applications. Simple fabrication and 3D-printing methods are used to generate tubes and filaments from self-assembled gels, which can be loaded in situ with gold nanoparticles, with the resulting gels encouraging stem cell proliferation.![]()
Collapse
Affiliation(s)
- Carmen C. Piras
- Department of Chemistry, University of York, Heslington, York, YO10 5DD, UK
| | - Alasdair G. Kay
- Department of Biology, University of York, Heslington, York, YO10 5DD, UK
| | - Paul G. Genever
- Department of Biology, University of York, Heslington, York, YO10 5DD, UK
| | - Juliette Fitremann
- IMRCP, UMR 5623, CNRS, Université de Toulouse, 118 Route de Narbonne, F-31062 Toulouse, France
| | - David K. Smith
- Department of Chemistry, University of York, Heslington, York, YO10 5DD, UK
| |
Collapse
|
40
|
Rizwana N, Agarwal V, Nune M. Antioxidant for Neurological Diseases and Neurotrauma and Bioengineering Approaches. Antioxidants (Basel) 2021; 11:72. [PMID: 35052576 PMCID: PMC8773039 DOI: 10.3390/antiox11010072] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Revised: 12/10/2021] [Accepted: 12/20/2021] [Indexed: 12/16/2022] Open
Abstract
Antioxidants are a class of molecules with an innate affinity to neutralize reactive oxygen species (ROS), which are known to cause oxidative stress. Oxidative stress has been associated with a wide range of diseases mediated by physiological damage to the cells. ROS play both beneficial and detrimental roles in human physiology depending on their overall concentration. ROS are an inevitable byproduct of the normal functioning of cells, which are produced as a result of the mitochondrial respiration process. Since the establishment of the detrimental effect of oxidative stress in neurological disorders and neurotrauma, there has been growing interest in exploring antioxidants to rescue remaining or surviving cells and reverse the neurological damage. In this review, we present the survey of different antioxidants studied in neurological applications including neurotrauma. We also delve into bioengineering approaches developed to deliver antioxidants to improve their cellular uptake in neurological applications.
Collapse
Affiliation(s)
- Nasera Rizwana
- Manipal Institute of Regenerative Medicine (MIRM), Bengaluru, Manipal Academy of Higher Education (MAHE), Manipal 576104, India;
| | - Vipul Agarwal
- Cluster for Advanced Macromolecular Design (CAMD), School of Chemical Engineering, University of New South Wales, Sydney, NSW 2052, Australia
| | - Manasa Nune
- Manipal Institute of Regenerative Medicine (MIRM), Bengaluru, Manipal Academy of Higher Education (MAHE), Manipal 576104, India;
| |
Collapse
|
41
|
Hydrogel, Electrospun and Composite Materials for Bone/Cartilage and Neural Tissue Engineering. MATERIALS 2021; 14:ma14226899. [PMID: 34832300 PMCID: PMC8624846 DOI: 10.3390/ma14226899] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 11/09/2021] [Accepted: 11/11/2021] [Indexed: 12/15/2022]
Abstract
Injuries of the bone/cartilage and central nervous system are still a serious socio-economic problem. They are an effect of diversified, difficult-to-access tissue structures as well as complex regeneration mechanisms. Currently, commercially available materials partially solve this problem, but they do not fulfill all of the bone/cartilage and neural tissue engineering requirements such as mechanical properties, biochemical cues or adequate biodegradation. There are still many things to do to provide complete restoration of injured tissues. Recent reports in bone/cartilage and neural tissue engineering give high hopes in designing scaffolds for complete tissue regeneration. This review thoroughly discusses the advantages and disadvantages of currently available commercial scaffolds and sheds new light on the designing of novel polymeric scaffolds composed of hydrogels, electrospun nanofibers, or hydrogels loaded with nano-additives.
Collapse
|
42
|
Nelson DW, Gilbert RJ. Extracellular Matrix-Mimetic Hydrogels for Treating Neural Tissue Injury: A Focus on Fibrin, Hyaluronic Acid, and Elastin-Like Polypeptide Hydrogels. Adv Healthc Mater 2021; 10:e2101329. [PMID: 34494398 PMCID: PMC8599642 DOI: 10.1002/adhm.202101329] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 08/23/2021] [Indexed: 12/19/2022]
Abstract
Neurological and functional recovery is limited following central nervous system injury and severe injury to the peripheral nervous system. Extracellular matrix (ECM)-mimetic hydrogels are of particular interest as regenerative scaffolds for the injured nervous system as they provide 3D bioactive interfaces that modulate cellular response to the injury environment and provide naturally degradable scaffolding for effective tissue remodeling. In this review, three unique ECM-mimetic hydrogels used in models of neural injury are reviewed: fibrin hydrogels, which rely on a naturally occurring enzymatic gelation, hyaluronic acid hydrogels, which require chemical modification prior to chemical crosslinking, and elastin-like polypeptide (ELP) hydrogels, which exhibit a temperature-sensitive gelation. The hydrogels are reviewed by summarizing their unique biological properties, their use as drug depots, and their combination with other biomaterials, such as electrospun fibers and nanoparticles. This review is the first to focus on these three ECM-mimetic hydrogels for their use in neural tissue engineering. Additionally, this is the first review to summarize the use of ELP hydrogels for nervous system applications. ECM-mimetic hydrogels have shown great promise in preclinical models of neural injury and future advancements in their design and use can likely lead to viable treatments for patients with neural injury.
Collapse
Affiliation(s)
- Derek W Nelson
- Department of Biomedical Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 110 8th St, Troy, NY, 12180, USA
| | - Ryan J Gilbert
- Department of Biomedical Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 110 8th St, Troy, NY, 12180, USA
| |
Collapse
|
43
|
Xu K, Liu X, Li X, Yin J, Wei P, Qian J, Sun J. Effect of Electrical and Electromechanical Stimulation on PC12 Cell Proliferation and Axon Outgrowth. Front Bioeng Biotechnol 2021; 9:757906. [PMID: 34746110 PMCID: PMC8566739 DOI: 10.3389/fbioe.2021.757906] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 09/15/2021] [Indexed: 11/13/2022] Open
Abstract
Peripheral nerve injuries have become a common clinical disease with poor prognosis and complicated treatments. The development of tissue engineering pointed a promising direction to produce nerve conduits for nerve regeneration. Electrical and mechanical stimulations have been incorporated with tissue engineering, since such external stimulations could promote nerve cell proliferation, migration and differentiation. However, the combination of electrical and mechanical stimulations (electromechanical stimulation) and its effects on neuron proliferation and axon outgrowth have been rarely investigated. Herein, silver nanowires (AgNWs) embedded polydimethylsiloxane (PDMS) electrodes were developed to study the effects of electromechanical stimulation on rat pheochromocytoma cells (PC12 cells) behaviors. AgNWs/PDMS electrodes demonstrated good biocompatibility and established a stable electric field during mechanical stretching. PC12 cells showed enhanced proliferation rate and axon outgrowth under electrical stimulation alone, and the cell number significantly increased with higher electrical stimulation intensity. The involvement of mechanical stretching in electrical stimulation reduced the cell proliferation rate and axon outgrowth, compared with the case of electrical stimulation alone. Interestingly, the cellular axons outgrowth was found to depend on the stretching direction, where the axons prefer to align perpendicularly to the stretch direction. These results suggested that AgNWs/PDMS electrodes provide an in vitro platform to investigate the effects of electromechanical stimulation on nerve cell behaviors and can be potentially used for nerve regeneration in the future.
Collapse
Affiliation(s)
- Kailei Xu
- Central Laboratory, Ningbo First Hospital, Ningbo, China
- The State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, China
- Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, School of Mechanical Engineering, Zhejiang University, Hangzhou, China
| | - Xixia Liu
- The State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, China
- Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, School of Mechanical Engineering, Zhejiang University, Hangzhou, China
- School of Mechanical Engineering, Guizhou University, Guiyang, China
| | - Xiaokeng Li
- The State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, China
- Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, School of Mechanical Engineering, Zhejiang University, Hangzhou, China
| | - Jun Yin
- The State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, China
- Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, School of Mechanical Engineering, Zhejiang University, Hangzhou, China
| | - Peng Wei
- Department of Hand and Foot Microsurgery, Ningbo First Hospital, Ningbo, China
| | - Jin Qian
- Key Laboratory of Soft Machines and Smart Devices of Zhejiang Province, Department of Engineering Mechanics, Zhejiang University, Hangzhou, China
| | - Jie Sun
- Central Laboratory, Ningbo First Hospital, Ningbo, China
- Department of Neurosurgery, Ningbo First Hospital, Ningbo, China
| |
Collapse
|
44
|
Burks SS, Diaz A, Haggerty AE, Oliva NDL, Midha R, Levi AD. Schwann cell delivery via a novel 3D collagen matrix conduit improves outcomes in critical length nerve gap repairs. J Neurosurg 2021; 135:1241-1251. [PMID: 33607621 DOI: 10.3171/2020.8.jns202349] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 08/25/2020] [Indexed: 11/06/2022]
Abstract
OBJECTIVE The current clinical standard of harvesting a nerve autograft for repair of long-segment peripheral nerve injuries (PNIs) is associated with many potential complications. Guidance channels offer an alternative therapy. The authors investigate whether autologous Schwann cells (SCs) implanted within a novel collagen-glycosaminoglycan conduit will improve axonal regeneration in a long-segment PNI model. METHODS Novel NeuraGen 3D collagen matrix conduits were implanted with autologous SCs to investigate axonal regeneration across a critical size defect (13 mm) in male Fischer rat sciatic nerve. Reversed sciatic nerve autografts served as positive controls, and conduits filled with serum only as negative controls. Electrophysiological assessments were made in vivo. Animals were killed at 4 or 16 weeks postinjury, muscle weights were measured, and grafts underwent immunohistochemical and morphometric analysis. RESULTS SC survival was confirmed by the presence of green fluorescent protein-labeled SCs within regenerated fibers. Regeneration and elongation of myelinated axons in all segments of the graft were significantly enhanced at 16 weeks in the SC-filled conduits compared to the conduit alone and were statistically similar to those of the autograft. Nerves repaired with SC-filled conduits exhibited onset latencies and nerve conduction amplitudes similar to those of the contralateral controls and autograft (p < 0.05). Adding SCs to the conduit also significantly reduced muscle atrophy compared to conduit alone (p < 0.0001). CONCLUSIONS Repair of long-segment PNI of rat sciatic nerve is significantly enhanced by SC-filled NeuraGen 3D conduits. Improvements in the total number of myelinated axons, axon diameter, and myelin thickness throughout SC-filled conduits allow for significant recovery in nerve conduction and a decrease in muscle atrophy.
Collapse
Affiliation(s)
- S Shelby Burks
- 1Department of Neurological Surgery and the Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, Florida; and
| | - Anthony Diaz
- 1Department of Neurological Surgery and the Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, Florida; and
| | - Agnes E Haggerty
- 1Department of Neurological Surgery and the Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, Florida; and
| | - Natalia de la Oliva
- 1Department of Neurological Surgery and the Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, Florida; and
| | - Rajiv Midha
- 2Department of Clinical Neurosciences and Hotchkiss Brain Institute, University of Calgary, Alberta, Canada
| | - Allan D Levi
- 1Department of Neurological Surgery and the Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, Florida; and
| |
Collapse
|
45
|
Bioactivated Oxidized Polyvinyl Alcohol towards Next-Generation Nerve Conduits Development. Polymers (Basel) 2021; 13:polym13193372. [PMID: 34641183 PMCID: PMC8512895 DOI: 10.3390/polym13193372] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 09/21/2021] [Accepted: 09/26/2021] [Indexed: 12/19/2022] Open
Abstract
The limitations and difficulties that nerve autografts create in normal nerve function recovery after injury is driving research towards using smart materials for next generation nerve conduits (NCs) setup. Here, the new polymer partially oxidized polyvinyl alcohol (OxPVA) was assayed to verify its future potential as a bioactivated platform for advanced/effective NCs. OxPVA-patterned scaffolds (obtained by a 3D-printed mold) with/without biochemical cues (peptide IKVAV covalently bound (OxPVA-IKVAV) or self-assembling peptide EAK (sequence: AEAEAKAKAEAEAKAK), mechanically incorporated (OxPVA+EAK) versus non-bioactivated scaffold (peptide-free OxPVA (PF-OxPVA) supports, OxPVA without IKVAV and OxPVA without EAK control scaffolds) were compared for their biological effect on neuronal SH-SY5Y cells. After cell seeding, adhesion/proliferation, mediated by (a) precise control over scaffolds surface ultrastructure; (b) functionalization efficacy guaranteed by bioactive cues (IKVAV/EAK), was investigated by MTT assay at 3, 7, 14 and 21 days. As shown by the results, the patterned groove alone stimulates colonization by cells; however, differences were observed when comparing the scaffold types over time. In the long period (21 days), patterned OxPVA+EAK scaffolds distinguished in bioactivity, assuring a significantly higher total cell amount than the other groups. Experimental evidence suggests patterned OxPVA-EAK potential for NCs device fabrication.
Collapse
|
46
|
Ramesh PA, Dhandapani R, Bagewadi S, Zennifer A, Radhakrishnan J, Sethuraman S, Subramanian A. Reverse engineering of an anatomically equivalent nerve conduit. J Tissue Eng Regen Med 2021; 15:998-1011. [PMID: 34551457 DOI: 10.1002/term.3245] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 07/29/2021] [Accepted: 09/02/2021] [Indexed: 01/12/2023]
Abstract
Reconstruction of peripheral nervous tissue remains challenging in critical-sized defects due to the lack of Büngner bands from the proximal to the distal nerve ends. Conventional nerve guides fail to bridge the large-sized defect owing to the formation of a thin fibrin cable. Hence, in the present study, an attempt was made to reverse engineer the intricate epi-, peri- and endo-neurial tissues using Fused Deposition Modeling based 3D printing. Bovine serum albumin protein nanoflowers (NF) exhibiting Viburnum opulus 'Roseum' morphology were ingrained into 3D printed constructs without affecting its secondary structure to enhance the axonal guidance from proximal to distal ends of denuded nerve ends. Scanning electron micrographs confirmed the uniform distribution of protein NF in 3D printed constructs. The PC-12 cells cultured on protein ingrained 3D printed scaffolds demonstrated cytocompatibility, improved cell adhesion and extended neuronal projections with significantly higher intensities of NF-200 and tubulin expressions. Further suture-free fixation designed in the current 3D printed construct aids facile implantation of printed conduits to the transected nerve ends. Hence the protein ingrained 3D printed construct would be a promising substitute to treat longer peripheral nerve defects as its structural equivalence of endo- and perineurial organization along with the ingrained protein NF promote the neuronal extension towards the distal ends by minimizing axonal dispersion.
Collapse
Affiliation(s)
- Preethy Amruthavarshini Ramesh
- Tissue Engineering and Additive Manufacturing (TEAM) Lab, Centre for Nanotechnology & Advanced Biomaterials, ABCDE Innovation Centre, School of Chemical & Biotechnology, SASTRA Deemed University, Thanjavur, India
| | - Ramya Dhandapani
- Tissue Engineering and Additive Manufacturing (TEAM) Lab, Centre for Nanotechnology & Advanced Biomaterials, ABCDE Innovation Centre, School of Chemical & Biotechnology, SASTRA Deemed University, Thanjavur, India
| | - Shambhavi Bagewadi
- Tissue Engineering and Additive Manufacturing (TEAM) Lab, Centre for Nanotechnology & Advanced Biomaterials, ABCDE Innovation Centre, School of Chemical & Biotechnology, SASTRA Deemed University, Thanjavur, India
| | - Allen Zennifer
- Tissue Engineering and Additive Manufacturing (TEAM) Lab, Centre for Nanotechnology & Advanced Biomaterials, ABCDE Innovation Centre, School of Chemical & Biotechnology, SASTRA Deemed University, Thanjavur, India
| | - Janani Radhakrishnan
- Tissue Engineering and Additive Manufacturing (TEAM) Lab, Centre for Nanotechnology & Advanced Biomaterials, ABCDE Innovation Centre, School of Chemical & Biotechnology, SASTRA Deemed University, Thanjavur, India
| | - Swaminathan Sethuraman
- Tissue Engineering and Additive Manufacturing (TEAM) Lab, Centre for Nanotechnology & Advanced Biomaterials, ABCDE Innovation Centre, School of Chemical & Biotechnology, SASTRA Deemed University, Thanjavur, India
| | - Anuradha Subramanian
- Tissue Engineering and Additive Manufacturing (TEAM) Lab, Centre for Nanotechnology & Advanced Biomaterials, ABCDE Innovation Centre, School of Chemical & Biotechnology, SASTRA Deemed University, Thanjavur, India
| |
Collapse
|
47
|
Cintron-Colon AF, Almeida-Alves G, VanGyseghem JM, Spitsbergen JM. GDNF to the rescue: GDNF delivery effects on motor neurons and nerves, and muscle re-innervation after peripheral nerve injuries. Neural Regen Res 2021; 17:748-753. [PMID: 34472460 PMCID: PMC8530131 DOI: 10.4103/1673-5374.322446] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Peripheral nerve injuries commonly occur due to trauma, like a traffic accident. Peripheral nerves get severed, causing motor neuron death and potential muscle atrophy. The current golden standard to treat peripheral nerve lesions, especially lesions with large (≥ 3 cm) nerve gaps, is the use of a nerve autograft or reimplantation in cases where nerve root avulsions occur. If not tended early, degeneration of motor neurons and loss of axon regeneration can occur, leading to loss of function. Although surgical procedures exist, patients often do not fully recover, and quality of life deteriorates. Peripheral nerves have limited regeneration, and it is usually mediated by Schwann cells and neurotrophic factors, like glial cell line-derived neurotrophic factor, as seen in Wallerian degeneration. Glial cell line-derived neurotrophic factor is a neurotrophic factor known to promote motor neuron survival and neurite outgrowth. Glial cell line-derived neurotrophic factor is upregulated in different forms of nerve injuries like axotomy, sciatic nerve crush, and compression, thus creating great interest to explore this protein as a potential treatment for peripheral nerve injuries. Exogenous glial cell line-derived neurotrophic factor has shown positive effects in regeneration and functional recovery when applied in experimental models of peripheral nerve injuries. In this review, we discuss the mechanism of repair provided by Schwann cells and upregulation of glial cell line-derived neurotrophic factor, the latest findings on the effects of glial cell line-derived neurotrophic factor in different types of peripheral nerve injuries, delivery systems, and complementary treatments (electrical muscle stimulation and exercise). Understanding and overcoming the challenges of proper timing and glial cell line-derived neurotrophic factor delivery is paramount to creating novel treatments to tend to peripheral nerve injuries to improve patients’ quality of life.
Collapse
Affiliation(s)
| | | | | | - John M Spitsbergen
- Biological Sciences Department, Western Michigan University, Kalamazoo, MI, USA
| |
Collapse
|
48
|
Litowczenko J, Woźniak-Budych MJ, Staszak K, Wieszczycka K, Jurga S, Tylkowski B. Milestones and current achievements in development of multifunctional bioscaffolds for medical application. Bioact Mater 2021; 6:2412-2438. [PMID: 33553825 PMCID: PMC7847813 DOI: 10.1016/j.bioactmat.2021.01.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 12/23/2020] [Accepted: 01/07/2021] [Indexed: 12/13/2022] Open
Abstract
Tissue engineering (TE) is a rapidly growing interdisciplinary field, which aims to restore or improve lost tissue function. Despite that TE was introduced more than 20 years ago, innovative and more sophisticated trends and technologies point to new challenges and development. Current challenges involve the demand for multifunctional bioscaffolds which can stimulate tissue regrowth by biochemical curves, biomimetic patterns, active agents and proper cell types. For those purposes especially promising are carefully chosen primary cells or stem cells due to its high proliferative and differentiation potential. This review summarized a variety of recently reported advanced bioscaffolds which present new functions by combining polymers, nanomaterials, bioactive agents and cells depending on its desired application. In particular necessity of study biomaterial-cell interactions with in vitro cell culture models, and studies using animals with in vivo systems were discuss to permit the analysis of full material biocompatibility. Although these bioscaffolds have shown a significant therapeutic effect in nervous, cardiovascular and muscle, tissue engineering, there are still many remaining unsolved challenges for scaffolds improvement.
Collapse
Affiliation(s)
- Jagoda Litowczenko
- NanoBioMedical Centre, Adam Mickiewicz University in Poznan, Wszechnicy Piastowskiej 3, Poznan, Poland
| | - Marta J. Woźniak-Budych
- NanoBioMedical Centre, Adam Mickiewicz University in Poznan, Wszechnicy Piastowskiej 3, Poznan, Poland
| | - Katarzyna Staszak
- Institute of Technology and Chemical Engineering, Poznan University of Technology, ul. Berdychowo 4, Poznan, Poland
| | - Karolina Wieszczycka
- Institute of Technology and Chemical Engineering, Poznan University of Technology, ul. Berdychowo 4, Poznan, Poland
| | - Stefan Jurga
- NanoBioMedical Centre, Adam Mickiewicz University in Poznan, Wszechnicy Piastowskiej 3, Poznan, Poland
| | - Bartosz Tylkowski
- Eurecat, Centre Tecnològic de Catalunya, Chemical Technologies Unit, Marcel·lí Domingo s/n, Tarragona, 43007, Spain
| |
Collapse
|
49
|
Apablaza JA, Lezcano MF, Lopez Marquez A, Godoy Sánchez K, Oporto GH, Dias FJ. Main Morphological Characteristics of Tubular Polymeric Scaffolds to Promote Peripheral Nerve Regeneration-A Scoping Review. Polymers (Basel) 2021; 13:polym13152563. [PMID: 34372166 PMCID: PMC8347244 DOI: 10.3390/polym13152563] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 07/27/2021] [Accepted: 07/28/2021] [Indexed: 11/16/2022] Open
Abstract
The "nerve guide conduits" (NGC) used in nerve regeneration must mimic the natural environment for proper cell behavior. OBJECTIVE To describe the main morphological characteristics of polymeric NGC to promote nerve regeneration. METHODS A scoping review was performed following the Preferred Reporting Items for Systematic reviews and Meta-Analyses extension for Scoping Reviews (PRISMA-ScR) criteria in the PubMed, Web of Science, Science Direct, and Scientific Electronic Library Online (SciELO) databases. Primary studies that considered/evaluated morphological characteristics of NGC to promote nerve regeneration were included. RESULT A total of 704 studies were found, of which 52 were selected. The NGC main morphological characteristics found in the literature were: (I) NGC diameter affects the mechanical properties of the scaffold. (II) Wall thickness of NGC determines the exchange of nutrients, molecules, and neurotrophins between the internal and external environment; and influences the mechanical properties and biodegradation, similarly to NGC (III) porosity, (IV) pore size, and (V) pore distribution. The (VI) alignment of the NGC fibers influences the phenotype of cells involved in nerve regeneration. In addition, the (VII) thickness of the polymeric fiber influences neurite extension and orientation. CONCLUSIONS An NGC should have its diameter adjusted to the nerve with wall thickness, porosity, pore size, and distribution of pores, to favor vascularization, permeability, and exchange of nutrients, and retention of neurotrophic factors, also favoring its mechanical properties and biodegradability.
Collapse
Affiliation(s)
- Josefa Alarcón Apablaza
- Research Centre in Dental Sciences (CICO-UFRO), Dental School—Facultad de Odontología, Universidad de La Frontera, Temuco 4780000, Chile; (J.A.A.); (M.F.L.); (G.H.O.)
- Program of Master in Dental Science, Dental School, Universidad de La Frontera, Temuco 4780000, Chile
| | - María Florencia Lezcano
- Research Centre in Dental Sciences (CICO-UFRO), Dental School—Facultad de Odontología, Universidad de La Frontera, Temuco 4780000, Chile; (J.A.A.); (M.F.L.); (G.H.O.)
- Department of Integral Adults Dentistry, Dental School—Facultad de Odontología, Universidad de La Frontera, Temuco 4780000, Chile
- Laboratorio de Cibernética, Departamento de Bioingeniería, Facultad de Ingeniería, Universidad Nacional de Entre Ríos, Oro Verde 3100, Argentina
| | - Alex Lopez Marquez
- HAWK—Hochschule für Angewandte Wissenschaften und Kunst, 37085 Göttingen, Germany;
| | - Karina Godoy Sánchez
- Scientific and Technological Bioresource Nucleus (BIOREN-UFRO), Universidad de La Frontera, Temuco 4780000, Chile;
- Center of Molecular Biology and Phamacogenetics, Universidad de La Frontera, Temuco 4780000, Chile
| | - Gonzalo H. Oporto
- Research Centre in Dental Sciences (CICO-UFRO), Dental School—Facultad de Odontología, Universidad de La Frontera, Temuco 4780000, Chile; (J.A.A.); (M.F.L.); (G.H.O.)
- Department of Integral Adults Dentistry, Dental School—Facultad de Odontología, Universidad de La Frontera, Temuco 4780000, Chile
| | - Fernando José Dias
- Research Centre in Dental Sciences (CICO-UFRO), Dental School—Facultad de Odontología, Universidad de La Frontera, Temuco 4780000, Chile; (J.A.A.); (M.F.L.); (G.H.O.)
- Department of Integral Adults Dentistry, Dental School—Facultad de Odontología, Universidad de La Frontera, Temuco 4780000, Chile
- Correspondence: ; Tel.: +56-9-8474-6702
| |
Collapse
|
50
|
Garrudo FFF, Nogueira DES, Rodrigues CAV, Ferreira FA, Paradiso P, Colaço R, Marques AC, Cabral JMS, Morgado J, Linhardt RJ, Ferreira FC. Electrical stimulation of neural-differentiating iPSCs on novel coaxial electroconductive nanofibers. Biomater Sci 2021; 9:5359-5382. [PMID: 34223566 DOI: 10.1039/d1bm00503k] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Neural tissue engineering strategies are paramount to create fully mature neurons, necessary for new therapeutic strategies for neurological diseases or the creation of reliable in vitro models. Scaffolds can provide physical support for these neurons and enable cues for enhancing neural cell differentiation, such as electrical current. Coaxial electrospinning fibers, designed to fulfill neural cell needs, bring together an electroconductive shell layer (PCL-PANI), able to mediate electrical stimulation of cells cultivated on fibers mesh surface, and a soft core layer (PGS), used to finetune fiber diameter (951 ± 465 nm) and mechanical properties (1.3 ± 0.2 MPa). Those dual functional coaxial fibers are electroconductive (0.063 ± 0.029 S cm-1, stable over 21 days) and biodegradable (72% weigh loss in 12 hours upon human lipase accelerated assay). For the first time, the long-term effects of electrical stimulation on induced neural progenitor cells were studied using such fibers. The results show increase in neural maturation (upregulation of MAP2, NEF-H and SYP), up-regulation of glutamatergic marker genes (VGLUT1 - 15-fold) and voltage-sensitive channels (SCN1α - 12-fold, CACNA1C - 32-fold), and a down-regulation of GABAergic marker (GAD67 - 0.09-fold), as detected by qRT-PCR. Therefore, this study suggest a shift from an inhibitory to an excitatory neural cell profile. This work shows that the PGS/PCL-PANI coaxial fibers here developed have potential applications in neural tissue engineering.
Collapse
Affiliation(s)
- Fábio F F Garrudo
- Department of Chemistry and Chemical Biology, Department of Chemistry & Chemical Biology, Rensselaer Polytechnic Institute, Biotechnology Center 4005, Troy, NY 12180, USA. and Department of Bioengineering and iBB - Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001, Lisboa, Portugal. and Associate Laboratory i4HB-Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal and Department of Bioengineering and Instituto de Telecomunicações, Universidade de Lisboa, Av. Rovisco Pais, P-1049-001, Lisboa, Portugal
| | - Diogo E S Nogueira
- Department of Bioengineering and iBB - Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001, Lisboa, Portugal. and Associate Laboratory i4HB-Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - Carlos A V Rodrigues
- Department of Bioengineering and iBB - Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001, Lisboa, Portugal. and Associate Laboratory i4HB-Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - Flávio A Ferreira
- Department of Bioengineering and iBB - Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001, Lisboa, Portugal. and Associate Laboratory i4HB-Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - Patrizia Paradiso
- IDMEC - Instituto de Engenharia Mecânica, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, P-1049-001 Lisboa, Portugal
| | - Rogério Colaço
- IDMEC - Instituto de Engenharia Mecânica, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, P-1049-001 Lisboa, Portugal
| | - Ana C Marques
- CERENA, DEQ, Instituto Superior Técnico, Universidade de Lisboa, Avenida Rovisco Pais, P-1049-001 Lisboa, Portugal
| | - Joaquim M S Cabral
- Department of Bioengineering and iBB - Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001, Lisboa, Portugal. and Associate Laboratory i4HB-Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - Jorge Morgado
- Department of Bioengineering and Instituto de Telecomunicações, Universidade de Lisboa, Av. Rovisco Pais, P-1049-001, Lisboa, Portugal
| | - Robert J Linhardt
- Department of Chemistry and Chemical Biology, Department of Chemistry & Chemical Biology, Rensselaer Polytechnic Institute, Biotechnology Center 4005, Troy, NY 12180, USA.
| | - Frederico Castelo Ferreira
- Department of Bioengineering and iBB - Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001, Lisboa, Portugal. and Associate Laboratory i4HB-Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| |
Collapse
|