1
|
Yin W, Jiang Y, Ma G, Mbituyimana B, Xu J, Shi Z, Yang G, Chen H. A review: Carrier-based hydrogels containing bioactive molecules and stem cells for ischemic stroke therapy. Bioact Mater 2025; 49:39-62. [PMID: 40124600 PMCID: PMC11928985 DOI: 10.1016/j.bioactmat.2025.01.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 12/22/2024] [Accepted: 01/13/2025] [Indexed: 03/25/2025] Open
Abstract
Ischemic stroke (IS), a cerebrovascular disease, is the leading cause of physical disability and death worldwide. Tissue plasminogen activator (tPA) and thrombectomy are limited by a narrow therapeutic time window. Although strategies such as drug therapies and cellular therapies have been used in preclinical trials, some important issues in clinical translation have not been addressed: low stem cell survival and drug delivery limited by the blood-brain barrier (BBB). Among the therapeutic options currently sought, carrier-based hydrogels hold great promise for the repair and regeneration of neural tissue in the treatment of ischemic stroke. The advantage lies in the ability to deliver drugs and cells to designated parts of the brain in an injectable manner to enhance therapeutic efficacy. Here, this article provides an overview of the use of carrier-based hydrogels in ischemic stroke therapy and focuses on the use of hydrogel scaffolds containing bioactive molecules and stem cells. In addition to this, we provide a more in-depth summary of the composition, physicochemical properties and physiological functions of the materials themselves. Finally, we also outline the prospects and challenges for clinical translation of hydrogel therapy for IS.
Collapse
Affiliation(s)
- Wenqi Yin
- Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yuchi Jiang
- Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Guangrui Ma
- Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Bricard Mbituyimana
- Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Jia Xu
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zhijun Shi
- Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Guang Yang
- Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Hong Chen
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| |
Collapse
|
2
|
Nochalabadi A, Khazaei M, Rezakhani L. Exosomes and tissue engineering: A novel therapeutic strategy for nerve regenerative. Tissue Cell 2025; 93:102676. [PMID: 39693896 DOI: 10.1016/j.tice.2024.102676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 11/10/2024] [Accepted: 12/06/2024] [Indexed: 12/20/2024]
Abstract
Damage to nerves negatively impacts quality of life and causes considerable morbidity. Self-regeneration is a special characteristic of the nervous system, yet how successful regeneration is accomplished remains unclear. Research on nerve regeneration is advancing and accelerating successful nerve recovery with potential new approaches. Eukaryote cells release extracellular vesicles (EVs), which control intercellular communication in both health and disease. More and more, EVs such as microvesicles and exosomes (EXOs) are being recognized as viable options for cell-free therapies that address complex tissue regeneration. The present study highlights the functional relevance of EVs in regenerative medicine for nerve-related regeneration. A subclass of EVs, EXOs were first identified as a way for cells to expel undesirable cell products. These nanovesicles have a diameter of 30-150 nm and are secreted by a variety of cells in conditions of both health and illness. Their benefits include the ability to promote endothelial cell growth, inhibit inflammation, encourage cell proliferation, and regulate cell differentiation. They are also known to transport functional proteins, metabolites, and nucleic acids to recipient cells, thus playing a significant role in cellular communication. EXOs impact an extensive array of physiological functions, including immunological responses, tissue regeneration, stem cell conservation, communication within the central nervous system, and pathological processes involving cardiovascular disorders, neurodegeneration, cancer, and inflammation. Their biocompatibility and bi-layered lipid structure (which shields the genetic consignment from deterioration and reduces immunogenicity) make them appealing as therapeutic vectors. They can pass through the blood brain barrier and other major biological membranes because of their small size and membrane composition. The creation of modified EXOs is a dynamic area of research that supports the evaluation of diverse therapeutic freights, improvement of target selectivity, and manufacturing optimization.
Collapse
Affiliation(s)
- Azadeh Nochalabadi
- Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mozafar Khazaei
- Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran; Department of Tissue Engineering, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| | - Leila Rezakhani
- Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran; Department of Tissue Engineering, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
3
|
Borah R, Diez Clarke D, Upadhyay J, Monaghan MG. From innovation to clinic: Emerging strategies harnessing electrically conductive polymers to enhance electrically stimulated peripheral nerve repair. Mater Today Bio 2025; 30:101415. [PMID: 39816667 PMCID: PMC11733191 DOI: 10.1016/j.mtbio.2024.101415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 12/07/2024] [Accepted: 12/17/2024] [Indexed: 01/18/2025] Open
Abstract
Peripheral nerve repair (PNR) is a major healthcare challenge due to the limited regenerative capacity of the nervous system, often leading to severe functional impairments. While nerve autografts are the gold standard, their implications are constrained by issues such as donor site morbidity and limited availability, necessitating innovative alternatives like nerve guidance conduits (NGCs). However, the inherently slow nerve growth rate (∼1 mm/day) and prolonged neuroinflammation, delay recovery even with the use of passive (no-conductive) NGCs, resulting in muscle atrophy and loss of locomotor function. Electrical stimulation (ES) has the ability to enhance nerve regeneration rate by modulating the innate bioelectrical microenvironment of nerve tissue while simultaneously fostering a reparative environment through immunoregulation. In this context, electrically conductive polymer (ECP)-based biomaterials offer unique advantages for nerve repair combining their flexibility, akin to traditional plastics, and mixed ionic-electronic conductivity, similar to ionically conductive nerve tissue, as well as their biocompatibility and ease of fabrication. This review focuses on the progress, challenges, and emerging techniques for integrating ECP based NGCs with ES for functional nerve regeneration. It critically evaluates the various approaches using ECP based scaffolds, identifying gaps that have hindered clinical translation. Key challenges discussed include designing effective 3D NGCs with high electroactivity, optimizing ES modules, and better understanding of immunoregulation during nerve repair. The review also explores innovative strategies in material development and wireless, self-powered ES methods. Furthermore, it emphasizes the need for non-invasive ES delivery methods combined with hybrid ECP based neural scaffolds, highlighting future directions for advancing preclinical and clinical translation. Together, ECP based NGCs combined with ES represent a promising avenue for advancing PNR and improving patient outcomes.
Collapse
Affiliation(s)
- Rajiv Borah
- Discipline of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin 2, Ireland
- Advanced Materials and BioEngineering Research (AMBER), Centre at Trinity College Dublin and the Royal College of Surgeons in Ireland, Dublin 2, Ireland
- Trinity Centre for Biomedical Engineering, Trinity College Dublin, Dublin 2, Ireland
| | - Daniel Diez Clarke
- Discipline of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin 2, Ireland
- Trinity Centre for Biomedical Engineering, Trinity College Dublin, Dublin 2, Ireland
| | - Jnanendra Upadhyay
- Department of Physics, Dakshin Kamrup College, Kamrup, Assam, 781125, India
| | - Michael G. Monaghan
- Discipline of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin 2, Ireland
- Advanced Materials and BioEngineering Research (AMBER), Centre at Trinity College Dublin and the Royal College of Surgeons in Ireland, Dublin 2, Ireland
- Trinity Centre for Biomedical Engineering, Trinity College Dublin, Dublin 2, Ireland
- CÚRAM, Research Ireland Centre for Research in Medical Devices, University of Galway, H91 W2TY Galway, Ireland
| |
Collapse
|
4
|
Steiner RC, Buchen JT, Phillips ER, Fellin CR, Yuan X, Jariwala SH. FRESH extrusion 3D printing of type-1 collagen hydrogels photocrosslinked using ruthenium. PLoS One 2025; 20:e0317350. [PMID: 39792905 PMCID: PMC11723599 DOI: 10.1371/journal.pone.0317350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 12/18/2024] [Indexed: 01/12/2025] Open
Abstract
The extrusion bioprinting of collagen material has many applications relevant to tissue engineering and regenerative medicine. Freeform Reversible Embedding of Suspended Hydrogels (FRESH) technology is capable of 3D printing collagen material with the specifications and details needed for precise tissue guidance, a crucial requirement for effective tissue repair. While FRESH has shown repeated success and reliability for extrusion printing, the mechanical properties of completed collagen prints can be improved further by post-print crosslinking methodologies. Photoinitiator-based crosslinking methods are simple and have proven effective in strengthening protein-based materials. The ruthenium and sodium persulfate photoinitiator system (Ru(bpy)3/SPS) has been suggested as an effective crosslinking method for collagen materials. Herein, we describe the procedure our group has developed to combine extrusion-based 3D printing of type-1 collagen using FRESH technology with Ru(bpy)3/SPS photoinitiated crosslinking methods to improve the strength and stability of 3D printed collagen structures. Mechanical testing and cell biocompatibility assessments were performed to investigate the impact of Ru(bpy)3/SPS photoinitiated crosslinking and highlight the potential limitations of this method. These results demonstrate a significant improvement in the compressive strength of type-1 collagen samples as the Ru(bpy)3/SPS concentration increases. Additionally, type-1 collagen samples crosslinked with up to 1/10 mM Ru(bpy)3/SPS support PC12 cell viability over a period of 7 days. The primary limitations that were observed and described in detail in this protocol are: the FRESH slurry preparation, printing environment, extrusion printer hardware, and quality of the ruthenium reagent.
Collapse
Affiliation(s)
- Richard C. Steiner
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, United States of America
- Department of Physical Medicine and Rehabilitation, The Center for Rehabilitation Sciences Research, Uniformed Services University of Health Sciences, Bethesda, Maryland, United States of America
| | - Jack T. Buchen
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, United States of America
- Department of Physical Medicine and Rehabilitation, The Center for Rehabilitation Sciences Research, Uniformed Services University of Health Sciences, Bethesda, Maryland, United States of America
| | - Evan R. Phillips
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, United States of America
- CytoSorbents Medical Inc., Princeton, New Jersey, United States of America
| | - Christopher R. Fellin
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, United States of America
- Department of Physical Medicine and Rehabilitation, The Center for Rehabilitation Sciences Research, Uniformed Services University of Health Sciences, Bethesda, Maryland, United States of America
| | - Xiaoning Yuan
- Department of Physical Medicine and Rehabilitation, The Center for Rehabilitation Sciences Research, Uniformed Services University of Health Sciences, Bethesda, Maryland, United States of America
| | - Shailly H. Jariwala
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, United States of America
- Department of Physical Medicine and Rehabilitation, The Center for Rehabilitation Sciences Research, Uniformed Services University of Health Sciences, Bethesda, Maryland, United States of America
| |
Collapse
|
5
|
Chang K, Wu JG, Ma TL, Hsu SH, Cho KS, Yu Z, Lennikov A, Ashok A, Rajagopalan A, Chen MH, Su WF, Utheim TP, Chen DF. Bioengineering strategy to promote CNS nerve growth and regeneration via chronic glutamate signaling. Acta Biomater 2024; 190:165-177. [PMID: 39427766 PMCID: PMC11614675 DOI: 10.1016/j.actbio.2024.10.023] [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: 01/19/2024] [Revised: 09/21/2024] [Accepted: 10/16/2024] [Indexed: 10/22/2024]
Abstract
Being part of the mature mammalian central nervous system, impairments of the retina and optic nerves caused by trauma or diseases often cannot be restored. Progressive degeneration of retinal ganglion cells (RGCs) in glaucoma and other optic neuropathies gradually leads to permanent vision loss, which currently has no cure. The purpose of this study is to develop a biocompatible scaffold to support RGC survival and guide axon growth, facilitating optic nerve repair and regeneration. We here report that electrical stimulation (ES) significantly promoted neurite outgrowth and elongation from primary RGCs, mediated through glutamate receptor signaling. To mimic prolonged glutamate stimulation and facilitate sustained nerve growth, we fabricated biocompatible poly-γ-benzyl-L-glutamate (PBG) scaffolds for controlled glutamate release. These PBG scaffolds supported RGC survival and robust long-distance nerve growth in both retinal explants and isolated RGC cultures. In contrast, control polycaprolactone (PCL) scaffolds with similar physical structures showed little benefits on RGC survival or nerve growth. Moreover, PBG scaffolds promoted the differentiation and neurite outgrowth from embryonic stem cell-derived RGC progenitors. The aligned PBG scaffold drove directed nerve elongation along the fiber alignment. Transplantation of PBG-coated biocompatible conduits induced robust optic nerve regeneration in adult mice following nerve transection. Together, the findings present the exciting possibility of driving optic nerve regeneration and RGC progenitor cell differentiation by imitating ES or glutamate signaling. PBG presents a permissive biomaterial in supporting robust and directed axon growth with promising clinical applications in the future. STATEMENT OF SIGNIFICANCE: We here reported compelling findings that demonstrate the potent regenerative effects of a bioengineered scaffold incorporating poly-γ-benzyl-L-glutamate (PBG) on the optic nerve. Retinal ganglion cell (RGC) axons, which form the optic nerve, are incapable of regenerating in adulthood, posing a significant hurdle in restoring vision for patients with optic nerve diseases or injuries. Built upon the finding that electrical stimulation promotes RGC axonal growth through glutamate signaling, we developed PBG scaffolds to provide sustained glutamate stimulation and showed their exceptional effects on driving directed axonal elongation in cultured RGCs and neural progenitors, as well as supporting robust optic nerve regeneration after transection in vivo. The findings hold great promise for reversing vision loss in patients with optic nerve conditions.
Collapse
Affiliation(s)
- Karen Chang
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA; Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway
| | - Jhih-Guang Wu
- Department of Materials Science and Engineering, National Taiwan University, Taipei, Taiwan
| | - Tien-Li Ma
- Department of Materials Science and Engineering, National Taiwan University, Taipei, Taiwan
| | - Sheng-Hao Hsu
- Graduate Institute of Clinical Dentistry, School of Dentistry, National Taiwan University, Taipei, Taiwan
| | - Kin-Sang Cho
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Zicheng Yu
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA; MRC Human Genetics Unit, Institute of Genetics and Cancer, The University of Edinburgh, Edinburgh, UK
| | - Anton Lennikov
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA; Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway
| | - Ajay Ashok
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA; Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway
| | - Aishwarya Rajagopalan
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA; Department of Biological Sciences, College of Agriculture and Life Sciences, Cornell University, Ithaca, NY, USA
| | - Min-Huey Chen
- Graduate Institute of Clinical Dentistry, School of Dentistry, National Taiwan University, Taipei, Taiwan
| | - Wei-Fang Su
- Department of Materials Science and Engineering, National Taiwan University, Taipei, Taiwan
| | - Tor Paaske Utheim
- Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway; Department of Ophthalmology, Oslo University Hospital, Oslo, Norway; Department of Ophthalmology, Drammen Hospital, Drammen, Norway
| | - Dong Feng Chen
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
6
|
Kalogeropoulou M, Kracher A, Fucile P, Mihăilă SM, Moroni L. Blueprints of Architected Materials: A Guide to Metamaterial Design for Tissue Engineering. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2408082. [PMID: 39370588 PMCID: PMC11586834 DOI: 10.1002/adma.202408082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 09/09/2024] [Indexed: 10/08/2024]
Abstract
Mechanical metamaterials are rationally designed structures engineered to exhibit extraordinary properties, often surpassing those of their constituent materials. The geometry of metamaterials' building blocks, referred to as unit cells, plays an essential role in determining their macroscopic mechanical behavior. Due to their hierarchical design and remarkable properties, metamaterials hold significant potential for tissue engineering; however their implementation in the field remains limited. The major challenge hindering the broader use of metamaterials lies in the complexity of unit cell design and fabrication. To address this gap, a comprehensive guide is presented detailing the design principles of well-established metamaterials. The essential unit cell geometric parameters and design constraints, as well as their influence on mechanical behavior, are summarized highlighting essential points for effective fabrication. Moreover, the potential integration of artificial intelligence techniques is explored in meta-biomaterial design for patient- and application-specific design. Furthermore, a comprehensive overview of current applications of mechanical metamaterials is provided in tissue engineering, categorized by tissue type, thereby showcasing the versatility of different designs in matching the mechanical properties of the target tissue. This review aims to provide a valuable resource for tissue engineering researchers and aid in the broader use of metamaterials in the field.
Collapse
Affiliation(s)
- Maria Kalogeropoulou
- Complex Tissue Regeneration DepartmentMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityMaastricht6229 ERThe Netherlands
| | - Anna Kracher
- Division of PharmacologyDepartment of Pharmaceutical SciencesUtrecht Institute for Pharmaceutical SciencesUtrecht UniversityUniversiteitsweg 99Utrecht3584 CGThe Netherlands
| | - Pierpaolo Fucile
- Complex Tissue Regeneration DepartmentMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityMaastricht6229 ERThe Netherlands
| | - Silvia M. Mihăilă
- Division of PharmacologyDepartment of Pharmaceutical SciencesUtrecht Institute for Pharmaceutical SciencesUtrecht UniversityUniversiteitsweg 99Utrecht3584 CGThe Netherlands
| | - Lorenzo Moroni
- Complex Tissue Regeneration DepartmentMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityMaastricht6229 ERThe Netherlands
| |
Collapse
|
7
|
Nawrotek K, Chyb M, Gatkowska J, Rudnicka K, Michlewska S, Jóźwiak P. Effect of sodium L-lactate on bioactive properties of chitosan-hydroxyapatite/polycaprolactone conduits for peripheral nerve tissue engineering. Int J Biol Macromol 2024; 281:136254. [PMID: 39366606 DOI: 10.1016/j.ijbiomac.2024.136254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 09/26/2024] [Accepted: 10/01/2024] [Indexed: 10/06/2024]
Abstract
Biomaterials and synthetic polymers have been widely used to replicate the regenerative microenvironment of the peripheral nervous system. Chitosan-based conduits have shown promise in the regeneration of nerve injuries. However, to mimic the regenerative microenvironment, the scaffold structure should possess bioactive properties. This can be achieved by the incorporation of biomolecules (e.g., proteins, peptides) or trophic factors that should preferably be aligned and/or released with controlled kinetics to activate the process of positive axon chemotaxis. In this study, sodium L-lactate has been used to enhance the bioactive properties of chitosan-hydroxyapatite/polycaprolactone electrodeposits. Next, two methods have been developed to incorporate NGF-loaded microspheres - Method 1 involves entrapment and co-deposition of NGF-loaded microspheres, while Method 2 is based on absorption of NGF-loaded microspheres. The study shows that modification of chitosan-hydroxyapatite/polycaprolactone conduits by sodium L-lactate significantly improves their bioactive, biological, and physicochemical properties. The obtained implants are cytocompatible, enhancing the neurite regeneration process by stimulating its elongation. The absorption of NGF-loaded microspheres into the conduit structure may be considered more favorable for the stimulation of axonal elongation compared to entrapment, as it allows for trophic factor dose-dependent controlled release. The developed conduits possess properties essential for the successful treatment of peripheral nerve discontinuities.
Collapse
Affiliation(s)
- Katarzyna Nawrotek
- Lodz University of Technology, Faculty of Process and Environmental Engineering, Department of Environmental Engineering, Wolczanska 213, 93-005 Lodz, Poland; Lodz University of Technology, International Centre for Research on Innovative Bio-based Materials, 2/22 Stefanowskiego, 90-537, Poland; Warsaw University of Technology, Centre for Advanced Materials and Technology (CEZAMAT), 19 Poleczki, 02-822 Warsaw, Poland.
| | - Maciej Chyb
- University of Lodz, Faculty of Biology and Environmental Protection, Department of Molecular Microbiology, 12/16 Banacha, 90-237 Lodz, Poland; Bio-Med-Chem Doctoral School of the University of Lodz and Lodz Institutes of the Polish Academy of Sciences Banacha 12/16, 90-237 Lodz, Poland.
| | - Justyna Gatkowska
- University of Lodz, Faculty of Biology and Environmental Protection, Department of Molecular Microbiology, 12/16 Banacha, 90-237 Lodz, Poland.
| | - Karolina Rudnicka
- University of Lodz, Faculty of Biology and Environmental Protection, Department of Immunology and Infectious Biology, 90-237 Lodz, Poland.
| | - Sylwia Michlewska
- University of Lodz, Faculty of Biology and Environmental Protection, Laboratory of Microscopic Imaging and Specialized Biological Techniques, Banacha 12/16, 90-237 Lodz, Poland.
| | - Piotr Jóźwiak
- University of Lodz, Faculty of Biology and Environmental Protection, Department of Invertebrates Zoology and Hydrobiology, Banacha 12/16, 90-324 Lodz, Poland.
| |
Collapse
|
8
|
Sumam P, Kumar P R A, Parameswaran R. Aligned Fibroporous Matrix Generated from a Silver Ion and Graphene Oxide-Incorporated Ethylene Vinyl Alcohol Copolymer as a Potential Biomaterial for Peripheral Nerve Repair. ACS APPLIED BIO MATERIALS 2024; 7:6617-6630. [PMID: 39295150 DOI: 10.1021/acsabm.4c00841] [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: 09/21/2024]
Abstract
Developing an ideal nerve conduit for proper nerve regeneration still faces several challenges. The attempts to fabricate aligned substrates for neuronal growth have enhanced the hope of successful nerve regeneration. In this wok, we have attempted to generate an electrospun matrix with aligned fibers from a silver and graphene oxide-incorporated ethylene vinyl alcohol copolymer (EVAL). The presence of silver was analyzed using UV-visible spectra, XPS spectra, and ICP. Raman spectra and FTIR spectra confirmed the presence of GO. The complexation of Ag+ with - OH of EVAL enabled the generation of aligned fibers. The fiber diameter (>1 μm) provided sufficient space for forming focal adhesion by the neurites and filopodia of N2a and C6 cells, respectively. The fiber diameter enabled the neurites and filopodia of the cells to align on the fibers. The incorporation of GO has contributed to the cell-material interactions. The morphological and mechanical properties of fibers obtained in the study ensure that the EVAL-Ag-GO-0.01 matrix is a potential substrate for developing a nerve guidance conduit/nerve wrap (NGC/W).
Collapse
Affiliation(s)
- Prima Sumam
- Division of Polymeric Medical Devices, Department of Medical Devices Engineering, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Poojappura, Trivandrum, 695 012 Kerala, India
| | - Anil Kumar P R
- Division of Tissue Culture, Department of Applied Biology, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Poojappura, Trivandrum, 695 012 Kerala, India
| | - Ramesh Parameswaran
- Division of Polymeric Medical Devices, Department of Medical Devices Engineering, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Poojappura, Trivandrum, 695 012 Kerala, India
| |
Collapse
|
9
|
Wang P, Li J, Li S, Liu Y, Gong J, He S, Wu W, Tan G, Liu S. Palladium-reduced graphene oxide nanocomposites enhance neurite outgrowth and protect neurons from Ishemic stroke. Mater Today Bio 2024; 28:101184. [PMID: 39221214 PMCID: PMC11364903 DOI: 10.1016/j.mtbio.2024.101184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 07/09/2024] [Accepted: 08/04/2024] [Indexed: 09/04/2024] Open
Abstract
Currently, the construction of novel biomimetic reduced graphene oxide (RGO)-based nanocomposites to induce neurite sprouting and repair the injured neurons represents a promising strategy in promoting neuronal development or treatment of cerebral anoxia or ischemia. Here, we present an effective method for constructing palladium-reduced graphene oxide (Pd-RGO) nanocomposites by covalently bonding Pd onto RGO surfaces to enhance neurite sprouting of cultured neurons. As described, the Pd-RGO nanocomposites exhibit the required physicochemical features for better biocompatibility without impacting cell viability. Primary neurons cultured on Pd-RGO nanocomposites had significantly increased number and length of neuronal processes, including both axons and dendrites, compared with the control. Western blotting showed that Pd-RGO nanocomposites improved the expression levels of growth associate protein-43 (GAP-43), as well as β-III tubulin, Tau-1, microtubule-associated protein-2 (MAP2), four proteins that are involved in regulating neurite sprouting and outgrowth. Importantly, Pd-RGO significantly promoted neurite length and complexity under oxygen-glucose deprivation/re-oxygenation (OGD/R) conditions, an in vitro cellular model of ischemic brain damage, that closely relates to neuronal GAP-43 expression. Furthermore, using the middle cerebral artery occlusion (MCAO) model in rats, we found Pd-RGO effectively reduced the infarct area, decreased neuronal apoptosis in the brain, and improved the rats' behavioral outcomes after MCAO. Together, these results indicate the great potential of Pd-RGO nanocomposites as a novel excellent biomimetic material for neural interfacing that shed light on its applications in brain injuries.
Collapse
Affiliation(s)
- Ping Wang
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Key Laboratory of Regenerative Medicine & Guangxi Engineering Center in Biomedical Material for Tissue and Organ Regeneration &Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory of Brain Science & Guangxi Colleges and Universities Key Laboratory of Biological Molecular Medicine Research, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
| | - Jinling Li
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Key Laboratory of Regenerative Medicine & Guangxi Engineering Center in Biomedical Material for Tissue and Organ Regeneration &Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory of Brain Science & Guangxi Colleges and Universities Key Laboratory of Biological Molecular Medicine Research, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
| | - Shuntang Li
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Key Laboratory of Regenerative Medicine & Guangxi Engineering Center in Biomedical Material for Tissue and Organ Regeneration &Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory of Brain Science & Guangxi Colleges and Universities Key Laboratory of Biological Molecular Medicine Research, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
| | - Yuanyuan Liu
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Key Laboratory of Regenerative Medicine & Guangxi Engineering Center in Biomedical Material for Tissue and Organ Regeneration &Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory of Brain Science & Guangxi Colleges and Universities Key Laboratory of Biological Molecular Medicine Research, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
| | - Jiangu Gong
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Key Laboratory of Regenerative Medicine & Guangxi Engineering Center in Biomedical Material for Tissue and Organ Regeneration &Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory of Brain Science & Guangxi Colleges and Universities Key Laboratory of Biological Molecular Medicine Research, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
| | - Shipei He
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Key Laboratory of Regenerative Medicine & Guangxi Engineering Center in Biomedical Material for Tissue and Organ Regeneration &Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory of Brain Science & Guangxi Colleges and Universities Key Laboratory of Biological Molecular Medicine Research, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
| | - Weifeng Wu
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Key Laboratory of Regenerative Medicine & Guangxi Engineering Center in Biomedical Material for Tissue and Organ Regeneration &Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Medical University, Nanning, Guangxi, China
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Guohe Tan
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Key Laboratory of Regenerative Medicine & Guangxi Engineering Center in Biomedical Material for Tissue and Organ Regeneration &Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory of Brain Science & Guangxi Colleges and Universities Key Laboratory of Biological Molecular Medicine Research, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
| | - Sijia Liu
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Key Laboratory of Regenerative Medicine & Guangxi Engineering Center in Biomedical Material for Tissue and Organ Regeneration &Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory of Brain Science & Guangxi Colleges and Universities Key Laboratory of Biological Molecular Medicine Research, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
10
|
Zimmermann KS, Aman M, Harhaus L, Boecker AH. Improving outcomes in traumatic peripheral nerve injuries to the upper extremity. EUROPEAN JOURNAL OF ORTHOPAEDIC SURGERY & TRAUMATOLOGY : ORTHOPEDIE TRAUMATOLOGIE 2024; 34:3687-3697. [PMID: 37864051 DOI: 10.1007/s00590-023-03751-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 09/25/2023] [Indexed: 10/22/2023]
Abstract
Peripheral nerve lesions of the upper extremity are common and are associated with devastating limitations for the patient. Rapid and accurate diagnosis of the lesion by electroneurography, neurosonography, or even MR neurography is important for treatment planning. There are different therapeutic approaches, which may show individual differences depending on the injured nerve. If a primary nerve repair is not possible, several strategies exist to bridge the gap. These may include autologous nerve grafts, bioartificial nerve conduits, or acellular nerve allografts. Tendon and nerve transfers are also of major importance in the treatment of nerve lesions in particular with long regeneration distances. As a secondary reconstruction, in addition to tendon transfers, there is also the option for free functional muscle transfer. In amputations, the prevention of neuroma is of great importance, for which different strategies exist, such as target muscle reinnervation, regenerative peripheral nerve interface, or neurotized flaps. In this article, we give an overview of the latest methods for the therapy of peripheral nerve lesions.
Collapse
Affiliation(s)
- Kim S Zimmermann
- Department of Hand, Plastic and Reconstructive Surgery, Burn Center, BG Trauma Center Ludwigshafen, Ludwig-Guttmann-Str. 13, 67071, Ludwigshafen, Germany
- Department of Hand and Plastic Surgery, University of Heidelberg, Heidelberg, Germany
| | - Martin Aman
- Department of Hand, Plastic and Reconstructive Surgery, Burn Center, BG Trauma Center Ludwigshafen, Ludwig-Guttmann-Str. 13, 67071, Ludwigshafen, Germany
- Department of Hand and Plastic Surgery, University of Heidelberg, Heidelberg, Germany
| | - Leila Harhaus
- Department of Hand, Plastic and Reconstructive Surgery, Burn Center, BG Trauma Center Ludwigshafen, Ludwig-Guttmann-Str. 13, 67071, Ludwigshafen, Germany
- Department of Hand and Plastic Surgery, University of Heidelberg, Heidelberg, Germany
| | - Arne H Boecker
- Department of Hand, Plastic and Reconstructive Surgery, Burn Center, BG Trauma Center Ludwigshafen, Ludwig-Guttmann-Str. 13, 67071, Ludwigshafen, Germany.
- Department of Hand and Plastic Surgery, University of Heidelberg, Heidelberg, Germany.
| |
Collapse
|
11
|
Bonetti L, Scalet G. 4D fabrication of shape-changing systems for tissue engineering: state of the art and perspectives. PROGRESS IN ADDITIVE MANUFACTURING 2024; 10:1913-1943. [PMID: 40125451 PMCID: PMC11926060 DOI: 10.1007/s40964-024-00743-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 07/30/2024] [Indexed: 03/25/2025]
Abstract
In recent years, four-dimensional (4D) fabrication has emerged as a powerful technology capable of revolutionizing the field of tissue engineering. This technology represents a shift in perspective from traditional tissue engineering approaches, which generally rely on static-or passive-structures (e.g., scaffolds, constructs) unable of adapting to changes in biological environments. In contrast, 4D fabrication offers the unprecedented possibility of fabricating complex designs with spatiotemporal control over structure and function in response to environment stimuli, thus mimicking biological processes. In this review, an overview of the state of the art of 4D fabrication technology for the obtainment of cellularized constructs is presented, with a focus on shape-changing soft materials. First, the approaches to obtain cellularized constructs are introduced, also describing conventional and non-conventional fabrication techniques with their relative advantages and limitations. Next, the main families of shape-changing soft materials, namely shape-memory polymers and shape-memory hydrogels are discussed and their use in 4D fabrication in the field of tissue engineering is described. Ultimately, current challenges and proposed solutions are outlined, and valuable insights into future research directions of 4D fabrication for tissue engineering are provided to disclose its full potential.
Collapse
Affiliation(s)
- Lorenzo Bonetti
- Department of Civil Engineering and Architecture (DICAr), University of Pavia, Via Ferrata 3, 27100 Pavia, Italy
| | - Giulia Scalet
- Department of Civil Engineering and Architecture (DICAr), University of Pavia, Via Ferrata 3, 27100 Pavia, Italy
| |
Collapse
|
12
|
Gao Y, Wang X, Fan C. Advances in graphene-based 2D materials for tendon, nerve, bone/cartilage regeneration and biomedicine. iScience 2024; 27:110214. [PMID: 39040049 PMCID: PMC11261022 DOI: 10.1016/j.isci.2024.110214] [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] [Indexed: 07/24/2024] Open
Abstract
Two-dimensional (2D) materials, especially graphene-based materials, have important implications for tissue regeneration and biomedicine due to their large surface area, transport properties, ease of functionalization, biocompatibility, and adsorption capacity. Despite remarkable progress in the field of tissue regeneration and biomedicine, there are still problems such as unclear long-term stability, lack of in vivo experimental data, and detection accuracy. This paper reviews recent applications of graphene-based materials in tissue regeneration and biomedicine and discusses current issues and prospects for the development of graphene-based materials with respect to promoting the regeneration of tendons, neuronal cells, bone, chondrocytes, blood vessels, and skin, as well as applications in sensing, detection, anti-microbial activity, and targeted drug delivery.
Collapse
Affiliation(s)
- Yuxin Gao
- Department of Orthopedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, China
| | - Xu Wang
- Department of Orthopedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Center for Orthopaedics, Shanghai, China
- Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai, China
| | - Cunyi Fan
- Department of Orthopedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Center for Orthopaedics, Shanghai, China
- Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai, China
| |
Collapse
|
13
|
Li L, Chu Z, Li S, Zheng T, Wei S, Zhao Y, Liu P, Lu Q. BDNF-loaded chitosan-based mimetic mussel polymer conduits for repair of peripheral nerve injury. Front Cell Dev Biol 2024; 12:1431558. [PMID: 39011392 PMCID: PMC11246889 DOI: 10.3389/fcell.2024.1431558] [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: 05/12/2024] [Accepted: 06/11/2024] [Indexed: 07/17/2024] Open
Abstract
Care for patients with peripheral nerve injury is multifaceted, as traditional methods are not devoid of limitations. Although the utilization of neural conduits shows promise as a therapeutic modality for peripheral nerve injury, its efficacy as a standalone intervention is limited. Hence, there is a pressing need to investigate a composite multifunctional neural conduit as an alternative treatment for peripheral nerve injury. In this study, a BDNF-loaded chitosan-based mimetic mussel polymer conduit was prepared. Its unique adhesion characteristics allow it to be suture-free, improve the microenvironment of the injury site, and have good antibacterial properties. Researchers utilized a rat sciatic nerve injury model to evaluate the progression of nerve regeneration at the 12-week postoperative stage. The findings of this study indicate that the chitosan-based mimetic mussel polymer conduit loaded with BDNF had a substantial positive effect on myelination and axon outgrowth. The observed impact demonstrated a favorable outcome in terms of sciatic nerve regeneration and subsequent functional restoration in rats with a 15-mm gap. Hence, this approach is promising for nerve tissue regeneration during peripheral nerve injury.
Collapse
Affiliation(s)
- Lei Li
- Department of Adult Joint Reconstructive Surgery, Beijing Jishuitan Hospital, Capital Medical University, Beijing, China
| | - Ziyue Chu
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Shihao Li
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Tong Zheng
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Shusheng Wei
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Yunpeng Zhao
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Peilai Liu
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Qunshan Lu
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, Shandong, China
| |
Collapse
|
14
|
Pinzon-Herrera L, Magness J, Apodaca-Reyes H, Sanchez J, Almodovar J. Surface Modification of Nerve Guide Conduits with ECM Coatings and Investigating Their Impact on Schwann Cell Response. Adv Healthc Mater 2024; 13:e2304103. [PMID: 38400540 DOI: 10.1002/adhm.202304103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 02/06/2024] [Indexed: 02/25/2024]
Abstract
In this study, layer-by-layer coatings composed of heparin and collagen are proposed as an extracellular mimetic environment on nerve guide conduits (NGC) to modulate the behavior of Schwann cells (hSCs). The authors evaluated the stability, degradation over time, and bioactivity of six bilayers of heparin/collagen layer-by-layer coatings, denoted as (HEP/COL)6. The stability study reveals that (HEP/COL)6 is stable after incubating the coatings in cell media for up to 21 days. The impact of (HEP/COL)6 on hSCs viability, protein expression, and migration is evaluated. These assays show that hSCs cultured in (HEP/COL)6 have enhanced protein expression and migration. This condition increases the expression of neurotrophic and immunomodulatory factors up to 1.5-fold compared to controls, and hSCs migrated 1.34 times faster than in the uncoated surfaces. Finally, (HEP/COL)6 is also applied to a commercial collagen-based NGC, NeuraGen, and hSC viability and adhesion are studied after 6 days of culture. The morphology of NeuraGen is not altered by the presence of (HEP/COL)6 and a nearly 170% increase of the cell viability is observed in the condition where NeuraGen is used with (HEP/COL)6. Additionally, cell adhesion on the coated samples is successfully demonstrated. This work demonstrates the reparative enhancing potential of extracellular mimetic coatings.
Collapse
Affiliation(s)
- Luis Pinzon-Herrera
- Ralph E. Martin Department of Chemical Engineering, University of Arkansas, 3202 Bell Engineering Center, Fayetteville, AR, 72701, USA
- Department of Chemical, Biochemical, and Environmental Engineering, University of Maryland Baltimore County, 1000 Hilltop Circle, Baltimore, MD, 21250, USA
| | - John Magness
- Ralph E. Martin Department of Chemical Engineering, University of Arkansas, 3202 Bell Engineering Center, Fayetteville, AR, 72701, USA
| | - Hector Apodaca-Reyes
- Ralph E. Martin Department of Chemical Engineering, University of Arkansas, 3202 Bell Engineering Center, Fayetteville, AR, 72701, USA
| | - Jesus Sanchez
- Science & Mathematics Division, Northwest Arkansas Community College, 1418 Burns Hall, Bentonville, AR, 72712, USA
| | - Jorge Almodovar
- Ralph E. Martin Department of Chemical Engineering, University of Arkansas, 3202 Bell Engineering Center, Fayetteville, AR, 72701, USA
- Department of Chemical, Biochemical, and Environmental Engineering, University of Maryland Baltimore County, 1000 Hilltop Circle, Baltimore, MD, 21250, USA
| |
Collapse
|
15
|
Sun R, Lang Y, Chang MW, Zhao M, Li C, Liu S, Wang B. Leveraging Oriented Lateral Walls of Nerve Guidance Conduit with Core-Shell MWCNTs Fibers for Peripheral Nerve Regeneration. Adv Healthc Mater 2024; 13:e2303867. [PMID: 38258406 DOI: 10.1002/adhm.202303867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Indexed: 01/24/2024]
Abstract
Peripheral nerve regeneration and functional recovery rely on the chemical, physical, and structural properties of nerve guidance conduits (NGCs). However, the limited support for long-distance nerve regeneration and axonal guidance has hindered the widespread use of NGCs. This study introduces a novel nerve guidance conduit with oriented lateral walls, incorporating multi-walled carbon nanotubes (MWCNTs) within core-shell fibers prepared in a single step using a modified electrohydrodynamic (EHD) printing technique to promote peripheral nerve regeneration. The structured conduit demonstrated exceptional stability, mechanical properties, and biocompatibility, significantly enhancing the functionality of NGCs. In vitro cell studies revealed that RSC96 cells adhered and proliferated effectively along the oriented fibers, demonstrating a favorable response to the distinctive architectures and properties. Subsequently, a rat sciatic nerve injury model demonstrated effective efficacy in promoting peripheral nerve regeneration and functional recovery. Tissue analysis and functional testing highlighted the significant impact of MWCNT concentration in enhancing peripheral nerve regeneration and confirming well-matured aligned axonal growth, muscle recovery, and higher densities of myelinated axons. These findings demonstrate the potential of oriented lateral architectures with coaxial MWCNT fibers as a promising approach to support long-distance regeneration and encourage directional nerve growth for peripheral nerve repair in clinical applications.
Collapse
Affiliation(s)
- Renyuan Sun
- State Key Laboratory of Reliability and Intelligence of Electrical Equipment, Tianjin Key Laboratory of Bio-Electromagnetic and Neural Engineering, Hebei Key Laboratory of Bioelectromagnetics and Neuroengineering, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin, 300132, China
| | - Yuna Lang
- State Key Laboratory of Reliability and Intelligence of Electrical Equipment, Tianjin Key Laboratory of Bio-Electromagnetic and Neural Engineering, Hebei Key Laboratory of Bioelectromagnetics and Neuroengineering, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin, 300132, China
| | - Ming-Wei Chang
- Nanotechnology and Integrated Bioengineering Centre, University of Ulster, Belfast, BT15 1AP, UK
| | - Mingkang Zhao
- State Key Laboratory of Reliability and Intelligence of Electrical Equipment, Tianjin Key Laboratory of Bio-Electromagnetic and Neural Engineering, Hebei Key Laboratory of Bioelectromagnetics and Neuroengineering, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin, 300132, China
| | - Chao Li
- State Key Laboratory of Reliability and Intelligence of Electrical Equipment, Tianjin Key Laboratory of Bio-Electromagnetic and Neural Engineering, Hebei Key Laboratory of Bioelectromagnetics and Neuroengineering, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin, 300132, China
| | - Shiheng Liu
- State Key Laboratory of Reliability and Intelligence of Electrical Equipment, Tianjin Key Laboratory of Bio-Electromagnetic and Neural Engineering, Hebei Key Laboratory of Bioelectromagnetics and Neuroengineering, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin, 300132, China
| | - Baolin Wang
- State Key Laboratory of Reliability and Intelligence of Electrical Equipment, Tianjin Key Laboratory of Bio-Electromagnetic and Neural Engineering, Hebei Key Laboratory of Bioelectromagnetics and Neuroengineering, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin, 300132, China
| |
Collapse
|
16
|
Ozcicek I, Aysit N, Balcikanli Z, Ayturk NU, Aydeger A, Baydas G, Aydin MS, Altintas E, Erim UC. Development of BDNF/NGF/IKVAV Peptide Modified and Gold Nanoparticle Conductive PCL/PLGA Nerve Guidance Conduit for Regeneration of the Rat Spinal Cord Injury. Macromol Biosci 2024; 24:e2300453. [PMID: 38224015 DOI: 10.1002/mabi.202300453] [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: 10/04/2023] [Revised: 12/22/2023] [Indexed: 01/16/2024]
Abstract
Spinal cord injuries are very common worldwide, leading to permanent nerve function loss with devastating effects in the affected patients. The challenges and inadequate results in the current clinical treatments are leading scientists to innovative neural regenerative research. Advances in nanoscience and neural tissue engineering have opened new avenues for spinal cord injury (SCI) treatment. In order for designed nerve guidance conduit (NGC) to be functionally useful, it must have ideal scaffold properties and topographic features that promote the linear orientation of damaged axons. In this study, it is aimed to develop channeled polycaprolactone (PCL)/Poly-D,L-lactic-co-glycolic acid (PLGA) hybrid film scaffolds, modify their surfaces by IKVAV pentapeptide/gold nanoparticles (AuNPs) or polypyrrole (PPy) and investigate the behavior of motor neurons on the designed scaffold surfaces in vitro under static/bioreactor conditions. Their potential to promote neural regeneration after implantation into the rat SCI by shaping the film scaffolds modified with neural factors into a tubular form is also examined. It is shown that channeled groups decorated with AuNPs highly promote neurite orientation under bioreactor conditions and also the developed optimal NGC (PCL/PLGA G1-IKVAV/BDNF/NGF-AuNP50) highly regenerates SCI. The results indicate that the designed scaffold can be an ideal candidate for spinal cord regeneration.
Collapse
Affiliation(s)
- Ilyas Ozcicek
- Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, 34810, Turkey
- Department of Medical Biology, School of Medicine, Istanbul Medipol University, Istanbul, 34815, Turkey
| | - Nese Aysit
- Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, 34810, Turkey
- Department of Medical Biology, School of Medicine, Istanbul Medipol University, Istanbul, 34815, Turkey
| | - Zeynep Balcikanli
- Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, 34810, Turkey
| | - Nilufer Ulas Ayturk
- Department of Histology and Embryology, Faculty of Medicine, Çanakkale Onsekiz Mart University, Canakkale, 17020, Turkey
| | - Asel Aydeger
- Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, 34810, Turkey
- Graduate School of Health Sciences, Istanbul Medipol University, Istanbul, 34815, Turkey
| | - Gulsena Baydas
- Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, 34810, Turkey
- Graduate School of Health Sciences, Istanbul Medipol University, Istanbul, 34815, Turkey
- Department of Physiology, School of Medicine, Istanbul Medipol University, Istanbul, 34815, Turkey
| | - Mehmet Serif Aydin
- Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, 34810, Turkey
| | - Esra Altintas
- Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, 34810, Turkey
- Graduate School of Health Sciences, Istanbul Medipol University, Istanbul, 34815, Turkey
| | - Umit Can Erim
- Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, 34810, Turkey
- Department of Analytical Chemistry, School of Pharmacy, Istanbul Medipol University, Istanbul, 34815, Turkey
| |
Collapse
|
17
|
Das JM, Upadhyay J, Monaghan MG, Borah R. Impact of the Reduction Time-Dependent Electrical Conductivity of Graphene Nanoplatelet-Coated Aligned Bombyx mori Silk Scaffolds on Electrically Stimulated Axonal Growth. ACS APPLIED BIO MATERIALS 2024; 7:2389-2401. [PMID: 38502100 PMCID: PMC11022174 DOI: 10.1021/acsabm.4c00052] [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: 01/12/2024] [Revised: 03/02/2024] [Accepted: 03/05/2024] [Indexed: 03/20/2024]
Abstract
Graphene-based nanomaterials, renowned for their outstanding electrical conductivity, have been extensively studied as electroconductive biomaterials (ECBs) for electrically stimulated tissue regeneration. However, using eco-friendly reducing agents like l-ascorbic acid (l-Aa) can result in lower conductive properties in these ECBs, limiting their full potential for smooth charge transfer in living tissues. Moreover, creating a flexible biomaterial scaffold using these materials that accurately mimics a specific tissue microarchitecture, such as nerves, poses additional challenges. To address these issues, this study developed a microfibrous scaffold of Bombyx mori (Bm) silk fibroin uniformly coated with graphene nanoplatelets (GNPs) through a vacuum coating method. The scaffold's electrical conductivity was optimized by varying the reduction period using l-Aa. The research systematically investigated how different reduction periods impact scaffold properties, focusing on electrical conductivity and its significance on electrically stimulated axonal growth in PC12 cells. Results showed that a 48 h reduction significantly increased surface electrical conductivity by 100-1000 times compared to a shorter or no reduction process. l-Aa contributed to stabilizing the reduced GNPs, demonstrated by a slow degradation profile and sustained conductivity even after 60 days in a proteolytic environment. β (III) tubulin immunostaining of PC12 cells on varied silk:GNP scaffolds under pulsed electrical stimulation (ES, 50 Hz frequency, 1 ms pulse width, and amplitudes of 100 and 300 mV/cm) demonstrates accelerated axonal growth on scaffolds exhibiting higher conductivity. This is supported by upregulated intracellular Ca2+ dynamics immediately after ES on the scaffolds with higher conductivity, subjected to a prolonged reduction period. The study showcases a sustainable reduction approach using l-Aa in combination with natural Bm silk fibroin to create a highly conductive, mechanically robust, and stable silk:GNP-based aligned fibrous scaffold. These scaffolds hold promise for functional regeneration in electrically excitable tissues such as nerves, cardiac tissue, and muscles.
Collapse
Affiliation(s)
- Jitu Mani Das
- Life
Sciences Division, Institute of Advanced
Study in Science & Technology, Guwahati 781035, India
| | - Jnanendra Upadhyay
- Department
of Physics, Dakshin Kamrup College, Kamrup, Mirza, Assam 781125, India
| | - Michael G. Monaghan
- Department
of Mechanical, Manufacturing and Biomedical Engineering, Trinity College Dublin, Dublin D2, Ireland
- Advanced
Materials and BioEngineering Research (AMBER), Centre at Trinity College Dublin and the Royal College of Surgeons
in Ireland, Dublin D2, Ireland
- Trinity
Centre for Biomedical Engineering, Trinity
College Dublin, Dublin D2, Ireland
- CÚRAM,
Centre for Research in Medical Devices, National University of Ireland, Galway H91 W2TY, Ireland
| | - Rajiv Borah
- Life
Sciences Division, Institute of Advanced
Study in Science & Technology, Guwahati 781035, India
- Department
of Mechanical, Manufacturing and Biomedical Engineering, Trinity College Dublin, Dublin D2, Ireland
- Advanced
Materials and BioEngineering Research (AMBER), Centre at Trinity College Dublin and the Royal College of Surgeons
in Ireland, Dublin D2, Ireland
- Trinity
Centre for Biomedical Engineering, Trinity
College Dublin, Dublin D2, Ireland
| |
Collapse
|
18
|
Loukelis K, Koutsomarkos N, Mikos AG, Chatzinikolaidou M. Advances in 3D bioprinting for regenerative medicine applications. Regen Biomater 2024; 11:rbae033. [PMID: 38845855 PMCID: PMC11153344 DOI: 10.1093/rb/rbae033] [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: 12/03/2023] [Revised: 03/13/2024] [Accepted: 03/17/2024] [Indexed: 06/09/2024] Open
Abstract
Biofabrication techniques allow for the construction of biocompatible and biofunctional structures composed from biomaterials, cells and biomolecules. Bioprinting is an emerging 3D printing method which utilizes biomaterial-based mixtures with cells and other biological constituents into printable suspensions known as bioinks. Coupled with automated design protocols and based on different modes for droplet deposition, 3D bioprinters are able to fabricate hydrogel-based objects with specific architecture and geometrical properties, providing the necessary environment that promotes cell growth and directs cell differentiation towards application-related lineages. For the preparation of such bioinks, various water-soluble biomaterials have been employed, including natural and synthetic biopolymers, and inorganic materials. Bioprinted constructs are considered to be one of the most promising avenues in regenerative medicine due to their native organ biomimicry. For a successful application, the bioprinted constructs should meet particular criteria such as optimal biological response, mechanical properties similar to the target tissue, high levels of reproducibility and printing fidelity, but also increased upscaling capability. In this review, we highlight the most recent advances in bioprinting, focusing on the regeneration of various tissues including bone, cartilage, cardiovascular, neural, skin and other organs such as liver, kidney, pancreas and lungs. We discuss the rapidly developing co-culture bioprinting systems used to resemble the complexity of tissues and organs and the crosstalk between various cell populations towards regeneration. Moreover, we report on the basic physical principles governing 3D bioprinting, and the ideal bioink properties based on the biomaterials' regenerative potential. We examine and critically discuss the present status of 3D bioprinting regarding its applicability and current limitations that need to be overcome to establish it at the forefront of artificial organ production and transplantation.
Collapse
Affiliation(s)
- Konstantinos Loukelis
- Department of Materials Science and Technology, University of Crete, Heraklion 70013, Greece
| | - Nikos Koutsomarkos
- Department of Materials Science and Technology, University of Crete, Heraklion 70013, Greece
| | - Antonios G Mikos
- Department of Bioengineering, Rice University, Houston, TX 77030, USA
| | - Maria Chatzinikolaidou
- Department of Materials Science and Technology, University of Crete, Heraklion 70013, Greece
- Institute of Electronic Structure and Laser (IESL), Foundation for Research and Technology Hellas (FORTH), Heraklion 70013, Greece
| |
Collapse
|
19
|
Rochkind S, Sirota S, Kushnir A. Nerve Reconstruction Using ActiGraft Blood Clot in Rabbit Acute Peripheral Injury Model: Preliminary Study. Bioengineering (Basel) 2024; 11:298. [PMID: 38671720 PMCID: PMC11047591 DOI: 10.3390/bioengineering11040298] [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: 02/05/2024] [Revised: 03/16/2024] [Accepted: 03/19/2024] [Indexed: 04/28/2024] Open
Abstract
This preliminary study aimed to investigate an ActiGraft blood clot implant (RedDress Ltd., Pardes-Hanna, Israel) attempting to treat and induce the regeneration of a completely injured peripheral nerve with a massive loss defect. The tibial portion of the sciatic nerve in 11 rabbits was transected, and a 25 mm nerve gap was reconnected using a collagen tube. A comparison was performed between the treatment group (eight rabbits; reconnection using a tube filled with ActiGraft blood clot) and the control group (three rabbits; gap reconnection using an empty tube). The post-operative follow-up period lasted 18 weeks and included electrophysiological and histochemical assessments. The pathological severity score was high in the tube cross sections of the control group (1.33) compared to the ActiGraft blood clot treatment group (0.63). Morphometric analysis showed a higher percentage of the positive myelin basic protein (MBP) stained area in the ActiGraft blood clot group (19.57%) versus the control group (3.67%). These differences were not statistically significant due to the small group sizes and the large intra-group variability. The results of this preliminary study suggest that the application of an ActiGraft blood clot (into the collagen tube) can enable nerve recovery. However, a future study using a larger animal group is required to achieve objective statistical results.
Collapse
Affiliation(s)
- Shimon Rochkind
- Sackler School of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Sharon Sirota
- RedDress Ltd., Pardes Hana 3701142, Israel; (S.S.); (A.K.)
| | - Alon Kushnir
- RedDress Ltd., Pardes Hana 3701142, Israel; (S.S.); (A.K.)
| |
Collapse
|
20
|
Hosseini SM, Borys B, Karimi-Abdolrezaee S. Neural stem cell therapies for spinal cord injury repair: an update on recent preclinical and clinical advances. Brain 2024; 147:766-793. [PMID: 37975820 DOI: 10.1093/brain/awad392] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 10/22/2023] [Accepted: 11/02/2023] [Indexed: 11/19/2023] Open
Abstract
Traumatic spinal cord injury (SCI) is a leading cause of lifelong disabilities. Permanent sensory, motor and autonomic impairments after SCI are substantially attributed to degeneration of spinal cord neurons and axons, and disintegration of neural network. To date, minimal regenerative treatments are available for SCI with an unmet need for new therapies to reconstruct the damaged spinal cord neuron-glia network and restore connectivity with the supraspinal pathways. Multipotent neural precursor cells (NPCs) have a unique capacity to generate neurons, oligodendrocytes and astrocytes. Due to this capacity, NPCs have been an attractive cell source for cellular therapies for SCI. Transplantation of NPCs has been extensively tested in preclinical models of SCI in the past two decades. These studies have identified opportunities and challenges associated with NPC therapies. While NPCs have the potential to promote neuroregeneration through various mechanisms, their low long-term survival and integration within the host injured spinal cord limit the functional benefits of NPC-based therapies for SCI. To address this challenge, combinatorial strategies have been developed to optimize the outcomes of NPC therapies by enriching SCI microenvironment through biomaterials, genetic and pharmacological therapies. In this review, we will provide an in-depth discussion on recent advances in preclinical NPC-based therapies for SCI. We will discuss modes of actions and mechanism by which engrafted NPCs contribute to the repair process and functional recovery. We will also provide an update on current clinical trials and new technologies that have facilitated preparation of medical-grade human NPCs suitable for transplantation in clinical studies.
Collapse
Affiliation(s)
- Seyed Mojtaba Hosseini
- Department of Physiology and Pathophysiology, Spinal Cord Research Centre, Rady Faculty of Health Sciences, University of Manitoba Winnipeg, Manitoba R3E 0J9, Canada
- Manitoba Multiple Sclerosis Research Center, Winnipeg, Manitoba R3E 0J9, Canada
| | - Ben Borys
- Department of Physiology and Pathophysiology, Spinal Cord Research Centre, Rady Faculty of Health Sciences, University of Manitoba Winnipeg, Manitoba R3E 0J9, Canada
| | - Soheila Karimi-Abdolrezaee
- Department of Physiology and Pathophysiology, Spinal Cord Research Centre, Rady Faculty of Health Sciences, University of Manitoba Winnipeg, Manitoba R3E 0J9, Canada
- Manitoba Multiple Sclerosis Research Center, Winnipeg, Manitoba R3E 0J9, Canada
- Children's Hospital Research Institute of Manitoba, Winnipeg, Manitoba R3E 3P4, Canada
| |
Collapse
|
21
|
Lin Y, Wang J, Liu X, Hu Y, Zhang Y, Jiang F. Synthesis, biological activity evaluation and mechanism analysis of new ganglioside GM3 derivatives as potential agents for nervous functional recovery. Eur J Med Chem 2024; 266:116108. [PMID: 38218125 DOI: 10.1016/j.ejmech.2023.116108] [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: 11/22/2023] [Revised: 12/27/2023] [Accepted: 12/27/2023] [Indexed: 01/15/2024]
Abstract
Neuronal regenerative ability is vital for the treatment of neurodegenerative diseases and neuronal injuries. Recent studies have revealed that Ganglioside GM3 and its derivatives may possess potential neuroprotective and neurite growth-promoting activities. Herein, six GM3 derivatives were synthesized and evaluated their potential neuroprotective effects and neurite outgrowth-promoting activities on a cellular model of Parkinson's disease and primary nerve cells. Amongst these derivatives, derivatives N-14 and 2C-12 demonstrated neuroprotective effects in the MPP + model in SH-SY5Y cells. 2C-12 combined with NGF (nerve growth factor) induced effecially neurite growth in primary nerve cells. Further action mechanism revealed that derivative 2C-12 exerts neuroprotective effects by regulating the Wnt signaling pathway, specifically involving the Wnt7b gene. Overall, this study establishes a foundation for further exploration and development of GM3 derivatives with neurotherapeutic potential.
Collapse
Affiliation(s)
- Yingjun Lin
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Juntao Wang
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Xiangwen Liu
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Yangfan Hu
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Yong Zhang
- School of Science and Biotechnology, Shanghai Jiao Tong University, No. 800 Dongchuan Rd., Minhang District, Shanghai, 200240, China
| | - Faqin Jiang
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
22
|
Zou X, Dong Y, Alhaskawi A, Zhou H, Ezzi SHA, Kota VG, Abdulla MHAH, Abdalbary SA, Lu H, Wang C. Techniques and graft materials for repairing peripheral nerve defects. Front Neurol 2024; 14:1307883. [PMID: 38318237 PMCID: PMC10839026 DOI: 10.3389/fneur.2023.1307883] [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: 10/05/2023] [Accepted: 12/15/2023] [Indexed: 02/07/2024] Open
Abstract
Peripheral nerve defects refer to damage or destruction occurring in the peripheral nervous system, typically affecting the limbs and face. The current primary approaches to address peripheral nerve defects involve the utilization of autologous nerve transplants or the transplantation of artificial material. Nevertheless, these methods possess certain limitations, such as inadequate availability of donor nerve or unsatisfactory regenerative outcomes post-transplantation. Biomaterials have been extensively studied as an alternative approach to promote the repair of peripheral neve defects. These biomaterials include both natural and synthetic materials. Natural materials consist of collagen, chitosan, and silk, while synthetic materials consist of polyurethane, polylactic acid, and polycaprolactone. Recently, several new neural repair technologies have also been developed, such as nerve regeneration bridging technology, electrical stimulation technology, and stem cell therapy technology. Overall, biomaterials and new neural repair technologies provide new methods and opportunities for repairing peripheral nerve defects. However, these methods still require further research and development to enhance their effectiveness and feasibility.
Collapse
Affiliation(s)
- Xiaodi Zou
- Department of Orthopedics, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Yanzhao Dong
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Ahmad Alhaskawi
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Haiying Zhou
- Faculty of Medicine, The Chinese University of Hong Kong School of Biomedical Science, Shatin, China
| | | | | | | | - Sahar Ahmed Abdalbary
- Department of Orthopedic Physical Therapy, Faculty of Physical Therapy, Nahda University in Beni Suef, Beni Suef, Egypt
| | - Hui Lu
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University, Hangzhou, China
- Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Zhejiang University, Hangzhou, China
| | - Changxin Wang
- Department of Orthopedics, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
23
|
Xiao B, Feturi F, Su AJA, Van der Merwe Y, Barnett JM, Jabbari K, Khatter NJ, Li B, Katzel EB, Venkataramanan R, Solari MG, Wagner WR, Steketee MB, Simons DJ, Washington KM. Nerve Wrap for Local Delivery of FK506/Tacrolimus Accelerates Nerve Regeneration. Int J Mol Sci 2024; 25:847. [PMID: 38255920 PMCID: PMC10815243 DOI: 10.3390/ijms25020847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 01/03/2024] [Accepted: 01/05/2024] [Indexed: 01/24/2024] Open
Abstract
Peripheral nerve injuries (PNIs) occur frequently and can lead to devastating and permanent sensory and motor function disabilities. Systemic tacrolimus (FK506) administration has been shown to hasten recovery and improve functional outcomes after PNI repair. Unfortunately, high systemic levels of FK506 can result in adverse side effects. The localized administration of FK506 could provide the neuroregenerative benefits of FK506 while avoiding systemic, off-target side effects. This study investigates the utility of a novel FK506-impregnated polyester urethane urea (PEUU) nerve wrap to treat PNI in a previously validated rat infraorbital nerve (ION) transection and repair model. ION function was assessed by microelectrode recordings of trigeminal ganglion cells responding to controlled vibrissae deflections in ION-transected and -repaired animals, with and without the nerve wrap. Peristimulus time histograms (PSTHs) having 1 ms bins were constructed from spike times of individual single units. Responses to stimulus onsets (ON responses) were calculated during a 20 ms period beginning 1 ms after deflection onset; this epoch captures the initial, transient phase of the whisker-evoked response. Compared to no-wrap controls, rats with PEUU-FK506 wraps functionally recovered earlier, displaying larger response magnitudes. With nerve wrap treatment, FK506 blood levels up to six weeks were measured nearly at the limit of quantification (LOQ ≥ 2.0 ng/mL); whereas the drug concentrations within the ION and muscle were much higher, demonstrating the local delivery of FK506 to treat PNI. An immunohistological assessment of ION showed increased myelin expression for animals assigned to neurorrhaphy with PEUU-FK506 treatment compared to untreated or systemic-FK506-treated animals, suggesting that improved PNI outcomes using PEUU-FK506 is mediated by the modulation of Schwann cell activity.
Collapse
Affiliation(s)
- Bo Xiao
- Department of Plastic Surgery, University of Pittsburgh School of Medicine, Veterans Administration Healthcare System, Pittsburgh, PA 15213, USA; (B.X.); (F.F.)
| | - Firuz Feturi
- Department of Plastic Surgery, University of Pittsburgh School of Medicine, Veterans Administration Healthcare System, Pittsburgh, PA 15213, USA; (B.X.); (F.F.)
| | - An-Jey A. Su
- Department of Plastic Surgery, University of Pittsburgh School of Medicine, Veterans Administration Healthcare System, Pittsburgh, PA 15213, USA; (B.X.); (F.F.)
- Department of Surgery, Division of Plastic Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | | | - Joshua M. Barnett
- Department of Plastic Surgery, University of Pittsburgh School of Medicine, Veterans Administration Healthcare System, Pittsburgh, PA 15213, USA; (B.X.); (F.F.)
| | - Kayvon Jabbari
- Department of Surgery, Division of Plastic Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Neil J. Khatter
- Department of Surgery, Division of Plastic Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Bing Li
- Department of Surgery, Division of Plastic Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Evan B. Katzel
- Department of Plastic Surgery, University of Pittsburgh School of Medicine, Veterans Administration Healthcare System, Pittsburgh, PA 15213, USA; (B.X.); (F.F.)
| | | | - Mario G. Solari
- Department of Plastic Surgery, University of Pittsburgh School of Medicine, Veterans Administration Healthcare System, Pittsburgh, PA 15213, USA; (B.X.); (F.F.)
| | - William R. Wagner
- McGowan Institute for Regenerative Medicine, Pittsburgh, PA 15219, USA; (W.R.W.); (D.J.S.)
| | - Michael B. Steketee
- Department of Ophthalmology, University of California, San Diego, CA 90095, USA
- McGowan Institute for Regenerative Medicine, Pittsburgh, PA 15219, USA; (W.R.W.); (D.J.S.)
| | - Daniel J. Simons
- McGowan Institute for Regenerative Medicine, Pittsburgh, PA 15219, USA; (W.R.W.); (D.J.S.)
| | - Kia M. Washington
- Department of Plastic Surgery, University of Pittsburgh School of Medicine, Veterans Administration Healthcare System, Pittsburgh, PA 15213, USA; (B.X.); (F.F.)
- Department of Surgery, Division of Plastic Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- McGowan Institute for Regenerative Medicine, Pittsburgh, PA 15219, USA; (W.R.W.); (D.J.S.)
| |
Collapse
|
24
|
Marques-Almeida T, Lanceros-Mendez S, Ribeiro C. State of the Art and Current Challenges on Electroactive Biomaterials and Strategies for Neural Tissue Regeneration. Adv Healthc Mater 2024; 13:e2301494. [PMID: 37843074 DOI: 10.1002/adhm.202301494] [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: 05/09/2023] [Revised: 09/22/2023] [Indexed: 10/17/2023]
Abstract
The loss or failure of an organ/tissue stands as one of the healthcare system's most prevalent, devastating, and costly challenges. Strategies for neural tissue repair and regeneration have received significant attention due to their particularly strong impact on patients' well-being. Many research efforts are dedicated not only to control the disease symptoms but also to find solutions to repair the damaged tissues. Neural tissue engineering (TE) plays a key role in addressing this problem and significant efforts are being carried out to develop strategies for neural repair treatment. In the last years, active materials allowing to tune cell-materials interaction are being increasingly used, representing a recent paradigm in TE applications. Among the most important stimuli influencing cell behavior are the electrical and mechanical ones. In this way, materials with the ability to provide this kind of stimuli to the neural cells seem to be appropriate to support neural TE. In this scope, this review summarizes the different biomaterials types used for neural TE, highlighting the relevance of using active biomaterials and electrical stimulation. Furthermore, this review provides not only a compilation of the most relevant studies and results but also strategies for novel and more biomimetic approaches for neural TE.
Collapse
Affiliation(s)
- Teresa Marques-Almeida
- Physics Centre of Minho and Porto Universities (CF-UM-UP), University of Minho, Braga, 4710-057, Portugal
- LaPMET - Laboratory of Physics for Materials and Emergent Technologies, University of Minho, Braga, 4710-057, Portugal
| | - Senentxu Lanceros-Mendez
- Physics Centre of Minho and Porto Universities (CF-UM-UP), University of Minho, Braga, 4710-057, Portugal
- LaPMET - Laboratory of Physics for Materials and Emergent Technologies, University of Minho, Braga, 4710-057, Portugal
- BCMaterials, Basque Center for Materials, Applications and Nanostructures, UPV/EHU Science Park, Leioa, 48940, Spain
- IKERBASQUE, Basque Foundation for Science, Bilbao, 48009, Spain
| | - Clarisse Ribeiro
- Physics Centre of Minho and Porto Universities (CF-UM-UP), University of Minho, Braga, 4710-057, Portugal
- LaPMET - Laboratory of Physics for Materials and Emergent Technologies, University of Minho, Braga, 4710-057, Portugal
| |
Collapse
|
25
|
Choinière W, Petit È, Monfette V, Pelletier S, Godbout-Lavoie C, Lauzon MA. Dynamic three-dimensional coculture model: The future of tissue engineering applied to the peripheral nervous system. J Tissue Eng 2024; 15:20417314241265916. [PMID: 39139455 PMCID: PMC11320398 DOI: 10.1177/20417314241265916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 06/18/2024] [Indexed: 08/15/2024] Open
Abstract
Traumatic injuries to the peripheral nervous system (PNI) can lead to severe consequences such as paralysis. Unfortunately, current treatments rarely allow for satisfactory functional recovery. The high healthcare costs associated with PNS injuries, worker disability, and low patient satisfaction press for alternative solutions that surpass current standards. For the treatment of injuries with a deficit of less than 30 mm to bridge, the use of synthetic nerve conduits (NGC) is favored. However, to develop such promising therapeutic strategies, in vitro models that more faithfully mimic nerve physiology are needed. The absence of a clinically scaled model with essential elements such as a three-dimension environment and dynamic coculture has hindered progress in this field. The presented research focuses on the development of an in vitro coculture model of the peripheral nervous system (PNS) involving the use of functional biomaterial which microstructure replicates nerve topography. Initially, the behavior of neuron-derived cell lines (N) and Schwann cells (SC) in contact with a short section of biomaterial (5 mm) was studied. Subsequent investigations, using fluorescent markers and survival assays, demonstrated the synergistic effects of coculture. These optimized parameters were then applied to longer biomaterials (30 mm), equivalent to clinically used NGC. The results obtained demonstrated the possibility of maintaining an extended coculture of SC and N over a 7-day period on a clinically scaled biomaterial, observing some functionality. In the long term, the knowledge gained from this work will contribute to a better understanding of the PNS regeneration process and promote the development of future therapeutic approaches while reducing reliance on animal experimentation. This model can be used for drug screening and adapted for personalized medicine trials. Ultimately, this work fills a critical gap in current research, providing a transformative approach to study and advance treatments for PNS injuries.
Collapse
Affiliation(s)
- William Choinière
- Department of Chemical Engineering and Biotechnological Engineering, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Ève Petit
- Department of Chemical Engineering and Biotechnological Engineering, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Vincent Monfette
- Department of Chemical Engineering and Biotechnological Engineering, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Samuel Pelletier
- Department of Electrical and Informatics Engineering, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Catherine Godbout-Lavoie
- Department of Chemical Engineering and Biotechnological Engineering, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Marc-Antoine Lauzon
- Department of Chemical Engineering and Biotechnological Engineering, Université de Sherbrooke, Sherbrooke, QC, Canada
- Research Center on Aging, CIUSS de l’ESTRIE-CHUS, Sherbrooke, QC, Canada
- The Quebec Network for Research on Protein Function, Engineering, and Applications, Montréal, QC, Canada
| |
Collapse
|
26
|
Dong X, Yang Y, Bao Z, Midgley AC, Li F, Dai S, Yang Z, Wang J, Liu L, Li W, Zheng Y, Liu S, Liu Y, Yu W, Liu J, Fan M, Zhu M, Shen Z, Xiaosong G, Kong D. Micro-nanofiber composite biomimetic conduits promote long-gap peripheral nerve regeneration in canine models. Bioact Mater 2023; 30:98-115. [PMID: 37560200 PMCID: PMC10406865 DOI: 10.1016/j.bioactmat.2023.06.015] [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: 03/31/2023] [Revised: 06/01/2023] [Accepted: 06/19/2023] [Indexed: 08/11/2023] Open
Abstract
Peripheral nerve injuries may result in severe long-gap interruptions that are challenging to repair. Autografting is the gold standard surgical approach for repairing long-gap nerve injuries but can result in prominent donor-site complications. Instead, imitating the native neural microarchitecture using synthetic conduits is expected to offer an alternative strategy for improving nerve regeneration. Here, we designed nerve conduits composed of high-resolution anisotropic microfiber grid-cordes with randomly organized nanofiber sheaths to interrogate the positive effects of these biomimetic structures on peripheral nerve regeneration. Anisotropic microfiber-grids demonstrated the capacity to directionally guide Schwann cells and neurites. Nanofiber sheaths conveyed adequate elasticity and permeability, whilst exhibiting a barrier function against the infiltration of fibroblasts. We then used the composite nerve conduits bridge 30-mm long sciatic nerve defects in canine models. At 12 months post-implant, the morphometric and histological recovery, gait recovery, electrophysiological function, and degree of muscle atrophy were assessed. The newly regenerated nerve tissue that formed within the composite nerve conduits showed restored neurological functions that were superior compared to sheaths-only scaffolds and Neurolac nerve conduit controls. Our findings demonstrate the feasibility of using synthetic biophysical cues to effectively bridge long-gap peripheral nerve injuries and indicates the promising clinical application prospects of biomimetic composite nerve conduits.
Collapse
Affiliation(s)
- Xianhao Dong
- State Key Laboratory of Medicinal Chemical Biology, The Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Science, Nankai University, Tianjin, China
| | - Yueyue Yang
- State Key Laboratory of Medicinal Chemical Biology, The Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Science, Nankai University, Tianjin, China
| | - Zheheng Bao
- Department of Orthopaedics, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, China
- Outpatient Department, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, China
| | - Adam C. Midgley
- State Key Laboratory of Medicinal Chemical Biology, The Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Science, Nankai University, Tianjin, China
| | - Feiyi Li
- State Key Laboratory of Medicinal Chemical Biology, The Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Science, Nankai University, Tianjin, China
| | - Shuxin Dai
- State Key Laboratory of Medicinal Chemical Biology, The Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Science, Nankai University, Tianjin, China
| | - Zhuangzhuang Yang
- State Key Laboratory of Medicinal Chemical Biology, The Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Science, Nankai University, Tianjin, China
| | - Jin Wang
- Outpatient Department, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, China
| | - Lihua Liu
- Department of Radiology, Tianjin First Central Hospital, Tianjin Medical Imaging Institute, School of Medicine, Nankai University, Tianjin, China
| | - Wenlei Li
- State Key Laboratory of Medicinal Chemical Biology, The Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Science, Nankai University, Tianjin, China
| | - Yayuan Zheng
- State Key Laboratory of Medicinal Chemical Biology, The Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Science, Nankai University, Tianjin, China
| | - Siyang Liu
- State Key Laboratory of Medicinal Chemical Biology, The Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Science, Nankai University, Tianjin, China
| | - Yang Liu
- Department of Radiology, Tianjin First Central Hospital, Tianjin Medical Imaging Institute, School of Medicine, Nankai University, Tianjin, China
| | - Weijian Yu
- State Key Laboratory of Medicinal Chemical Biology, The Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Science, Nankai University, Tianjin, China
| | - Jun Liu
- Clinical School/College of Orthopedics, Tianjin Medical University, Tianjin, China
- Department of Joint, Tianjin Hospital, Tianjin, China
| | - Meng Fan
- Department of Orthopaedics, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, China
| | - Meifeng Zhu
- State Key Laboratory of Medicinal Chemical Biology, The Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Science, Nankai University, Tianjin, China
- Haihe Laboratory of Sustainable Chemical Transformations, Keyan West Road, Tianjin, 300192, China
| | - Zhongyang Shen
- Institute of Transplantation Medicine, NHC Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, China
| | - Gu Xiaosong
- Jiangsu Key Laboratory of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Deling Kong
- State Key Laboratory of Medicinal Chemical Biology, The Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Science, Nankai University, Tianjin, China
- Institute of Transplantation Medicine, NHC Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, China
- Haihe Laboratory of Sustainable Chemical Transformations, Keyan West Road, Tianjin, 300192, China
| |
Collapse
|
27
|
Ron T, Leon A, Kafri A, Ashraf A, Na J, Babu A, Banerjee R, Brookbank H, Muddaluri SR, Little KJ, Aghion E, Pixley S. Nerve Regeneration with a Scaffold Incorporating an Absorbable Zinc-2% Iron Alloy Filament to Improve Axonal Guidance. Pharmaceutics 2023; 15:2595. [PMID: 38004574 PMCID: PMC10674795 DOI: 10.3390/pharmaceutics15112595] [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: 10/09/2023] [Revised: 10/30/2023] [Accepted: 11/02/2023] [Indexed: 11/26/2023] Open
Abstract
Peripheral nerve damage that results in lost segments requires surgery, but currently available hollow scaffolds have limitations that could be overcome by adding internal guidance support. A novel solution is to use filaments of absorbable metals to supply physical support and guidance for nerve regeneration that then safely disappear from the body. Previously, we showed that thin filaments of magnesium metal (Mg) would support nerve regeneration. Here, we tested another absorbable metal, zinc (Zn), using a proprietary zinc alloy with 2% iron (Zn-2%Fe) that was designed to overcome the limitations of both Mg and pure Zn metal. Non-critical-sized gaps in adult rat sciatic nerves were repaired with silicone conduits plus single filaments of Zn-2%Fe, Mg, or no metal, with autografts as controls. After seventeen weeks, all groups showed equal recovery of function and axonal density at the distal end of the conduit. The Zn alloy group showed some improvements in early rat health and recovery of function. The alloy had a greater local accumulation of degradation products and inflammatory cells than Mg; however, both metals had an equally thin capsule (no difference in tissue irritation) and no toxicity or inflammation in neighboring nerve tissues. Therefore, Zn-2%Fe, like Mg, is biocompatible and has great potential for use in nervous tissue regeneration and repair.
Collapse
Affiliation(s)
- Tomer Ron
- Department of Materials Engineering, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Avi Leon
- Department of Materials Engineering, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Alon Kafri
- Department of Materials Engineering, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
- Nuclear Research Centre-Negev, Beer-Sheva 84190, Israel
| | - Ahmed Ashraf
- College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - John Na
- College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Ashvin Babu
- School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Runima Banerjee
- College of Engineering & Applied Sciences, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Hunter Brookbank
- College of Arts & Sciences, University of Cincinnati, Cincinnati, OH 45221, USA
| | | | - Kevin J. Little
- Department of Orthopedic Surgery, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
- Pediatric Hand & Upper Extremity Center, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Eli Aghion
- Department of Materials Engineering, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Sarah Pixley
- Department of Pharmacology & Systems Physiology, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| |
Collapse
|
28
|
Fontana S, Caramazza L, Marracino P, Cuenca Ortolá I, Colella M, Dolciotti N, Paffi A, Gisbert Roca F, Ivashchenko S, Más Estellés J, Consales C, Balucani M, Apollonio F, Liberti M. Electric field bridging-effect in electrified microfibrils' scaffolds. Front Bioeng Biotechnol 2023; 11:1264406. [PMID: 37954020 PMCID: PMC10634785 DOI: 10.3389/fbioe.2023.1264406] [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: 07/20/2023] [Accepted: 10/03/2023] [Indexed: 11/14/2023] Open
Abstract
Introduction: The use of biocompatible scaffolds combined with the implantation of neural stem cells, is increasingly being investigated to promote the regeneration of damaged neural tissue, for instance, after a Spinal Cord Injury (SCI). In particular, aligned Polylactic Acid (PLA) microfibrils' scaffolds are capable of supporting cells, promoting their survival and guiding their differentiation in neural lineage to repair the lesion. Despite its biocompatible nature, PLA is an electrically insulating material and thus it could be detrimental for increasingly common scaffolds' electric functionalization, aimed at accelerating the cellular processes. In this context, the European RISEUP project aims to combine high intense microseconds pulses and DC stimulation with neurogenesis, supported by a PLA microfibrils' scaffold. Methods: In this paper a numerical study on the effect of microfibrils' scaffolds on the E-field distribution, in planar interdigitated electrodes, is presented. Realistic microfibrils' 3D CAD models have been built to carry out a numerical dosimetry study, through Comsol Multiphysics software. Results: Under a voltage of 10 V, microfibrils redistribute the E-field values focalizing the field streamlines in the spaces between the fibers, allowing the field to pass and reach maximum values up to 100 kV/m and values comparable with the bare electrodes' device (without fibers). Discussion: Globally the median E-field inside the scaffolded electrodes is the 90% of the nominal field, allowing an adequate cells' exposure.
Collapse
Affiliation(s)
- Sara Fontana
- BioEM Lab, Department of Information Engineering, Electronics and Telecommunications (DIET), Sapienza University of Rome, Rome, Italy
- Center for Life Nano- & Neuro-Science, Fondazione Istituto Italiano di Tecnologia (IIT), Rome, Italy
| | - Laura Caramazza
- BioEM Lab, Department of Information Engineering, Electronics and Telecommunications (DIET), Sapienza University of Rome, Rome, Italy
- Center for Life Nano- & Neuro-Science, Fondazione Istituto Italiano di Tecnologia (IIT), Rome, Italy
| | | | - Irene Cuenca Ortolá
- Center for Biomaterials and Tissue Engineering, Universitat Politècnica de València, Valencia, Spain
| | - Micol Colella
- BioEM Lab, Department of Information Engineering, Electronics and Telecommunications (DIET), Sapienza University of Rome, Rome, Italy
| | - Noemi Dolciotti
- BioEM Lab, Department of Information Engineering, Electronics and Telecommunications (DIET), Sapienza University of Rome, Rome, Italy
| | - Alessandra Paffi
- BioEM Lab, Department of Information Engineering, Electronics and Telecommunications (DIET), Sapienza University of Rome, Rome, Italy
| | - Fernando Gisbert Roca
- Center for Biomaterials and Tissue Engineering, Universitat Politècnica de València, Valencia, Spain
| | - Sergiy Ivashchenko
- Center for Biomaterials and Tissue Engineering, Universitat Politècnica de València, Valencia, Spain
| | - Jorge Más Estellés
- Center for Biomaterials and Tissue Engineering, Universitat Politècnica de València, Valencia, Spain
| | - Claudia Consales
- Division of Health Protection Technologies, ENEA-Italian National Agency for New Technologies, Energy and Sustainable Economic Development, Rome, Italy
| | | | - Francesca Apollonio
- BioEM Lab, Department of Information Engineering, Electronics and Telecommunications (DIET), Sapienza University of Rome, Rome, Italy
- Center for Life Nano- & Neuro-Science, Fondazione Istituto Italiano di Tecnologia (IIT), Rome, Italy
| | - Micaela Liberti
- BioEM Lab, Department of Information Engineering, Electronics and Telecommunications (DIET), Sapienza University of Rome, Rome, Italy
- Center for Life Nano- & Neuro-Science, Fondazione Istituto Italiano di Tecnologia (IIT), Rome, Italy
| |
Collapse
|
29
|
Abstract
Many soft tissues of the human body possess hierarchically anisotropic structures, exhibiting orientation-specific mechanical properties and biological functionality. Hydrogels have been proposed as promising scaffold materials for tissue engineering applications due to their water-rich composition, excellent biocompatibility, and tunable physico-chemical properties. However, conventional hydrogels with homogeneous structures often exhibit isotropic properties that differ from those of biological tissues, limiting their further application. Recently, magnetically anisotropic hydrogels containing long-range ordered magneto-structures have attracted widespread interest owing to their highly controllable assembly strategy, rapid magnetic responsiveness and remote spatiotemporal regulation. In this review, we summarize the latest progress of magnetically anisotropic hydrogels for tissue engineering. The fabrication strategy of magnetically anisotropic hydrogels from magnetic nanofillers with different dimensions is systemically introduced. Then, the effects of magnetically anisotropic cues on the physicochemical properties of hydrogels and the cellular biological behavior are discussed. And the applications of magnetically anisotropic hydrogels in the engineering of different tissues are emphasized. Finally, the current challenges and the future perspectives for magnetically anisotropic hydrogels are presented.
Collapse
Affiliation(s)
- Lili Hao
- Research Institute for Biomaterials, Tech Institute for Advanced Materials, College of Materials Science and Engineering, Nanjing Tech University, Nanjing 211816, China.
| | - Hongli Mao
- Research Institute for Biomaterials, Tech Institute for Advanced Materials, College of Materials Science and Engineering, Nanjing Tech University, Nanjing 211816, China.
| |
Collapse
|
30
|
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: 12] [Impact Index Per Article: 6.0] [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
|
31
|
Du W, Wang T, Hu S, Luan J, Tian F, Ma G, Xue J. Engineering of electrospun nanofiber scaffolds for repairing brain injury. ENGINEERED REGENERATION 2023; 4:289-303. [DOI: 10.1016/j.engreg.2023.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/14/2023] Open
|
32
|
Zhang Y, Bai P, Lu J, Lui KHW, Zhao T, Wen D, He B, Zhu Z. Effect of growth differentiation factor 11 expression after peripheral nerve injury in Sprague-Dawley rats. Neurol Res 2023; 45:835-842. [PMID: 37220327 DOI: 10.1080/01616412.2023.2211446] [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: 11/02/2022] [Accepted: 05/01/2023] [Indexed: 05/25/2023]
Abstract
OBJECTIVES We attempt to investigate the expression pattern of GDF11 in the sciatic nerves after injury. METHODS Thirty-six healthy male Sprague Dawley (SD) rats were divided into three groups at random and were labelled as: day 1, day 4, and day 7 post-surgery. The sciatic nerve crush model was established on the left-hind limb, while the right limb was untreated, and served as the control. Nerve samples were collected at post-injury day 1, day 4 and day 7. Nerve samples collected from the proximal and distal stump of the injury site underwent immunofluorescence staining with GDF11, NF200 and CD31. GDF11 mRNA expression was analyzed by qRT-PCR. CCK-8 assay, after si-GDF11 transfection in Schwann cells (RSC96) was applied to verify its effect in cell proliferation rate. RESULTS GDF11 was abundantly expressed in axons stained with NF200 and Schwann cells stained with S100. However, no GDF11 expression was observed in vascular endothelial tissues stained with CD31. From day 4 onwards, the level of GDF11 showed an increasing trend, up to a twofold level at day 7 after injury. Proliferation rate of RSC96 cells showed a significant decrease after the down-regulation of GDF11 by siRNAs compared to the control group. CONCLUSIONS GDF11 may play a role in the proliferation of Schwann cell during nerve regeneration process.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Plastic Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Peiwen Bai
- Department of Plastic Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jiamin Lu
- Department of Plastic Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Koon Hei Winson Lui
- Department of Plastic Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Tianjiao Zhao
- Department of Plastic Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Di Wen
- Department of Plastic Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Bo He
- Joint and Orthopedic Trauma, Orthopedic Department, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zhaowei Zhu
- Department of Plastic Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
33
|
Agarwal T, Chiesa I, Costantini M, Lopamarda A, Tirelli MC, Borra OP, Varshapally SVS, Kumar YAV, Koteswara Reddy G, De Maria C, Zhang LG, Maiti TK. Chitosan and its derivatives in 3D/4D (bio) printing for tissue engineering and drug delivery applications. Int J Biol Macromol 2023; 246:125669. [PMID: 37406901 DOI: 10.1016/j.ijbiomac.2023.125669] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 06/19/2023] [Accepted: 07/01/2023] [Indexed: 07/07/2023]
Abstract
Tissue engineering research has undergone to a revolutionary improvement, thanks to technological advancements, such as the introduction of bioprinting technologies. The ability to develop suitable customized biomaterial inks/bioinks, with excellent printability and ability to promote cell proliferation and function, has a deep impact on such improvements. In this context, printing inks based on chitosan and its derivatives have been instrumental. Thus, the current review aims at providing a comprehensive overview on chitosan-based materials as suitable inks for 3D/4D (bio)printing and their applicability in creating advanced drug delivery platforms and tissue engineered constructs. Furthermore, relevant strategies to improve the mechanical and biological performances of this biomaterial are also highlighted.
Collapse
Affiliation(s)
- Tarun Agarwal
- Department of Bio-Technology, Koneru Lakshmaiah Education Foundation, Vaddeswaram, AP, India.
| | - Irene Chiesa
- Research Center "E. Piaggio", Department of Information Engineering, University of Pisa, Largo Lucio Lazzarino 1, 56122 Pisa, Italy
| | - Marco Costantini
- Institute of Physical Chemistry, Polish Academy of Sciences, 01-224 Warsaw, Poland.
| | - Anna Lopamarda
- Research Center "E. Piaggio", Department of Information Engineering, University of Pisa, Largo Lucio Lazzarino 1, 56122 Pisa, Italy
| | | | - Om Prakash Borra
- Department of Bio-Technology, Koneru Lakshmaiah Education Foundation, Vaddeswaram, AP, India
| | | | | | - G Koteswara Reddy
- Department of Bio-Technology, Koneru Lakshmaiah Education Foundation, Vaddeswaram, AP, India
| | - Carmelo De Maria
- Research Center "E. Piaggio", Department of Information Engineering, University of Pisa, Largo Lucio Lazzarino 1, 56122 Pisa, Italy.
| | - Lijie Grace Zhang
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC 20052, USA; Department of Electrical Engineering, The George Washington University, Washington, DC 20052, USA; Department of Biomedical Engineering, The George Washington University, Washington, DC 20052, USA; Department of Medicine, The George Washington University, Washington, DC 20052, USA
| | - Tapas Kumar Maiti
- Department of Biotechnology, Indian Institute of technology Kharagpur, West Bengal 721302, India
| |
Collapse
|
34
|
Lee D, Yang K, Xie J. Advances in Nerve Injury Models on a Chip. Adv Biol (Weinh) 2023; 7:e2200227. [PMID: 36709421 DOI: 10.1002/adbi.202200227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 12/19/2022] [Indexed: 01/30/2023]
Abstract
Regeneration and functional recovery of the damaged nerve are challenging due to the need for effective therapeutic drugs, biomaterials, and approaches. The poor outcome of the treatment of nerve injury stems from the incomplete understanding of axonal biology and interactions between neurons and the surrounding environment, such as glial cells and extracellular matrix. Microfluidic devices, in combination with various injury techniques, have been applied to test biological hypotheses in nerve injury and nerve regeneration. The microfluidic devices provide multiple advantages over the in vitro cell culture on a petri dish and in vivo animal models because a specific part of the neuronal environment can be manipulated using physical and chemical interventions. In addition, single-cell behavior and interactions between neurons and glial cells can be visualized and quantified on microfluidic platforms. In this article, current in vitro nerve injury models on a chip that mimics in vivo axonal injuries and the regeneration process of axons are summarized. The microfluidic-based nerve injury models could enhance the understanding of the physiological and pathophysiological mechanisms of nerve tissues and simultaneously serve as powerful drug and biomaterial screening platforms.
Collapse
Affiliation(s)
- Donghee Lee
- Department of Surgery-Transplant and Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Kai Yang
- Department of Surgery-Plastic Surgery, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Jingwei Xie
- Department of Surgery-Transplant and Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Department of Mechanical and Materials Engineering, College of Engineering, University of Nebraska Lincoln, Lincoln, NE, 68588, USA
| |
Collapse
|
35
|
Takeya H, Itai S, Kimura H, Kurashina Y, Amemiya T, Nagoshi N, Iwamoto T, Sato K, Shibata S, Matsumoto M, Onoe H, Nakamura M. Schwann cell-encapsulated chitosan-collagen hydrogel nerve conduit promotes peripheral nerve regeneration in rodent sciatic nerve defect models. Sci Rep 2023; 13:11932. [PMID: 37488180 PMCID: PMC10366170 DOI: 10.1038/s41598-023-39141-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 07/20/2023] [Indexed: 07/26/2023] Open
Abstract
Chitosan has various tissue regeneration effects. This study was designed to investigate the nerve regeneration effect of Schwann cell (SC)-encapsulated chitosan-collagen hydrogel nerve conduit (CCN) transplanted into a rat model of sciatic nerve defect. We prepared a CCN consisting of an outer layer of chitosan hydrogel and an inner layer of collagen hydrogel to encapsulate the intended cells. Rats with a 10-mm sciatic nerve defect were treated with SCs encapsulated in CCN (CCN+), CCN without SCs (CCN-), SC-encapsulated silicone tube (silicone+), and autologous nerve transplanting (auto). Behavioral and histological analyses indicated that motor functional recovery, axonal regrowth, and myelination of the CCN+ group were superior to those of the CCN- and silicone+ groups. Meanwhile, the CCN- and silicone+ groups showed no significant differences in the recovery of motor function and nerve histological restoration. In conclusion, SC-encapsulated CCN has a synergistic effect on peripheral nerve regeneration, especially axonal regrowth and remyelination of host SCs. In the early phase after transplantation, SC-encapsulated CCNs have a positive effect on recovery. Therefore, using SC-encapsulated CCNs may be a promising approach for massive peripheral nerve defects.
Collapse
Affiliation(s)
- Hiroaki Takeya
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi Shinjuku-Ku, Tokyo, 160-8582, Japan
| | - Shun Itai
- Department of Mechanical Engineering, Faculty of Science and Technology, Keio University, 3-14-1 Hiyoshi, Kohoku-Ku, Yokohama-Shi, Kanagawa, 223-8522, Japan
- Division of Medical Science, Graduate School of Biomedical Engineering, Tohoku University, 1-1 Seiryomachi, Aoba-Ku, Sendai, Miyagi, 980-8574, Japan
| | - Hiroo Kimura
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi Shinjuku-Ku, Tokyo, 160-8582, Japan.
| | - Yuta Kurashina
- Division of Advanced Mechanical Systems Engineering, Institute of Engineering, Tokyo University of Agriculture and Technology, 2-24-16 Nakacho, Koganei-Shi, Tokyo, 184-8588, Japan
| | - Tsuyoshi Amemiya
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi Shinjuku-Ku, Tokyo, 160-8582, Japan
| | - Narihito Nagoshi
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi Shinjuku-Ku, Tokyo, 160-8582, Japan
| | - Takuji Iwamoto
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi Shinjuku-Ku, Tokyo, 160-8582, Japan
| | - Kazuki Sato
- Institute for Integrated Sports Medicine, Keio University School of Medicine, 35 Shinanomachi Shinjuku-Ku, Tokyo, Japan
| | - Shinsuke Shibata
- Division of Microscopic Anatomy, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-Dori, Chuo-Ku, Niigata, 951-8510, Japan
| | - Morio Matsumoto
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi Shinjuku-Ku, Tokyo, 160-8582, Japan
| | - Hiroaki Onoe
- Department of Mechanical Engineering, Faculty of Science and Technology, Keio University, 3-14-1 Hiyoshi, Kohoku-Ku, Yokohama-Shi, Kanagawa, 223-8522, Japan
| | - Masaya Nakamura
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi Shinjuku-Ku, Tokyo, 160-8582, Japan
| |
Collapse
|
36
|
Venturini C, Sáez P. A multi-scale clutch model for adhesion complex mechanics. PLoS Comput Biol 2023; 19:e1011250. [PMID: 37450544 PMCID: PMC10393167 DOI: 10.1371/journal.pcbi.1011250] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 06/07/2023] [Indexed: 07/18/2023] Open
Abstract
Cell-matrix adhesion is a central mechanical function to a large number of phenomena in physiology and disease, including morphogenesis, wound healing, and tumor cell invasion. Today, how single cells respond to different extracellular cues has been comprehensively studied. However, how the mechanical behavior of the main individual molecules that form an adhesion complex cooperatively responds to force within the adhesion complex is still poorly understood. This is a key aspect of cell adhesion because how these cell adhesion molecules respond to force determines not only cell adhesion behavior but, ultimately, cell function. To answer this question, we develop a multi-scale computational model for adhesion complexes mechanics. We extend the classical clutch hypothesis to model individual adhesion chains made of a contractile actin network, a talin rod, and an integrin molecule that binds at individual adhesion sites on the extracellular matrix. We explore several scenarios of integrins dynamics and analyze the effects of diverse extracellular matrices on the behavior of the adhesion molecules and on the whole adhesion complex. Our results describe how every single component of the adhesion chain mechanically responds to the contractile actomyosin force and show how they control the traction forces exerted by the cell on the extracellular space. Importantly, our computational results agree with previous experimental data at the molecular and cellular levels. Our multi-scale clutch model presents a step forward not only to further understand adhesion complexes mechanics but also to impact, e.g., the engineering of biomimetic materials, tissue repairment, or strategies to arrest tumor progression.
Collapse
Affiliation(s)
- Chiara Venturini
- Laboratori de Càlcul Numèric (LaCaN), Universitat Politècnica de Catalunya, Barcelona, Spain
| | - Pablo Sáez
- Laboratori de Càlcul Numèric (LaCaN), Universitat Politècnica de Catalunya, Barcelona, Spain
- E.T.S. de Ingeniería de Caminos, Universitat Politècnica de Catalunya, Barcelona, Spain
- Institut de Matemàtiques de la UPC-BarcelonaTech (IMTech), Universitat Politècnica de Catalunya, Barcelona, Spain
| |
Collapse
|
37
|
Shen Y, Luo Y, Liao P, Zuo Y, Jiang R. Role of the Voltage-Gated Proton Channel Hv1 in Nervous Systems. Neurosci Bull 2023; 39:1157-1172. [PMID: 37029856 PMCID: PMC10313628 DOI: 10.1007/s12264-023-01053-6] [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: 10/12/2022] [Accepted: 02/20/2023] [Indexed: 04/09/2023] Open
Abstract
Hv1 is the only voltage-gated proton-selective channel in mammalian cells. It contains a conserved voltage-sensor domain, shared by a large class of voltage-gated ion channels, but lacks a pore domain. Its primary role is to extrude protons from the cytoplasm upon pH reduction and membrane depolarization. The best-known function of Hv1 is the regulation of cytosolic pH and the nicotinamide adenine dinucleotide phosphate oxidase-dependent production of reactive oxygen species. Accumulating evidence indicates that Hv1 is expressed in nervous systems, in addition to immune cells and others. Here, we summarize the molecular properties, distribution, and physiological functions of Hv1 in the peripheral and central nervous systems. We describe the recently discovered functions of Hv1 in various neurological diseases, including brain or spinal cord injury, ischemic stroke, demyelinating diseases, and pain. We also summarize the current advances in the discovery and application of Hv1-targeted small molecules in neurological diseases. Finally, we discuss the current limitations of our understanding of Hv1 and suggest future research directions.
Collapse
Affiliation(s)
- Yu Shen
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610000, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610000, China
| | - Yuncheng Luo
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610000, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610000, China
| | - Ping Liao
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610000, China
| | - Yunxia Zuo
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610000, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610000, China
| | - Ruotian Jiang
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610000, China.
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610000, China.
| |
Collapse
|
38
|
Dresvyanina EN, Tagandurdyyeva NA, Kodolova-Chukhontseva VV, Dobrovol'skaya IP, Kamalov AM, Nashchekina YA, Nashchekin AV, Ivanov AG, Yukina GY, Yudin VE. Structure and Properties of Composite Fibers Based on Chitosan and Single-Walled Carbon Nanotubes for Peripheral Nerve Regeneration. Polymers (Basel) 2023; 15:2860. [PMID: 37447506 DOI: 10.3390/polym15132860] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 06/19/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
This study focused on a potential application of electrically conductive, biocompatible, bioresorbable fibers for tubular conduits aimed at the regeneration of peripheral nerves. The conducting, mechanical, and biological properties of composite fibers based on chitosan and single-walled carbon nanotubes were investigated in this paper. It was shown that introducing 0.5 wt.% of SWCNT into the composite fibers facilitated the formation of a denser fiber structure, resulting in improved strength (σ = 260 MPa) and elastic (E = 14 GPa) characteristics. Additionally, the composite fibers were found to be biocompatible and did not cause significant inflammation or deformation during in vivo studies. A thin layer of connective tissue formed around the fiber.
Collapse
Affiliation(s)
- Elena N Dresvyanina
- Institute of Textile and Fashion, Saint Petersburg State University of Industrial Technologies and Design, B. Morskaya Str., 18, Saint Petersburg 191186, Russia
| | - Nurjemal A Tagandurdyyeva
- Institute of Biomedical Systems and Biotechnology, Peter the Great Saint Petersburg Polytechnic University, Polytekhnicheskaya Str., 29, Saint Petersburg 195251, Russia
| | - Vera V Kodolova-Chukhontseva
- Institute of Biomedical Systems and Biotechnology, Peter the Great Saint Petersburg Polytechnic University, Polytekhnicheskaya Str., 29, Saint Petersburg 195251, Russia
- Institute of Macromolecular Compounds of Russian Academy of Sciences, VO Bolshoy pr., 31, Saint Petersburg 199004, Russia
| | - Irina P Dobrovol'skaya
- Institute of Macromolecular Compounds of Russian Academy of Sciences, VO Bolshoy pr., 31, Saint Petersburg 199004, Russia
| | - Almaz M Kamalov
- Institute of Biomedical Systems and Biotechnology, Peter the Great Saint Petersburg Polytechnic University, Polytekhnicheskaya Str., 29, Saint Petersburg 195251, Russia
| | - Yulia A Nashchekina
- Institute of Cytology Russian Academy of Sciences, Tikhoretsky Ave., 4, Saint Petersburg 194064, Russia
| | - Alexey V Nashchekin
- Ioffe Institute, Polytekhnicheskaya Str., 26, Saint Petersburg 194021, Russia
| | - Alexey G Ivanov
- Institute of Macromolecular Compounds of Russian Academy of Sciences, VO Bolshoy pr., 31, Saint Petersburg 199004, Russia
| | - Galina Yu Yukina
- Pavlov First Saint Petersburg State Medical University, L'va Tolstogo Str. 6-8, Saint Petersburg 197022, Russia
| | - Vladimir E Yudin
- Institute of Macromolecular Compounds of Russian Academy of Sciences, VO Bolshoy pr., 31, Saint Petersburg 199004, Russia
| |
Collapse
|
39
|
Zhao W, Tu H, Chen J, Wang J, Liu H, Zhang F, Li J. Functionalized hydrogels in neural injury repairing. Front Neurosci 2023; 17:1199299. [PMID: 37404462 PMCID: PMC10315583 DOI: 10.3389/fnins.2023.1199299] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 04/27/2023] [Indexed: 07/06/2023] Open
Abstract
Repairing injuries to the nervous system has always been a prominent topic in clinical research. Direct suturing and nerve displacement surgery are the primary treatment options, but they may not be suitable for long nerve injuries and may require sacrificing the functionality of other autologous nerves. With the emergence of tissue engineering, hydrogel materials have been identified as a promising technology with clinical translation potential for repairing nervous system injuries due to their excellent biocompatibility and ability to release or deliver functional ions. By controlling their composition and structure, hydrogels can be Functionalized and almost fully matched with nerve tissue and even simulate nerve conduction function and mechanical properties. Thus, they are suitable for repairing injuries to both the central and peripheral nervous systems. This article provides a review of recent research progress in functionalized hydrogels for nerve injury repair, highlighting the design differences among various materials and future research directions. We strongly believe that the development of functionalized hydrogels has great potential for improving the clinical treatment of nerve injuries.
Collapse
Affiliation(s)
- Wenqian Zhao
- College of Medical Technology and Engineering, Henan University of Science and Technology, Luoyang, China
| | - Hui Tu
- College of Medical Technology and Engineering, Henan University of Science and Technology, Luoyang, China
| | - Jianxiao Chen
- Department of Nephrology, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang, China
| | - Jing Wang
- College of Medical Technology and Engineering, Henan University of Science and Technology, Luoyang, China
| | - Haoting Liu
- College of Medical Technology and Engineering, Henan University of Science and Technology, Luoyang, China
| | - Fengshou Zhang
- College of Medical Technology and Engineering, Henan University of Science and Technology, Luoyang, China
| | - Jing Li
- Office of Science and Technology, Henan University of Science and Technology, Luoyang, China
| |
Collapse
|
40
|
Mays EA, Ellis EB, Hussain Z, Parajuli P, Sundararaghavan HG. Enzyme-Mediated Nerve Growth Factor Release from Nanofibers Using Gelatin Microspheres. Tissue Eng Part A 2023; 29:333-343. [PMID: 37016821 DOI: 10.1089/ten.tea.2022.0205] [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: 04/06/2023] Open
Abstract
Spinal cord injury is a complex environment, with many conflicting growth factors present at different times throughout the injury timeline. Delivery of multiple growth factors has received mixed results, highlighting a need to consider the timing of delivery for possibly antagonistic growth factors. Cell-mediated degradation of delivery vehicles for delayed release of growth factors offers an attractive way to exploit the highly active immune response in the spinal cord injury environment. In this study, growth factor-loaded gelatin microspheres (GMS) combined with methacrylated hyaluronic acid (MeHA) were electrospun to create GMS fibers (GMSF) for delayed release of growth factors (GFs). GMS were successfully combined with MeHA while electrospinning, with an average fiber diameter of 365 ± 10 nm and 44% ± 8% fiber alignment. GMSF with nerve growth factor (NGF) was tested on dissociated chick dorsal root ganglia cells. We further tested the effect of M1 macrophage-conditioned media (M1CM) to simulate macrophage invasion after spinal cord injury for cell-mediated degradation. We hypothesized that neurons grown on GMSF with loaded NGF would exhibit longer neurites in M1CM, showing a release of functional NGF, as compared with controls. GMSF in M1CM was significantly different from MeHA in serum-free media (SFM) and M0-conditioned media (M0CM), as well as GMSF in M0CM (p < 0.05). Moreover, GMSF + NGF in all media conditions were significantly different from MeHA in SFM and M0CM (p < 0.05). The goal of this study was to develop a biomaterial system where drug delivery is triggered by immune response, allowing for more control and longer exposure to encapsulated drugs. The spinal cord injury microenvironment is known to have a robust immune response, making this immune-medicated drug release system particularly significant for directed repair.
Collapse
Affiliation(s)
- Elizabeth A Mays
- Department of Biomedical Engineering, Wayne State University, Detroit, Michigan, USA
| | - Eric B Ellis
- Department of Chemical Engineering and Materials Science, Wayne State University, Detroit, Michigan, USA
| | - Zahin Hussain
- School of Medicine, Michigan State University, East Lansing, Michigan, USA
| | - Prahlad Parajuli
- Department of Pharmaceutical Sciences, Wayne State University, Detroit, Michigan, USA
| | | |
Collapse
|
41
|
Liu Y, Zhang X, Xiao C, Liu B. Engineered hydrogels for peripheral nerve repair. Mater Today Bio 2023; 20:100668. [PMID: 37273791 PMCID: PMC10232914 DOI: 10.1016/j.mtbio.2023.100668] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 05/06/2023] [Accepted: 05/16/2023] [Indexed: 06/06/2023] Open
Abstract
Peripheral nerve injury (PNI) is a complex disease that often appears in young adults. It is characterized by a high incidence, limited treatment options, and poor clinical outcomes. This disease not only causes dysfunction and psychological disorders in patients but also brings a heavy burden to the society. Currently, autologous nerve grafting is the gold standard in clinical treatment, but complications, such as the limited source of donor tissue and scar tissue formation, often further limit the therapeutic effect. Recently, a growing number of studies have used tissue-engineered materials to create a natural microenvironment similar to the nervous system and thus promote the regeneration of neural tissue and the recovery of impaired neural function with promising results. Hydrogels are often used as materials for the culture and differentiation of neurogenic cells due to their unique physical and chemical properties. Hydrogels can provide three-dimensional hydration networks that can be integrated into a variety of sizes and shapes to suit the morphology of neural tissues. In this review, we discuss the recent advances of engineered hydrogels for peripheral nerve repair and analyze the role of several different therapeutic strategies of hydrogels in PNI through the application characteristics of hydrogels in nerve tissue engineering (NTE). Furthermore, the prospects and challenges of the application of hydrogels in the treatment of PNI are also discussed.
Collapse
Affiliation(s)
- Yao Liu
- Hand and Foot Surgery Department, First Hospital of Jilin University, Xinmin Street, Changchun, 130061, PR China
| | - Xiaonong Zhang
- Key Laboratory of Polymer Ecomaterials, Jilin Biomedical Polymers Engineering Laboratory, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China
| | - Chunsheng Xiao
- Key Laboratory of Polymer Ecomaterials, Jilin Biomedical Polymers Engineering Laboratory, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China
| | - Bin Liu
- Hand and Foot Surgery Department, First Hospital of Jilin University, Xinmin Street, Changchun, 130061, PR China
| |
Collapse
|
42
|
Jiang L, Ouyang X, Zhang D, Wang G, Zhang Z, Wang W, Yan H. The role of Gel-Ppy-modified nerve conduit on the repair of sciatic nerve defect in rat model. FASEB J 2023; 37:e22921. [PMID: 37052612 DOI: 10.1096/fj.202201969r] [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: 11/24/2022] [Revised: 02/14/2023] [Accepted: 03/30/2023] [Indexed: 04/14/2023]
Abstract
The serious clinical challenge of peripheral nerve injury (PNI) is nerve regeneration. Nerve conduit represents a promising strategy to contribute to nerve regeneration by bridging injured nerve gaps. However, due to a unique microenvironment of nerve tissue, autologous nerves have not been substituted by nerve conduit. Nerve regeneration after nerve conduit implantation depends on many factors, such as conductivity and biocompatibility. Therefore, Gelatin (Gel) with biocompatibility and polypyrrole (Ppy) with conductivity is highly concerned. In this paper, Gel-Ppy modified nerve conduit was fabricated with great biocompatibility and conductivity to evaluate its properties of enhancing nerve regeneration in vivo and in vitro. The proliferation of Schwann cells on Gel-Ppy modified nerve conduit was remarkably increased. Consistent with in vitro results, the Gel-Ppy nerve conduit could contribute to the regeneration of Schwann cell in vivo. The axon diameters and myelin sheath thickness were also enhanced, resulting in the amelioration of muscle atrophy, nerve conduction, and motor function recovery. To explain this interesting phenomenon, western blot results indicated that the Gel-Ppy conduit facilitated nerve regeneration via upregulating the Rap1 pathway to induce neurite outgrowth. Therefore, the above results demonstrated that Gel-Ppy modified nerve conduit could provide an acceptable microenvironment for nerve regeneration and be popularized as a novel therapeutic strategy of PNI.
Collapse
Affiliation(s)
- Liangfu Jiang
- Key Laboratory of Orthopedics of Zhejiang Province, Department of Orthopedics (Division of Wound Repair), The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Xingyu Ouyang
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Dupiao Zhang
- Key Laboratory of Orthopedics of Zhejiang Province, Department of Orthopedics (Division of Hand Surgery), The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Gang Wang
- College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, China
| | - Zhe Zhang
- Key Laboratory of Orthopedics of Zhejiang Province, Department of Orthopedics (Division of Hand Surgery), The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Wei Wang
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Hede Yan
- Key Laboratory of Orthopedics of Zhejiang Province, Department of Orthopedics (Division of Hand Surgery), The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
43
|
Sumam P, Parameswaran R. Neuronal cell response on aligned fibroporous electrospun mat generated from silver ion complexed ethylene vinyl alcohol copolymer. J Biomed Mater Res B Appl Biomater 2023; 111:782-794. [PMID: 36333924 DOI: 10.1002/jbm.b.35189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 09/12/2022] [Accepted: 10/10/2022] [Indexed: 11/07/2022]
Abstract
Generating electrospun mats with aligned fibers and obtaining neurite extension in the aligned fiber direction could provide hope for fabricating nerve guidance conduits or wraps through an easy method. The growing interest in generating electrospun mats with aligned fibers for tissue engineering is looking for simple methods to generate the same. Here, in this study, ethylene vinyl alcohol copolymer (EVAL) chains were complexed with silver ions (Ag+ ) to generate aligned fibers during the electrospinning process. The fibers thus produced were subjected to physico-chemical characterization and biological studies to ensure their properties and to examine whether suitable for neuronal cell attachment and neurite extension that may be useful in making nerve guidance conduits or wraps. The presence of silver ions and its complex formation with -OH of EVAL has been confirmed with EDX and XPS analysis respectively. The alignment of fibers was visualized from SEM analysis and confirmed using directionality analysis using Fiji-ImageJ software. Mechanical properties done with dumbbells punched out in longitudinal and transverse directions also substantiated the alignment of fibers. The results obtained from direct contact, MTT, and live/dead assay showed the cells are viable on the material. From the actin staining and immunostaining assays, it was evident that the PC12 cells could attach and extend their neurites in an aligned manner on the fibers. The maximum neurite extension was up to 200 μm in length. These properties of electrospun EVAL-Ag mat with aligned fibers indicated that it could be developed as a biocompatible nerve guidance conduit or wrap.
Collapse
Affiliation(s)
- Prima Sumam
- Division of Polymeric Medical Devices, Department of Medical Devices Engineering, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, India
| | - Ramesh Parameswaran
- Division of Polymeric Medical Devices, Department of Medical Devices Engineering, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, India
| |
Collapse
|
44
|
Tarim EA, Anil Inevi M, Ozkan I, Kecili S, Bilgi E, Baslar MS, Ozcivici E, Oksel Karakus C, Tekin HC. Microfluidic-based technologies for diagnosis, prevention, and treatment of COVID-19: recent advances and future directions. Biomed Microdevices 2023; 25:10. [PMID: 36913137 PMCID: PMC10009869 DOI: 10.1007/s10544-023-00649-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/21/2023] [Indexed: 03/14/2023]
Abstract
The COVID-19 pandemic has posed significant challenges to existing healthcare systems around the world. The urgent need for the development of diagnostic and therapeutic strategies for COVID-19 has boomed the demand for new technologies that can improve current healthcare approaches, moving towards more advanced, digitalized, personalized, and patient-oriented systems. Microfluidic-based technologies involve the miniaturization of large-scale devices and laboratory-based procedures, enabling complex chemical and biological operations that are conventionally performed at the macro-scale to be carried out on the microscale or less. The advantages microfluidic systems offer such as rapid, low-cost, accurate, and on-site solutions make these tools extremely useful and effective in the fight against COVID-19. In particular, microfluidic-assisted systems are of great interest in different COVID-19-related domains, varying from direct and indirect detection of COVID-19 infections to drug and vaccine discovery and their targeted delivery. Here, we review recent advances in the use of microfluidic platforms to diagnose, treat or prevent COVID-19. We start by summarizing recent microfluidic-based diagnostic solutions applicable to COVID-19. We then highlight the key roles microfluidics play in developing COVID-19 vaccines and testing how vaccine candidates perform, with a focus on RNA-delivery technologies and nano-carriers. Next, microfluidic-based efforts devoted to assessing the efficacy of potential COVID-19 drugs, either repurposed or new, and their targeted delivery to infected sites are summarized. We conclude by providing future perspectives and research directions that are critical to effectively prevent or respond to future pandemics.
Collapse
Affiliation(s)
- E Alperay Tarim
- Department of Bioengineering, Izmir Institute of Technology, Izmir, Turkey
| | - Muge Anil Inevi
- Department of Bioengineering, Izmir Institute of Technology, Izmir, Turkey
| | - Ilayda Ozkan
- Department of Bioengineering, Izmir Institute of Technology, Izmir, Turkey
| | - Seren Kecili
- Department of Bioengineering, Izmir Institute of Technology, Izmir, Turkey
| | - Eyup Bilgi
- Department of Bioengineering, Izmir Institute of Technology, Izmir, Turkey
| | - M Semih Baslar
- Department of Bioengineering, Izmir Institute of Technology, Izmir, Turkey
| | - Engin Ozcivici
- Department of Bioengineering, Izmir Institute of Technology, Izmir, Turkey
| | | | - H Cumhur Tekin
- Department of Bioengineering, Izmir Institute of Technology, Izmir, Turkey.
- METU MEMS Center, Ankara, Turkey.
| |
Collapse
|
45
|
Fan Z, Wen X, Ding X, Wang Q, Wang S, Yu W. Advances in biotechnology and clinical therapy in the field of peripheral nerve regeneration based on magnetism. Front Neurol 2023; 14:1079757. [PMID: 36970536 PMCID: PMC10036769 DOI: 10.3389/fneur.2023.1079757] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 02/07/2023] [Indexed: 03/12/2023] Open
Abstract
Peripheral nerve injury (PNI) is one of the most common neurological diseases. Recent studies on nerve cells have provided new ideas for the regeneration of peripheral nerves and treatment of physical trauma or degenerative disease-induced loss of sensory and motor neuron functions. Accumulating evidence suggested that magnetic fields might have a significant impact on the growth of nerve cells. Studies have investigated different magnetic field properties (static or pulsed magnetic field) and intensities, various magnetic nanoparticle-encapsulating cytokines based on superparamagnetism, magnetically functionalized nanofibers, and their relevant mechanisms and clinical applications. This review provides an overview of these aspects as well as their future developmental prospects in related fields.
Collapse
|
46
|
de Assis ACC, Reis ALS, Nunes LV, Ferreira LFR, Bilal M, Iqbal HMN, Soriano RN. Stem Cells and Tissue Engineering-Based Therapeutic Interventions: Promising Strategies to Improve Peripheral Nerve Regeneration. Cell Mol Neurobiol 2023; 43:433-454. [PMID: 35107689 PMCID: PMC11415205 DOI: 10.1007/s10571-022-01199-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Accepted: 01/21/2022] [Indexed: 02/05/2023]
Abstract
Unlike the central nervous system, the peripheral one has the ability to regenerate itself after injury; however, this natural regeneration process is not always successful. In fact, even with some treatments, the prognosis is poor, and patients consequently suffer with the functional loss caused by injured nerves, generating several impacts on their quality of life. In the present review we aimed to address two strategies that may considerably potentiate peripheral nerve regeneration: stem cells and tissue engineering. In vitro studies have shown that pluripotent cells associated with neural scaffolds elaborated by tissue engineering can increase functional recovery, revascularization, remyelination, neurotrophin expression and reduce muscle atrophy. Although these results are very promising, it is important to note that there are some barriers to be circumvented: the host's immune response, the oncogenic properties attributed to stem cells and the duration of the pro-regenerative effects. After all, more studies are still needed to overcome the limitations of these treatments; those that address techniques for manipulating the lesion microenvironment combining different therapies seem to be the most promising and proactive ones.
Collapse
Affiliation(s)
- Ana Carolina Correa de Assis
- Department of Medicine, Federal University of Juiz de Fora (UFJF-GV), 241 Manoel Byrro St., Governador Valadares, MG, 35032-620, Brazil
| | - Amanda Luiza Silva Reis
- Department of Medicine, Federal University of Juiz de Fora (UFJF-GV), 241 Manoel Byrro St., Governador Valadares, MG, 35032-620, Brazil
| | - Leonardo Vieira Nunes
- School of Medicine, Federal University of Juiz de Fora (UFJF-JF), Eugênio do Nascimento Avenue, Juiz de Fora, MG, 36038-330, Brazil
| | - Luiz Fernando Romanholo Ferreira
- Graduate Program in Process Engineering, Tiradentes University (UNIT), 300 Murilo Dantas Ave., Aracaju, SE, 49032-490, Brazil
- Institute of Technology and Research (ITP), Tiradentes University (UNIT), 300 Murilo Dantas Ave., Aracaju, SE, 49032-490, Brazil
| | - Muhammad Bilal
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huaian, 223003, China.
| | - Hafiz M N Iqbal
- Tecnologico de Monterrey, School of Engineering and Sciences, Campus Monterrey, Ave. Eugenio Garza Sada 2501, CP 64849, Monterrey, NL , Mexico
| | - Renato Nery Soriano
- Division of Physiology and Biophysics, Department of Basic Life Sciences, Federal University of Juiz de Fora (UFJF-GV), 1167 Moacir Paleta Ave., Governador Valadares, MG, 35020-360, Brazil.
| |
Collapse
|
47
|
Perrelle JM, Boreland AJ, Gamboa JM, Gowda P, Murthy NS. Biomimetic Strategies for Peripheral Nerve Injury Repair: An Exploration of Microarchitecture and Cellularization. BIOMEDICAL MATERIALS & DEVICES (NEW YORK, N.Y.) 2023; 1:21-37. [PMID: 38343513 PMCID: PMC10857769 DOI: 10.1007/s44174-022-00039-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 09/14/2022] [Indexed: 02/15/2024]
Abstract
Injuries to the nervous system present formidable challenges to scientists, clinicians, and patients. While regeneration within the central nervous system is minimal, peripheral nerves can regenerate, albeit with limitations. The regenerative mechanisms of the peripheral nervous system thus provide fertile ground for clinical and scientific advancement, and opportunities to learn fundamental lessons regarding nerve behavior in the context of regeneration, particularly the relationship of axons to their support cells and the extracellular matrix environment. However, few current interventions adequately address peripheral nerve injuries. This article aims to elucidate areas in which progress might be made toward developing better interventions, particularly using synthetic nerve grafts. The article first provides a thorough review of peripheral nerve anatomy, physiology, and the regenerative mechanisms that occur in response to injury. This is followed by a discussion of currently available interventions for peripheral nerve injuries. Promising biomaterial fabrication techniques which aim to recapitulate nerve architecture, along with approaches to enhancing these biomaterial scaffolds with growth factors and cellular components, are then described. The final section elucidates specific considerations when developing nerve grafts, including utilizing induced pluripotent stem cells, Schwann cells, nerve growth factors, and multilayered structures that mimic the architectures of the natural nerve.
Collapse
Affiliation(s)
- Jeremy M. Perrelle
- Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Andrew J. Boreland
- Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
- Graduate Program in Molecular Biosciences, Rutgers University, Piscataway, NJ, USA
| | - Jasmine M. Gamboa
- Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Prarthana Gowda
- Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - N. Sanjeeva Murthy
- Laboratory for Biomaterials Research, Department of Chemistry and Chemical Biology, Rutgers University, Piscataway, NJ 08854, USA
| |
Collapse
|
48
|
Li Y, Takanari K, Nakamura R, Kambe M, Ebisawa K, Oishi M, Kamei Y. Artificial PGA/Collagen-based Bilayer Conduit in Short Gap Interposition Setting Provides Comparable Regenerative Potential to Direct Suture. PLASTIC AND RECONSTRUCTIVE SURGERY-GLOBAL OPEN 2023; 11:e4875. [PMID: 36998535 PMCID: PMC10043553 DOI: 10.1097/gox.0000000000004875] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 01/27/2023] [Indexed: 03/29/2023]
Abstract
The aim of this study was to evaluate whether the Nerbridge, an artificial polyglycolic acid conduit with collagen matrix, is comparable to direct nerve suture in a rat sciatic nerve injury model in a short-gap interposition (SGI) setting. Methods Sixty-six female Lewis rats were randomly divided into the sham group (n = 13); no reconstruction (no-recon) group (n = 13; rat model with 10 mm sciatic nerve defect); direct group (n = 20; rat sciatic nerve injury directly connected by 10-0 Nylon); and SGI group (n = 20; sciatic nerve injury repaired using 5-mm Nerbridge). Motor function and histological recovery were evaluated. The sciatic nerve and gastrocnemius muscle were harvested for quantification of the degree of nerve regeneration and muscle atrophy. Results The SGI and direct groups achieved equal recovery in both functional and histological outcomes. At weeks 3 and 8 postsurgery, there was a significant improvement in the sciatic functional index of the SGI group when compared with that of the no-recon group (P < 0.05). Furthermore, the direct and SGI groups had less muscle atrophy at 4 and 8 weeks postsurgery compared with the no-recon group (P < 0.05). The axon density and diameter at the distal site in the SGI group were significantly higher than that in the no-recon group and comparable to that in the direct and sham groups. Conclusion An artificial nerve conduit has equal potential as direct suture in motor nerve reconstruction when used in the SGI setting.
Collapse
|
49
|
Tatu R, White LG, Yun Y, Hopkins T, An X, Ashraf A, Little KJ, Hershcovitch M, Hom DB, Pixley S. Effects of Altering Magnesium Metal Surfaces on Degradation In Vitro and In Vivo during Peripheral Nerve Regeneration. MATERIALS (BASEL, SWITZERLAND) 2023; 16:1195. [PMID: 36770202 PMCID: PMC9920421 DOI: 10.3390/ma16031195] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/23/2023] [Accepted: 01/25/2023] [Indexed: 06/18/2023]
Abstract
In vivo use of biodegradable magnesium (Mg) metal can be plagued by too rapid a degradation rate that removes metal support before physiological function is repaired. To advance the use of Mg biomedical implants, the degradation rate may need to be adjusted. We previously demonstrated that pure Mg filaments used in a nerve repair scaffold were compatible with regenerating peripheral nerve tissues, reduced inflammation, and improved axonal numbers across a short-but not long-gap in sciatic nerves in rats. To determine if the repair of longer gaps would be improved by a slower Mg degradation rate, we tested, in vitro and in vivo, the effects of Mg filament polishing followed by anodization using plasma electrolytic oxidation (PEO) with non-toxic electrolytes. Polishing removed oxidation products from the surface of as-received (unpolished) filaments, exposed more Mg on the surface, produced a smoother surface, slowed in vitro Mg degradation over four weeks after immersion in a physiological solution, and improved attachment of cultured epithelial cells. In vivo, treated Mg filaments were used to repair longer (15 mm) injury gaps in adult rat sciatic nerves after placement inside hollow poly (caprolactone) nerve conduits. The addition of single Mg or control titanium filaments was compared to empty conduits (negative control) and isografts (nerves from donor rats, positive control). After six weeks in vivo, live animal imaging with micro computed tomography (micro-CT) showed that Mg metal degradation rates were slowed by polishing vs. as-received Mg, but not by anodization, which introduced greater variability. After 14 weeks in vivo, functional return was seen only with isograft controls. However, within Mg filament groups, the amount of axonal growth across the injury site was improved with slower Mg degradation rates. Thus, anodization slowed degradation in vitro but not in vivo, and degradation rates do affect nerve regeneration.
Collapse
Affiliation(s)
- Rigwed Tatu
- Department of Pharmacology & Systems Physiology, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
- PSN Labs (Plastics Services Network), Erie, PA 16510, USA
| | - Leon G. White
- Department of Mechanical Engineering, Department of Bioengineering, North Carolina Agricultural & Technical State University, Greensboro, NC 27411, USA
- Northrop Grumman, Baltimore, MD 21240, USA
| | - Yeoheung Yun
- Department of Mechanical Engineering, Department of Bioengineering, North Carolina Agricultural & Technical State University, Greensboro, NC 27411, USA
| | - Tracy Hopkins
- Department of Pharmacology & Systems Physiology, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Xiaoxian An
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Ahmed Ashraf
- Department of Pharmacology & Systems Physiology, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Kevin J. Little
- College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Meir Hershcovitch
- College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - David B. Hom
- Department of Pharmacology & Systems Physiology, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
- Head & Neck Surgery, San Diego Medical Center, University of California, San Diego, CA 92103, USA
| | - Sarah Pixley
- Department of Pharmacology & Systems Physiology, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| |
Collapse
|
50
|
Babanejad N, Mfoafo K, Thumma A, Omidi Y, Omidian H. Advances in cryostructures and their applications in biomedical and pharmaceutical products. Polym Bull (Berl) 2023. [DOI: 10.1007/s00289-023-04683-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|