1
|
Zhang C, Zhang J, Xie D, Guo G, Jalili S. The effects of local delivery of laurus nobilis extract and adipose derived stem cells via electrospun gelatin scaffold on spinal cord injury inflammatoradscy response and its regeneration. Regen Ther 2024; 26:879-888. [PMID: 39444416 PMCID: PMC11497134 DOI: 10.1016/j.reth.2024.09.008] [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: 08/20/2024] [Revised: 09/03/2024] [Accepted: 09/23/2024] [Indexed: 10/25/2024] Open
Abstract
When subjected to injury, the spinal cord's inherent complexity poses significant challenges for effective healing. In this study, gelatin nanofibers loaded with Laurus nobilis extract were developed to serve as a delivery system for adipose-derived stem cells (ADSCs), aiming to explore its potential immunomodulatory effects in a rat model of spinal cord injury. Through a series of in vitro assessments including scanning electron microscopy imaging, cell viability, anti-inflammatory, cell adhesion, biodegradation, and hemocompatibility assays, the characteristics of the delivery system were thoroughly evaluated. The in vitro studies revealed both the biocompatibility of the scaffolds and their notable anti-inflammatory properties, laying the groundwork for further investigation. Subsequent in vivo experiments demonstrated that rats treated with Laurus nobilis extract and ADSCs loaded scaffolds exhibited heightened functional recovery (BBB score of 14.66 ± 1.52 % and hot plate latency time of 8.33 0.26 s) and histological restoration at the 8-week mark post-injury. Notably, ELISA assay results revealed a significant reduction in tissue expression levels of key pro-inflammatory cytokines, including TNF-α, IL-1β, and IL-6, suggesting a pronounced immunomodulatory effect of the Laurus nobilis extract-loaded scaffolds. The findings underscore the potential of this novel delivery system to improve clinical outcomes in spinal cord injury by enhancing functional recovery and reducing inflammation. This approach could lead to the development of new, natural-based therapeutic strategies for spinal cord injury, with potential extensions to other inflammatory or degenerative conditions. Future research should focus on optimizing this strategy in larger animal models and eventually translating these findings into human clinical trials.
Collapse
Affiliation(s)
- Chong Zhang
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Jin Zhang
- Department of Emergency Traumatology, Gejiu People's Hospital, Yunnan, 661000, China
| | - Daotao Xie
- Norxin International Science and Technology Base, Xi'an, 710032, China
| | - Gang Guo
- Norxin International Science and Technology Base, Xi'an, 710032, China
| | - Saman Jalili
- Department of Materials Science, Isfahan University of Technology, Isfahan, Iran
| |
Collapse
|
2
|
Banaeeyeh S, Afkhami-Goli A, Moosavi Z, Razavi BM, Hosseinzadeh H. Anti-inflammatory, antioxidant and anti-mitophagy effects of trans sodium crocetinate on experimental autoimmune encephalomyelitis in BALB/C57 mice. Metab Brain Dis 2024; 39:783-801. [PMID: 38739183 DOI: 10.1007/s11011-024-01349-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 05/04/2024] [Indexed: 05/14/2024]
Abstract
Multiple sclerosis (MS) is an autoimmune disorder characterized by the degeneration of myelin and inflammation in the central nervous system. Trans sodium crocetinate (TSC), a novel synthetic carotenoid compound, possesses antioxidant, anti-inflammatory and neuroprotective effects. This study aimed to evaluate the protective effects of TSC against the development of experimental autoimmune encephalomyelitis (EAE), a well-established model for MS. Female BALB/C57 mice were divided into different groups, including control, EAE, vehicle, TSC-treated (25, 50, and 100 mg/kg, administered via gavage) + EAE, methyl prednisone acetate + EAE, and TSC-treated (100 mg/kg, administered via gavage for 28 days) groups. EAE was induced using MOG35-55, complete Freund's adjuvant, and pertussis toxin. In the mice spinal cord tissues, the oxidative markers (GSH and MDA) were measured using spectrophotometry and histological evaluation was performed. Mitophagic pathway proteins (PINK1and PARKIN) and inflammatory factors (IL-1β and TNF-α) were evaluated by western blot. Following 21 days post-induction, EAE mice exhibited weight loss, and the paralysis scores increased on day 13 but recovered after TSC (100 mg/kg) administration on day 16. Furthermore, TSC (50 and 100 mg/kg) reversed the altered levels of MDA and GSH in the spinal cord tissue of EAE mice. TSC (100 mg/kg) also decreased microgliosis, demyelination, and the levels of inflammatory markers IL-1β and TNF-α. Notably, TSC (100 mg/kg) modulated the mitophagy pathway by reducing PINK1 and Parkin protein levels. These findings demonstrate that TSC protects spinal cord tissue against EAE-induced MS through anti-inflammatory, antioxidant, and anti-mitophagy mechanisms.
Collapse
Affiliation(s)
- Sara Banaeeyeh
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Afkhami-Goli
- Division of Pharmacology, Department of Basic Sciences, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
- Stem Cell Biology and Regenerative Medicine Research Group, Research Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Zahra Moosavi
- Department of Pathobiology, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Bibi Marjan Razavi
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hossein Hosseinzadeh
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
3
|
Wang Z, Li J, Xu T, Guo B, Xie Z, Li M. The Efficacy of Different Material Scaffold-Guided Cell Transplantation in the Treatment of Spinal Cord Injury in Rats: A Systematic Review and Network Meta-analysis. Cell Mol Neurobiol 2024; 44:43. [PMID: 38703332 PMCID: PMC11069479 DOI: 10.1007/s10571-024-01465-6] [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: 12/17/2023] [Accepted: 02/23/2024] [Indexed: 05/06/2024]
Abstract
Cell transplantation is a promising treatment option for spinal cord injury (SCI). However, there is no consensus on the choice of carrier scaffolds to host the cells. This study aims to evaluate the efficacy of different material scaffold-mediated cell transplantation in treating SCI in rats. According to PRISMA's principle, Embase, PubMed, Web of Science, and Cochrane databases were searched, and relevant literature was referenced. Only original research on cell transplantation plus natural or synthetic scaffolds in SCI rats was included. Direct and indirect evidence for improving hind limb motor function was pooled through meta-analysis. A subgroup analysis of some factors that may affect the therapeutic effect was conducted to understand the results fully. In total, 25 studies met the inclusion criteria, in which 293 rats received sham surgery, 78 rats received synthetic material scaffolds, and 219 rats received natural materials scaffolds. The network meta-analysis demonstrated that although synthetic scaffolds were slightly inferior to natural scaffolds in terms of restoring motor function in cell transplantation of SCI rats, no statistical differences were observed between the two (MD: -0.35; 95% CI -2.6 to 1.9). Moreover, the subgroup analysis revealed that the type and number of cells may be important factors in therapeutic efficacy (P < 0.01). Natural scaffolds and synthetic scaffolds are equally effective in cell transplantation of SCI rats without significant differences. In the future, the findings need to be validated in multicenter, large-scale, randomized controlled trials in clinical practice. Trial registration: Registration ID CRD42024459674 (PROSPERO).
Collapse
Affiliation(s)
- Zhihua Wang
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, No.17, Yongwai Street, Nanchang, 330006, Jiangxi Province, China
- Postdoctoral Innovation Practice Base, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Jun Li
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, No.17, Yongwai Street, Nanchang, 330006, Jiangxi Province, China
- Department of the Second Clinical Medical College of Nanchang University, No.460, BaYi Street, Nanchang, 330006, Jiangxi Province, China
| | - Tianqi Xu
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, No.17, Yongwai Street, Nanchang, 330006, Jiangxi Province, China
- Department of the Second Clinical Medical College of Nanchang University, No.460, BaYi Street, Nanchang, 330006, Jiangxi Province, China
| | - Boyu Guo
- Department of the First Clinical Medical College of Nanchang University, No.460, BaYi Street, Nanchang, 330006, Jiangxi Province, China
| | - Zhiping Xie
- Department of Neurosurgery, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, No.152 Aiguo Road, Nanchang, 330006, Jiangxi Province, China.
- Department of Neurosurgery, Xiangya Hospital Jiangxi Hospital, Central South University, Nanchang, Jiangxi Province, China.
| | - Meihua Li
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, No.17, Yongwai Street, Nanchang, 330006, Jiangxi Province, China.
| |
Collapse
|
4
|
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: 34] [Impact Index Per Article: 34.0] [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
|
5
|
Giorgi Z, Veneruso V, Petillo E, Veglianese P, Perale G, Rossi F. Biomaterials and Cell Therapy Combination in Central Nervous System Treatments. ACS APPLIED BIO MATERIALS 2024; 7:80-98. [PMID: 38158393 PMCID: PMC10792669 DOI: 10.1021/acsabm.3c01058] [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: 11/09/2023] [Revised: 12/07/2023] [Accepted: 12/11/2023] [Indexed: 01/03/2024]
Abstract
Current pharmacological and surgical therapies for the central nervous system (CNS) show a limited capacity to reduce the damage progression; that together with the intrinsic limited capability of the CNS to regenerate greatly reduces the hopes of recovery. Among all the therapies proposed, the tissue engineering strategies supplemented with therapeutic stem cells remain the most promising. Neural tissue engineering strategies are based on the development of devices presenting optimal physical, chemical, and mechanical properties which, once inserted in the injured site, can support therapeutic cells, limiting the effect of a hostile environment and supporting regenerative processes. Thus, this review focuses on the employment of hydrogel and nanofibrous scaffolds supplemented with stem cells as promising therapeutic tools for the central and peripheral nervous systems in preclinical and clinical applications.
Collapse
Affiliation(s)
- Zoe Giorgi
- Department
of Chemistry, Materials and Chemical Engineering “Giulio Natta”, Politecnico di Milano, piazza Leonardo da Vinci 32, 20133, Milan, Italy
| | - Valeria Veneruso
- Istituto
di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milan, Italy
- Faculty
of Biomedical Sciences, University of Southern
Switzerland (USI), Via
Buffi 13, 6900 Lugano, Switzerland
| | - Emilia Petillo
- Department
of Chemistry, Materials and Chemical Engineering “Giulio Natta”, Politecnico di Milano, piazza Leonardo da Vinci 32, 20133, Milan, Italy
- Istituto
di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milan, Italy
| | - Pietro Veglianese
- Istituto
di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milan, Italy
- Faculty
of Biomedical Sciences, University of Southern
Switzerland (USI), Via
Buffi 13, 6900 Lugano, Switzerland
| | - Giuseppe Perale
- Faculty
of Biomedical Sciences, University of Southern
Switzerland (USI), Via
Buffi 13, 6900 Lugano, Switzerland
- Ludwig
Boltzmann Institute for Experimental and Clinical Traumatology, Donaueschingenstrasse 13, 1200 Vienna, Austria
| | - Filippo Rossi
- Department
of Chemistry, Materials and Chemical Engineering “Giulio Natta”, Politecnico di Milano, piazza Leonardo da Vinci 32, 20133, Milan, Italy
- Faculty
of Biomedical Sciences, University of Southern
Switzerland (USI), Via
Buffi 13, 6900 Lugano, Switzerland
| |
Collapse
|
6
|
Yan B, Hua Y, Wang J, Shao T, Wang S, Gao X, Gao J. Surface Modification Progress for PLGA-Based Cell Scaffolds. Polymers (Basel) 2024; 16:165. [PMID: 38201830 PMCID: PMC10780542 DOI: 10.3390/polym16010165] [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: 12/04/2023] [Revised: 12/28/2023] [Accepted: 12/29/2023] [Indexed: 01/12/2024] Open
Abstract
Poly(lactic-glycolic acid) (PLGA) is a biocompatible bio-scaffold material, but its own hydrophobic and electrically neutral surface limits its application as a cell scaffold. Polymer materials, mimics ECM materials, and organic material have often been used as coating materials for PLGA cell scaffolds to improve the poor cell adhesion of PLGA and enhance tissue adaptation. These coating materials can be modified on the PLGA surface via simple physical or chemical methods, and coating multiple materials can simultaneously confer different functions to the PLGA scaffold; not only does this ensure stronger cell adhesion but it also modulates cell behavior and function. This approach to coating could facilitate the production of more PLGA-based cell scaffolds. This review focuses on the PLGA surface-modified materials, methods, and applications, and will provide guidance for PLGA surface modification.
Collapse
Affiliation(s)
- Bohua Yan
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; (B.Y.); (J.W.); (T.S.); (S.W.)
| | - Yabing Hua
- Department of Pharmacy, Xuzhou Medical University Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, China;
| | - Jinyue Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; (B.Y.); (J.W.); (T.S.); (S.W.)
| | - Tianjiao Shao
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; (B.Y.); (J.W.); (T.S.); (S.W.)
| | - Shan Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; (B.Y.); (J.W.); (T.S.); (S.W.)
| | - Xiang Gao
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; (B.Y.); (J.W.); (T.S.); (S.W.)
| | - Jing Gao
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; (B.Y.); (J.W.); (T.S.); (S.W.)
| |
Collapse
|
7
|
Mahdian M, Tabatabai TS, Abpeikar Z, Rezakhani L, Khazaei M. Nerve regeneration using decellularized tissues: challenges and opportunities. Front Neurosci 2023; 17:1295563. [PMID: 37928728 PMCID: PMC10620322 DOI: 10.3389/fnins.2023.1295563] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Accepted: 10/06/2023] [Indexed: 11/07/2023] Open
Abstract
In tissue engineering, the decellularization of organs and tissues as a biological scaffold plays a critical role in the repair of neurodegenerative diseases. Various protocols for cell removal can distinguish the effects of treatment ability, tissue structure, and extracellular matrix (ECM) ability. Despite considerable progress in nerve regeneration and functional recovery, the slow regeneration and recovery potential of the central nervous system (CNS) remains a challenge. The success of neural tissue engineering is primarily influenced by composition, microstructure, and mechanical properties. The primary objective of restorative techniques is to guide existing axons properly toward the distal end of the damaged nerve and the target organs. However, due to the limitations of nerve autografts, researchers are seeking alternative methods with high therapeutic efficiency and without the limitations of autograft transplantation. Decellularization scaffolds, due to their lack of immunogenicity and the preservation of essential factors in the ECM and high angiogenic ability, provide a suitable three-dimensional (3D) substrate for the adhesion and growth of axons being repaired toward the target organs. This study focuses on mentioning the types of scaffolds used in nerve regeneration, and the methods of tissue decellularization, and specifically explores the use of decellularized nerve tissues (DNT) for nerve transplantation.
Collapse
Affiliation(s)
- Maryam Mahdian
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Tayebeh Sadat Tabatabai
- Student Research Committee, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Zahra Abpeikar
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Fasa University of Medical Sciences, Fasa, 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
| | - 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
| |
Collapse
|
8
|
Li Y, Meng Q, Chen S, Ling P, Kuss MA, Duan B, Wu S. Advances, challenges, and prospects for surgical suture materials. Acta Biomater 2023; 168:78-112. [PMID: 37516417 DOI: 10.1016/j.actbio.2023.07.041] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 07/07/2023] [Accepted: 07/24/2023] [Indexed: 07/31/2023]
Abstract
As one of the long-established and necessary medical devices, surgical sutures play an essentially important role in the closing and healing of damaged tissues and organs postoperatively. The recent advances in multiple disciplines, like materials science, engineering technology, and biomedicine, have facilitated the generation of various innovative surgical sutures with humanization and multi-functionalization. For instance, the application of numerous absorbable materials is assuredly a marvelous progression in terms of surgical sutures. Moreover, some fantastic results from recent laboratory research cannot be ignored either, ranging from the fiber generation to the suture structure, as well as the suture modification, functionalization, and even intellectualization. In this review, the suture materials, including natural or synthetic polymers, absorbable or non-absorbable polymers, and metal materials, were first introduced, and then their advantages and disadvantages were summarized. Then we introduced and discussed various fiber fabrication strategies for the production of surgical sutures. Noticeably, advanced nanofiber generation strategies were highlighted. This review further summarized a wide and diverse variety of suture structures and further discussed their different features. After that, we covered the advanced design and development of surgical sutures with multiple functionalizations, which mainly included surface coating technologies and direct drug-loading technologies. Meanwhile, the review highlighted some smart and intelligent sutures that can monitor the wound status in a real-time manner and provide on-demand therapies accordingly. Furthermore, some representative commercial sutures were also introduced and summarized. At the end of this review, we discussed the challenges and future prospects in the field of surgical sutures in depth. This review aims to provide a meaningful reference and guidance for the future design and fabrication of innovative surgical sutures. STATEMENT OF SIGNIFICANCE: This review article introduces the recent advances of surgical sutures, including material selection, fiber morphology, suture structure and construction, as well as suture modification, functionalization, and even intellectualization. Importantly, some innovative strategies for the construction of multifunctional sutures with predetermined biological properties are highlighted. Moreover, some important commercial suture products are systematically summarized and compared. This review also discusses the challenges and future prospects of advanced sutures in a deep manner. In all, this review is expected to arouse great interest from a broad group of readers in the fields of multifunctional biomaterials and regenerative medicine.
Collapse
Affiliation(s)
- Yiran Li
- College of Textiles & Clothing, Qingdao University, Qingdao, 266071, China
| | - Qi Meng
- College of Textiles & Clothing, Qingdao University, Qingdao, 266071, China
| | - Shaojuan Chen
- College of Textiles & Clothing, Qingdao University, Qingdao, 266071, China
| | - Peixue Ling
- Shandong Academy of Pharmaceutical Science, Jinan, 250101, China
| | - Mitchell A Kuss
- Mary & Dick Holland Regenerative Medicine Program and Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Bin Duan
- Mary & Dick Holland Regenerative Medicine Program and Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Shaohua Wu
- College of Textiles & Clothing, Qingdao University, Qingdao, 266071, China; Shandong Academy of Pharmaceutical Science, Jinan, 250101, China.
| |
Collapse
|
9
|
Xin W, Baokun Z, Zhiheng C, Qiang S, Erzhu Y, Jianguang X, Xiaofeng L. Biodegradable bilayer hydrogel membranes loaded with bazedoxifene attenuate blood-spinal cord barrier disruption via the NF-κB pathway after acute spinal cord injury. Acta Biomater 2023; 159:140-155. [PMID: 36736849 DOI: 10.1016/j.actbio.2023.01.056] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 01/25/2023] [Accepted: 01/26/2023] [Indexed: 02/04/2023]
Abstract
After spinal cord injury (SCI), blood-spinal cord barrier (BSCB) disruption and hemorrhage lead to blood cell infiltration and progressive secondary injuries. Therefore, early restoration of the BSCB represents a key step in the treatment of SCI. Bazedoxifene (BZA), a third-generation estrogen receptor modulator, has recently been reported to inhibit inflammation and alleviate blood-brain barrier disruption caused by traumatic brain injury, attracting great interest in the field of central nervous system injury and repair. However, whether BZA can attenuate BSCB disruption and contribute to SCI repair remains unknown. Here, we developed a new type of biomaterial carrier and constructed a BZA-loaded HSPT (hyaluronic acid (HA), sodium alginate (SA), polyvinyl alcohol (PVA), tetramethylpropane (TPA) material construction) (HSPT@Be) system to effectively deliver BZA to the site of SCI. We found that HSPT@Be could significantly reduce inflammation in the spinal cord in SCI rats and attenuate BSCB disruption by providing covering scaffold, inhibiting oxidative stress, and upregulating tight junction proteins, which was mediated by regulation of the NF-κB/MMP signaling pathway. Importantly, functional assessment showed the evident improvement of behavioral functions in the HSPT@Be-treated SCI rats. These results indicated that HSPT@Be can attenuate BSCB disruption via the NF-κB pathway after SCI, shedding light on its potential therapeutic benefit for SCI. STATEMENT OF SIGNIFICANCE: After spinal cord injury, blood-spinal cord barrier disruption and hemorrhage lead to blood cell infiltration and progressive secondary injuries. Bazedoxifene has recently been reported to inhibit inflammation and alleviate blood-brain barrier disruption caused by traumatic brain injury. However, whether BZA can attenuate BSCB disruption and contribute to SCI repair remains unknown. In this study, we developed a new type of biomaterial carrier and constructed a bazedoxifene-loaded HSPT (HSPT@Be) system to efficiently treat SCI. HSPT@Be could provide protective coverage, inhibit oxidative stress, and upregulate tight junction proteins through NF-κB/MMP pathway both in vivo and in vitro, therefore attenuating BSCB disruption. Our study fills the application gap of biomaterials in BSCB restoration.
Collapse
Affiliation(s)
- Wang Xin
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Zhang Baokun
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Chen Zhiheng
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Shi Qiang
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Yang Erzhu
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Xu Jianguang
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China.
| | - Lian Xiaofeng
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China.
| |
Collapse
|
10
|
Feng C, Deng L, Yong YY, Wu JM, Qin DL, Yu L, Zhou XG, Wu AG. The Application of Biomaterials in Spinal Cord Injury. Int J Mol Sci 2023; 24:816. [PMID: 36614259 PMCID: PMC9821025 DOI: 10.3390/ijms24010816] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 12/19/2022] [Accepted: 12/30/2022] [Indexed: 01/05/2023] Open
Abstract
The spinal cord and the brain form the central nervous system (CNS), which is the most important part of the body. However, spinal cord injury (SCI) caused by external forces is one of the most difficult types of neurological injury to treat, resulting in reduced or even absent motor, sensory and autonomic functions. It leads to the reduction or even disappearance of motor, sensory and self-organizing nerve functions. Currently, its incidence is increasing each year worldwide. Therefore, the development of treatments for SCI is urgently needed in the clinic. To date, surgery, drug therapy, stem cell transplantation, regenerative medicine, and rehabilitation therapy have been developed for the treatment of SCI. Among them, regenerative biomaterials that use tissue engineering and bioscaffolds to transport cells or drugs to the injured site are considered the most promising option. In this review, we briefly introduce SCI and its molecular mechanism and summarize the application of biomaterials in the repair and regeneration of tissue in various models of SCI. However, there is still limited evidence about the treatment of SCI with biomaterials in the clinic. Finally, this review will provide inspiration and direction for the future study and application of biomaterials in the treatment of SCI.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Xiao-Gang Zhou
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - An-Guo Wu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| |
Collapse
|
11
|
Sellahewa SG, Li JY, Xiao Q. Updated Perspectives on Direct Vascular Cellular Reprogramming and Their Potential Applications in Tissue Engineered Vascular Grafts. J Funct Biomater 2022; 14:21. [PMID: 36662068 PMCID: PMC9866165 DOI: 10.3390/jfb14010021] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 12/25/2022] [Accepted: 12/27/2022] [Indexed: 01/03/2023] Open
Abstract
Cardiovascular disease is a globally prevalent disease with far-reaching medical and socio-economic consequences. Although improvements in treatment pathways and revascularisation therapies have slowed disease progression, contemporary management fails to modulate the underlying atherosclerotic process and sustainably replace damaged arterial tissue. Direct cellular reprogramming is a rapidly evolving and innovative tissue regenerative approach that holds promise to restore functional vasculature and restore blood perfusion. The approach utilises cell plasticity to directly convert somatic cells to another cell fate without a pluripotent stage. In this narrative literature review, we comprehensively analyse and compare direct reprogramming protocols to generate endothelial cells, vascular smooth muscle cells and vascular progenitors. Specifically, we carefully examine the reprogramming factors, their molecular mechanisms, conversion efficacies and therapeutic benefits for each induced vascular cell. Attention is given to the application of these novel approaches with tissue engineered vascular grafts as a therapeutic and disease-modelling platform for cardiovascular diseases. We conclude with a discussion on the ethics of direct reprogramming, its current challenges, and future perspectives.
Collapse
Affiliation(s)
- Saneth Gavishka Sellahewa
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Jojo Yijiao Li
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Qingzhong Xiao
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
- Key Laboratory of Cardiovascular Diseases, School of Basic Medical Sciences, Guangzhou Institute of Cardiovascular Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| |
Collapse
|
12
|
Abadi B, Goshtasbi N, Bolourian S, Tahsili J, Adeli-Sardou M, Forootanfar H. Electrospun hybrid nanofibers: Fabrication, characterization, and biomedical applications. Front Bioeng Biotechnol 2022; 10:986975. [PMID: 36561047 PMCID: PMC9764016 DOI: 10.3389/fbioe.2022.986975] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 11/16/2022] [Indexed: 12/05/2022] Open
Abstract
Nanotechnology is one of the most promising technologies available today, holding tremendous potential for biomedical and healthcare applications. In this field, there is an increasing interest in the use of polymeric micro/nanofibers for the construction of biomedical structures. Due to its potential applications in various fields like pharmaceutics and biomedicine, the electrospinning process has gained considerable attention for producing nano-sized fibers. Electrospun nanofiber membranes have been used in drug delivery, controlled drug release, regenerative medicine, tissue engineering, biosensing, stent coating, implants, cosmetics, facial masks, and theranostics. Various natural and synthetic polymers have been successfully electrospun into ultrafine fibers. Although biopolymers demonstrate exciting properties such as good biocompatibility, non-toxicity, and biodegradability, they possess poor mechanical properties. Hybrid nanofibers from bio and synthetic nanofibers combine the characteristics of biopolymers with those of synthetic polymers, such as high mechanical strength and stability. In addition, a variety of functional agents, such as nanoparticles and biomolecules, can be incorporated into nanofibers to create multifunctional hybrid nanofibers. Due to the remarkable properties of hybrid nanofibers, the latest research on the unique properties of hybrid nanofibers is highlighted in this study. Moreover, various established hybrid nanofiber fabrication techniques, especially the electrospinning-based methods, as well as emerging strategies for the characterization of hybrid nanofibers, are summarized. Finally, the development and application of electrospun hybrid nanofibers in biomedical applications are discussed.
Collapse
Affiliation(s)
- Banafshe Abadi
- Herbal and Traditional Medicines Research Center, Kerman University of Medical Sciences, Kerman, Iran,Brain Cancer Research Core (BCRC), Universal Scientific Education and Research Network (USERN), Kerman, Iran
| | - Nazanin Goshtasbi
- Department of Pharmaceutics, Faculty of Pharmacy and Pharmaceutical Sciences, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Saman Bolourian
- Department of Biology, Faculty of Science, Alzahra University, Tehran, Iran
| | - Jaleh Tahsili
- Department of Plant Biology, Faculty of Biological Science, Tarbiat Modares University, Tehran, Iran
| | - Mahboubeh Adeli-Sardou
- Medical Mycology and Bacteriology Research Center, Kerman University of Medical Sciences, Kerman, Iran,Student Research Committee, Kerman University of Medical Sciences, Kerman, Iran,*Correspondence: Mahboubeh Adeli-Sardou, ; Hamid Forootanfar,
| | - Hamid Forootanfar
- Pharmaceutical Sciences and Cosmetic Products Research Center, Kerman University of Medical Sciences, Kerman, Iran,Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran,*Correspondence: Mahboubeh Adeli-Sardou, ; Hamid Forootanfar,
| |
Collapse
|
13
|
Yousefifard M, Askarian-Amiri S, Nasseri Maleki S, Rafiei Alavi SN, Madani Neishaboori A, Haghani L, Vaccaro AR, Harrop JS, Lu Y, Rahimi-Movaghar V, Hosseini M. Combined application of neural stem/progenitor cells and scaffolds on locomotion recovery following spinal cord injury in rodents: a systematic review and meta-analysis. Neurosurg Rev 2022; 45:3469-3488. [DOI: 10.1007/s10143-022-01859-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 07/20/2022] [Accepted: 09/01/2022] [Indexed: 11/29/2022]
|
14
|
Asghari Niari S, Rahbarghazi R, Salehi R, Kazemi L, Fathi Karkan S, Karimipour M. Fabrication, characterization and evaluation of the effect of PLGA and PLGA-PEG biomaterials on the proliferation and neurogenesis potential of human neural SH-SY5Y cells. Microsc Res Tech 2021; 85:1433-1443. [PMID: 34859937 DOI: 10.1002/jemt.24006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 10/26/2021] [Accepted: 11/05/2021] [Indexed: 12/18/2022]
Abstract
In recent years with regard to the development of nanotechnology and neural stem cell discovery, the combinatorial therapeutic strategies of neural progenitor cells and appropriate biomaterials have raised the hope for brain regeneration following neurological disorders. This study aimed to explore the proliferation and neurogenic effect of PLGA and PLGA-PEG nanofibers on human SH-SY5Y cells in in vitro condition. Nanofibers of PLGA and PLGA-PEG biomaterials were synthesized and fabricated using electrospinning method. Physicochemical features were examined using HNMR, FT-IR, and water contact angle assays. Ultrastructural morphology, the orientation of nanofibers, cell distribution and attachment were visualized by SEM imaging. Cell survival and proliferation rate were measured. Differentiation capacity was monitored by immunofluorescence staining of Map-2. HNMR, FT-IR assays confirmed the integration of PEG to PLGA backbone. Water contact angel assay showed increasing surface hydrophilicity in PLGA-PEG biomaterial compared to the PLGA substrate. SEM analysis revealed the reduction of PLGA-PEG nanofibers' diameter compared to the PLGA group. Cell attachment was observed in both groups while PLGA-PEG had a superior effect in the promotion of survival rate compared to other groups (p < .05). Compared to the PLGA group, PLGA-PEG increased the number of Ki67+ cells (p < .01). PLGA-PEG biomaterial induced neural maturation by increasing protein Map-2 compared to the PLGA scaffold in a three-dimensional culture system. According to our data, structural modification of PLGA with PEG could enhance orientated differentiation and the dynamic growth of neural cells.
Collapse
Affiliation(s)
- Shabnam Asghari Niari
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Roya Salehi
- Department of Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Leila Kazemi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sonia Fathi Karkan
- Department of Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Karimipour
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
15
|
Niari SA, Rahbarghazi R, Geranmayeh MH, Karimipour M. Biomaterials patterning regulates neural stem cells fate and behavior: The interface of biology and material science. J Biomed Mater Res A 2021; 110:725-737. [PMID: 34751503 DOI: 10.1002/jbm.a.37321] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 09/19/2021] [Accepted: 10/06/2021] [Indexed: 11/12/2022]
Abstract
The combination of nanotechnology and stem cell biology is one of the most promising advances in the field of regenerative medicine. This novel combination has widely been utilized in vitro settings in an attempt to develop efficient therapeutic strategies to overcome the limited capacity of the central nervous system (CNS) in replacing degenerating neural cells with functionally normal cells after the onset of acute and chronic neurological disorders. Importantly, biomaterials, not only, enhance the endogenous CNS neurogenesis and plasticity, but also, could provide a desirable supportive microenvironment to harness the full potential of the in vitro expanded neural stem cells (NSCs) for regenerative purposes. Here, first, we discuss how the physical and biochemical properties of biomaterials, such as their stiffness and elasticity, could influence the behavior of NSCs. Then, since the NSCs niche or microenvironment is of fundamental importance in controlling the dynamic destiny of NSCs such as their quiescent and proliferative states, topographical effects of surface diversity in biomaterials, that is, the micro-and nano-patterned surfaces will be discussed in detail. Finally, the influence of biomaterials as artificial microenvironments on the behavior of NSCs through the specific mechanotransduction signaling pathway mediated by focal adhesion formation will be reviewed.
Collapse
Affiliation(s)
- Shabnam Asghari Niari
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Hossein Geranmayeh
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.,Neurosciences Research Center (NSRC), Imam Reza Medical Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Karimipour
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
16
|
Pinho TS, Cunha CB, Lanceros-Méndez S, Salgado AJ. Electroactive Smart Materials for Neural Tissue Regeneration. ACS APPLIED BIO MATERIALS 2021; 4:6604-6618. [PMID: 35006964 DOI: 10.1021/acsabm.1c00567] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Repair in the human nervous system is a complex and intertwined process that offers significant challenges to its study and comprehension. Taking advantage of the progress in fields such as tissue engineering and regenerative medicine, the scientific community has witnessed a strong increase of biomaterial-based approaches for neural tissue regenerative therapies. Electroactive materials, increasingly being used as sensors and actuators, also find application in neurosciences due to their ability to deliver electrical signals to the cells and tissues. The use of electrical signals for repairing impaired neural tissue therefore presents an interesting and innovative approach to bridge the gap between fundamental research and clinical applications in the next few years. In this review, first a general overview of electroactive materials, their historical origin, and characteristics are presented. Then a comprehensive view of the applications of electroactive smart materials for neural tissue regeneration is presented, with particular focus on the context of spinal cord injury and brain repair. Finally, the major challenges of the field are discussed and the main challenges for the near future presented. Overall, it is concluded that electroactive smart materials play an ever-increasing role in neural tissue regeneration, appearing as potentially valuable biomaterials for regenerative purposes.
Collapse
Affiliation(s)
- Tiffany S Pinho
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, 4710-057/4805-017 Braga/Guimarães, Portugal.,Stemmatters, Biotecnologia e Medicina Regenerativa SA, 4805-017 Guimarães, Portugal
| | - Cristiana B Cunha
- Stemmatters, Biotecnologia e Medicina Regenerativa SA, 4805-017 Guimarães, Portugal
| | - Senentxu Lanceros-Méndez
- Center of Physics, University of Minho, 4710-058 Braga, Portugal.,BCMaterials, Basque Center for Materials, Applications and Nanostructures, UPV/EHU Science Park, 48940 Leioa, Spain.,Ikerbasque, Basque Foundation for Science, 48009 Bilbao, Spain
| | - António J Salgado
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, 4710-057/4805-017 Braga/Guimarães, Portugal
| |
Collapse
|
17
|
Mousa AH, Agha Mohammad S, Rezk HM, Muzaffar KH, Alshanberi AM, Ansari SA. Nanoparticles in traumatic spinal cord injury: therapy and diagnosis. F1000Res 2021. [DOI: 10.12688/f1000research.55472.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Nanotechnology has been previously employed for constructing drug delivery vehicles, biosensors, solar cells, lubricants and as antimicrobial agents. The advancement in synthesis procedure makes it possible to formulate nanoparticles (NPs) with precise control over physico-chemical and optical properties that are desired for specific clinical or biological applications. The surface modification technology has further added impetus to the specific applications of NPs by providing them with desirable characteristics. Hence, nanotechnology is of paramount importance in numerous biomedical and industrial applications due to their biocompatibility and stability even in harsh environments. Traumatic spinal cord injuries (TSCIs) are one of the major traumatic injuries that are commonly associated with severe consequences to the patient that may reach to the point of paralysis. Several processes occurring at a biochemical level which exacerbate the injury may be targeted using nanotechnology. This review discusses possible nanotechnology-based approaches for the diagnosis and therapy of TSCI, which have a bright future in clinical practice.
Collapse
|
18
|
Gilmour AD, Reshamwala R, Wright AA, Ekberg JAK, St John JA. Optimizing Olfactory Ensheathing Cell Transplantation for Spinal Cord Injury Repair. J Neurotrauma 2021; 37:817-829. [PMID: 32056492 DOI: 10.1089/neu.2019.6939] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Cell transplantation constitutes an important avenue for development of new treatments for spinal cord injury (SCI). These therapies are aimed at supporting neural repair and/or replacing lost cells at the injury site. To date, various cell types have been trialed, with most studies focusing on different types of stem cells or glial cells. Here, we review commonly used cell transplantation approaches for spinal cord injury (SCI) repair, with focus on transplantation of olfactory ensheathing cells (OECs), the glial cells of the primary olfactory nervous system. OECs are promising candidates for promotion of neural repair given that they support continuous regeneration of the olfactory nerve that occurs throughout life. Further, OECs can be accessed from the nasal mucosa (olfactory neuroepithelium) at the roof of the nasal cavity and can be autologously transplanted. OEC transplantation has been trialed in many animal models of SCI, as well as in human clinical trials. While several studies have been promising, outcomes are variable and the method needs improvement to enhance aspects such as cell survival, integration, and migration. As a case study, we include the approaches used by our team (the Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Nathan, QLD, Australia) to address the current problems with OEC transplantation and discuss how the therapeutic potential of OEC transplantation can be improved. Our approach includes discovery research to improve our knowledge of OEC biology, identifying natural and synthetic compounds to stimulate the neural repair properties of OECs, and designing three-dimensional cell constructs to create stable and transplantable cell structures.
Collapse
Affiliation(s)
- Aaron D Gilmour
- Clem Jones Centre for Neurobiology and Stem Cell Research and Griffith University, Nathan, Queensland, Australia.,Menzies Health Institute Queensland, Griffith University, Southport, Queensland, Australia
| | - Ronak Reshamwala
- Clem Jones Centre for Neurobiology and Stem Cell Research and Griffith University, Nathan, Queensland, Australia.,Menzies Health Institute Queensland, Griffith University, Southport, Queensland, Australia.,Griffith Institute for Drug Discovery, Griffith University, Nathan, Queensland, Australia
| | - Alison A Wright
- Clem Jones Centre for Neurobiology and Stem Cell Research and Griffith University, Nathan, Queensland, Australia.,Menzies Health Institute Queensland, Griffith University, Southport, Queensland, Australia
| | - Jenny A K Ekberg
- Clem Jones Centre for Neurobiology and Stem Cell Research and Griffith University, Nathan, Queensland, Australia.,Menzies Health Institute Queensland, Griffith University, Southport, Queensland, Australia.,Griffith Institute for Drug Discovery, Griffith University, Nathan, Queensland, Australia
| | - James A St John
- Clem Jones Centre for Neurobiology and Stem Cell Research and Griffith University, Nathan, Queensland, Australia.,Menzies Health Institute Queensland, Griffith University, Southport, Queensland, Australia.,Griffith Institute for Drug Discovery, Griffith University, Nathan, Queensland, Australia
| |
Collapse
|
19
|
Paterson TE, Dhowre HS, Villanueva D, Holland JW, Reddy Kethiri A, Singh V, Claeyssens F, MacNeil S, Ortega Asencio I. Tuning Electrospun Substrate Stiffness for the Fabrication of a Biomimetic Amniotic Membrane Substitute for Corneal Healing. ACS APPLIED BIO MATERIALS 2021; 4:5638-5649. [DOI: 10.1021/acsabm.1c00436] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Thomas E. Paterson
- Automatic Control and Systems Engineering, University of Sheffield, Sheffield S1 3JD, United Kingdom
| | - Hala S. Dhowre
- School of Dentistry, University of Sheffield, Sheffield S10 2TA, United Kingdom
| | - Danilo Villanueva
- School of Dentistry, University of Sheffield, Sheffield S10 2TA, United Kingdom
| | - Joseph W. Holland
- School of Dentistry, University of Sheffield, Sheffield S10 2TA, United Kingdom
| | - Abhinav Reddy Kethiri
- Centre for Ocular Regeneration, Prof. Brien Holden Eye Research Centre, L V Prasad Eye Institute, Hyderabad 500034, India
| | - Vivek Singh
- Centre for Ocular Regeneration, Prof. Brien Holden Eye Research Centre, L V Prasad Eye Institute, Hyderabad 500034, India
| | - Frederik Claeyssens
- The Kroto Research Institute, North Campus, University of Sheffield, Sheffield S3 7HQ, United Kingdom
| | - Sheila MacNeil
- The Kroto Research Institute, North Campus, University of Sheffield, Sheffield S3 7HQ, United Kingdom
| | | |
Collapse
|
20
|
Tejeda G, Ciciriello AJ, Dumont CM. Biomaterial Strategies to Bolster Neural Stem Cell-Mediated Repair of the Central Nervous System. Cells Tissues Organs 2021; 211:655-669. [PMID: 34120118 DOI: 10.1159/000515351] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 02/12/2021] [Indexed: 01/25/2023] Open
Abstract
Stem cell therapies have the potential to not only repair, but to regenerate tissue of the central nervous system (CNS). Recent studies demonstrate that transplanted stem cells can differentiate into neurons and integrate with the intact circuitry after traumatic injury. Unfortunately, the positive findings described in rodent models have not been replicated in clinical trials, where the burden to maintain the cell viability necessary for tissue repair becomes more challenging. Low transplant survival remains the greatest barrier to stem cell-mediated repair of the CNS, often with fewer than 1-2% of the transplanted cells remaining after 1 week. Strategic transplantation parameters, such as injection location, cell concentration, and transplant timing achieve only modest improvements in stem cell transplant survival and appear inconsistent across studies. Biomaterials provide researchers with a means to significantly improve stem cell transplant survival through two mechanisms: (1) a vehicle to deliver and protect the stem cells and (2) a substrate to control the cytotoxic injury environment. These biomaterial strategies can alleviate cell death associated with delivery to the injury and can be used to limit cell death after transplantation by limiting cell exposure to cytotoxic signals. Moreover, it is likely that control of the injury environment with biomaterials will lead to a more reliable support for transplanted cell populations. This review will highlight the challenges associated with cell delivery in the CNS and the advances in biomaterial development and deployment for stem cell therapies necessary to bolster stem cell-mediated repair.
Collapse
Affiliation(s)
- Giancarlo Tejeda
- Department of Biomedical Engineering, University of Miami, Coral Gables, Florida, USA.,Biomedical Nanotechnology Institute at the University of Miami (BioNIUM), University of Miami, Miami, Florida, USA
| | - Andrew J Ciciriello
- Department of Biomedical Engineering, University of Miami, Coral Gables, Florida, USA.,Biomedical Nanotechnology Institute at the University of Miami (BioNIUM), University of Miami, Miami, Florida, USA
| | - Courtney M Dumont
- Department of Biomedical Engineering, University of Miami, Coral Gables, Florida, USA.,Biomedical Nanotechnology Institute at the University of Miami (BioNIUM), University of Miami, Miami, Florida, USA
| |
Collapse
|
21
|
Zhang X, Gong B, Zhai J, Zhao Y, Lu Y, Zhang L, Xue J. A Perspective: Electrospun Fibers for Repairing Spinal Cord Injury. Chem Res Chin Univ 2021. [DOI: 10.1007/s40242-021-1162-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
22
|
Roy HS, Singh R, Ghosh D. SARS-CoV-2 and tissue damage: current insights and biomaterial-based therapeutic strategies. Biomater Sci 2021; 9:2804-2824. [PMID: 33666206 DOI: 10.1039/d0bm02077j] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The effect of SARS-CoV-2 infection on humanity has gained worldwide attention and importance due to the rapid transmission, lack of treatment options and high mortality rate of the virus. While scientists across the world are searching for vaccines/drugs that can control the spread of the virus and/or reduce the risks associated with infection, patients infected with SARS-CoV-2 have been reported to have tissue/organ damage. With most tissues/organs having limited regenerative potential, interventions that prevent further damage or facilitate healing would be helpful. In the past few decades, biomaterials have gained prominence in the field of tissue engineering, in view of their major role in the regenerative process. Here we describe the effect of SARS-CoV-2 on multiple tissues/organs, and provide evidence for the positive role of biomaterials in aiding tissue repair. These findings are further extrapolated to explore their prospects as a therapeutic platform to address the tissue/organ damage that is frequently observed during this viral outbreak. This study suggests that the biomaterial-based approach could be an effective strategy for regenerating tissues/organs damaged by SARS-CoV-2.
Collapse
Affiliation(s)
- Himadri Shekhar Roy
- Department of Biological Science, Institute of Nanoscience and Technology (INST), Habitat Centre, Sector 64, Phase 10, Mohali-160062, Punjab, India.
| | - Rupali Singh
- Department of Biological Science, Institute of Nanoscience and Technology (INST), Habitat Centre, Sector 64, Phase 10, Mohali-160062, Punjab, India.
| | - Deepa Ghosh
- Department of Biological Science, Institute of Nanoscience and Technology (INST), Habitat Centre, Sector 64, Phase 10, Mohali-160062, Punjab, India.
| |
Collapse
|
23
|
Badekila AK, Kini S, Jaiswal AK. Fabrication techniques of biomimetic scaffolds in three-dimensional cell culture: A review. J Cell Physiol 2021; 236:741-762. [PMID: 32657458 DOI: 10.1002/jcp.29935] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 07/03/2020] [Indexed: 12/20/2022]
Abstract
In the last four decades, several researchers worldwide have routinely and meticulously exercised cell culture experiments in two-dimensional (2D) platforms. Using traditionally existing 2D models, the therapeutic efficacy of drugs has been inappropriately validated due to the failure in generating the precise therapeutic response. Fortunately, a 3D model addresses the foregoing limitations by recapitulating the in vivo environment. In this context, one has to contemplate the design of an appropriate scaffold for favoring the organization of cell microenvironment. Instituting pertinent model on the platter will pave way for a precise mimicking of in vivo conditions. It is because animal cells in scaffolds oblige spontaneous formation of 3D colonies that molecularly, phenotypically, and histologically resemble the native environment. The 3D culture provides insight into the biochemical aspects of cell-cell communication, plasticity, cell division, cytoskeletal reorganization, signaling mechanisms, differentiation, and cell death. Focusing on these criteria, this paper discusses in detail, the diversification of polymeric scaffolds based on their available resources. The paper also reviews the well-founded and latest techniques of scaffold fabrication, and their applications pertaining to tissue engineering, drug screening, and tumor model development.
Collapse
Affiliation(s)
- Anjana K Badekila
- Nitte University Centre for Science Education and Research, Nitte (Deemed to be University), Mangalore, Karnataka, India
| | - Sudarshan Kini
- Nitte University Centre for Science Education and Research, Nitte (Deemed to be University), Mangalore, Karnataka, India
| | - Amit K Jaiswal
- Centre for Biomaterials, Cellular, and Molecular Theranostics, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| |
Collapse
|
24
|
Zimmermann R, Vieira Alves Y, Sperling LE, Pranke P. Nanotechnology for the Treatment of Spinal Cord Injury. TISSUE ENGINEERING PART B-REVIEWS 2020; 27:353-365. [PMID: 33135599 DOI: 10.1089/ten.teb.2020.0188] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Spinal cord injury (SCI) affects the central nervous system (CNS) and there is currently no treatment with the potential for rehabilitation. Although several clinical treatments have been developed, they are still at an early stage and have not shown success in repairing the broken fiber, which prevents cellular regeneration and integral restoration of motor and sensory functions. Considering the importance of nanotechnology and tissue engineering for neural tissue injuries, this review focuses on the latest advances in nanotechnology for SCI treatment and tissue repair. The PubMed database was used for the bibliographic survey. Initial research using the following keywords "tissue engineering and spinal cord injury" revealed 970 articles published in the last 10 years. The articles were further analyzed, excluding those not related to SCI or with results that did not pertain to the field of interest, including the reviews. It was observed that a total of 811 original articles used the quoted keywords. When the word "treatment" was added, 662 articles were found and among them, 529 were original ones. Finally, when the keywords "Nanotechnology and spinal cord injury" were used, 102 articles were found, 65 being original articles. A search concerning the biomaterials used for SCI found 700 articles with 589 original articles. A total of 107 articles were included in the discussion of this review and some are used for the theoretical framework. Recent progress in nanotechnology and tissue engineering has shown promise for repairing CNS damage. A variety of in vivo animal testing for SCI has been used with or without cells and some of these in vivo studies have shown successful results. However, there is no translation to humans using nanotechnology for SCI treatment, although there is one ongoing trial that employs a tissue engineering approach, among other technologies. The first human surgical scaffold implantation will elucidate the possibility of this use for further clinical trials. This review concludes that even though tissue engineering and nanotechnology are being investigated as a possibility for SCI treatment, tests with humans are still in the theoretical stage. Impact statement Thousands of people are affected by spinal cord injury (SCI) per year in the world. This type of lesion is one of the most severe conditions that can affect humans and usually causes permanent loss of strength, sensitivity, and motor function below the injury site. This article reviews studies on the PubMed database, assessing the publications on SCI in the study field of tissue engineering, focusing on the use of nanotechnology for the treatment of SCI. The review makes an evaluation of the biomaterials used for the treatment of this condition and the techniques applied for the production of nanostructured biomaterials.
Collapse
Affiliation(s)
- Rafaela Zimmermann
- Hematology and Stem Cell Laboratory, Faculty of Pharmacy, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Yuri Vieira Alves
- Hematology and Stem Cell Laboratory, Faculty of Pharmacy, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Laura E Sperling
- Hematology and Stem Cell Laboratory, Faculty of Pharmacy, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Health School, Faculty of Medicine, UNISINOS, São Leopoldo, Brazil
| | - Patricia Pranke
- Hematology and Stem Cell Laboratory, Faculty of Pharmacy, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Stem Cell Research Institute, Porto Alegre, Brazil
| |
Collapse
|
25
|
Guedan-Duran A, Jemni-Damer N, Orueta-Zenarruzabeitia I, Guinea GV, Perez-Rigueiro J, Gonzalez-Nieto D, Panetsos F. Biomimetic Approaches for Separated Regeneration of Sensory and Motor Fibers in Amputee People: Necessary Conditions for Functional Integration of Sensory-Motor Prostheses With the Peripheral Nerves. Front Bioeng Biotechnol 2020; 8:584823. [PMID: 33224936 PMCID: PMC7670549 DOI: 10.3389/fbioe.2020.584823] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Accepted: 09/25/2020] [Indexed: 12/22/2022] Open
Abstract
The regenerative capacity of the peripheral nervous system after an injury is limited, and a complete function is not recovered, mainly due to the loss of nerve tissue after the injury that causes a separation between the nerve ends and to the disorganized and intermingled growth of sensory and motor nerve fibers that cause erroneous reinnervations. Even though the development of biomaterials is a very promising field, today no significant results have been achieved. In this work, we study not only the characteristics that should have the support that will allow the growth of nerve fibers, but also the molecular profile necessary for a specific guidance. To do this, we carried out an exhaustive study of the molecular profile present during the regeneration of the sensory and motor fibers separately, as well as of the effect obtained by the administration and inhibition of different factors involved in the regeneration. In addition, we offer a complete design of the ideal characteristics of a biomaterial, which allows the growth of the sensory and motor neurons in a differentiated way, indicating (1) size and characteristics of the material; (2) necessity to act at the microlevel, on small groups of neurons; (3) combination of molecules and specific substrates; and (4) temporal profile of those molecules expression throughout the regeneration process. The importance of the design we offer is that it respects the complexity and characteristics of the regeneration process; it indicates the appropriate temporal conditions of molecular expression, in order to obtain a synergistic effect; it takes into account the importance of considering the process at the group of neuron level; and it gives an answer to the main limitations in the current studies.
Collapse
Affiliation(s)
- Atocha Guedan-Duran
- Neuro-computing and Neuro-robotics Research Group, Complutense University of Madrid, Madrid, Spain
- Innovation Group, Institute for Health Research San Carlos Clinical Hospital (IdISSC), Madrid, Spain
- Department of Biomedical Engineering, Tufts University, Medford, MA, United States
| | - Nahla Jemni-Damer
- Neuro-computing and Neuro-robotics Research Group, Complutense University of Madrid, Madrid, Spain
- Innovation Group, Institute for Health Research San Carlos Clinical Hospital (IdISSC), Madrid, Spain
| | - Irune Orueta-Zenarruzabeitia
- Neuro-computing and Neuro-robotics Research Group, Complutense University of Madrid, Madrid, Spain
- Innovation Group, Institute for Health Research San Carlos Clinical Hospital (IdISSC), Madrid, Spain
| | - Gustavo Víctor Guinea
- Center for Biomedical Technology, Universidad Politécnica de Madrid, Madrid, Spain
- Department of Material Science, Civil Engineering Superior School, Universidad Politécnica de Madrid, Madrid, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
- Silk Biomed SL, Madrid, Spain
| | - José Perez-Rigueiro
- Center for Biomedical Technology, Universidad Politécnica de Madrid, Madrid, Spain
- Department of Material Science, Civil Engineering Superior School, Universidad Politécnica de Madrid, Madrid, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
- Silk Biomed SL, Madrid, Spain
| | - Daniel Gonzalez-Nieto
- Center for Biomedical Technology, Universidad Politécnica de Madrid, Madrid, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
- Silk Biomed SL, Madrid, Spain
| | - Fivos Panetsos
- Neuro-computing and Neuro-robotics Research Group, Complutense University of Madrid, Madrid, Spain
- Innovation Group, Institute for Health Research San Carlos Clinical Hospital (IdISSC), Madrid, Spain
- Silk Biomed SL, Madrid, Spain
| |
Collapse
|
26
|
Superior Synaptogenic Effect of Electrospun PLGA-PEG Nanofibers Versus PLGA Nanofibers on Human Neural SH-SY5Y Cells in a Three-Dimensional Culture System. J Mol Neurosci 2020; 70:1967-1976. [PMID: 32436197 DOI: 10.1007/s12031-020-01596-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 05/14/2020] [Indexed: 10/24/2022]
Abstract
Synapses are touted as the main structural and functional components of neural cells within in the nervous system, providing tissue connectivity and integration via the formation of perineuronal nets. In the present study, we evaluated the synaptogenic activity of electrospun PLGA and PLGA-PEG nanofibers on human SH-SY5Y cells after 14 days in vitro. Electrospun PLGA and PLGA-PEG nanofibers were fabricated and physicochemical properties were examined using the HNMR technique. The cells were classified into three random groups, i.e., control (laminin-coated surface), PLGA, and PLGA-PEG. Scaffolds' features, cell morphology, attachment, and alignment were monitored by SEM imaging. We performed MTT assay to measure cell survival rate. To evaluate neurite formation and axonal outgrowth, cells were stained with an antibody against β-tubulin III using immunofluorescence imaging. Antibodies against synapsin-1 and synaptophysin were used to explore the impact of PLGA and PLGA-PEG scaffolds on synaptogenesis and functional activity of synapses. According to SEM analysis, the PLGA-PEG scaffold had less thick nanofibers compared with the PLGA scaffold. Cell attachment, expansion, neurite outgrowth, and orientation were promoted in the PLGA-PEG group in comparison with the PLGA substrate (p < 0.05). MTT assay revealed that both scaffolds did not exert any neurotoxic effects on cell viability. Notably, PLGA-PEG surface increased cell viability compared to PLGA by time (p < 0.05). Immunofluorescence staining indicated an increased β-tubulin III level in the PLGA-PEG group days coincided with axonal outgrowth and immature neuron marker after seven compared with the PLGA and control groups (p < 0.05). Based on our data, both synaptogenesis and functional connectivity were induced in cells plated on the PLGA-PEG surface that coincide with the increase of synapsin-1 and synaptophysin in comparsion with the PLGA and control groups (p < 0.05). Taken together, our results imply that the PLGA-PEG nanofibers could provide the desirable microenvironment to develop perineuronal net formation, contributing to efficient synaptogenesis and neuron-to-neuron crosstalk.
Collapse
|
27
|
Ghane N, Beigi MH, Labbaf S, Nasr-Esfahani MH, Kiani A. Design of hydrogel-based scaffolds for the treatment of spinal cord injuries. J Mater Chem B 2020; 8:10712-10738. [DOI: 10.1039/d0tb01842b] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Hydrogel-based scaffold design approaches for the treatment of spinal cord injuries.
Collapse
Affiliation(s)
- Nazanin Ghane
- Department of Cellular Biotechnology Cell Science Research Center
- Royan Institute for Biotechnology
- ACECR
- Isfahan
- Iran
| | - Mohammad-Hossein Beigi
- Department of Cellular Biotechnology Cell Science Research Center
- Royan Institute for Biotechnology
- ACECR
- Isfahan
- Iran
| | - Sheyda Labbaf
- Biomaterials Research Group
- Department of Materials Engineering
- Isfahan University of Technology
- Isfahan
- Iran
| | | | - Amirkianoosh Kiani
- Silicon Hall: Micro/Nano Manufacturing Facility
- Faculty of Engineering and Applied Science
- Ontario Tech University
- Ontario
- Canada
| |
Collapse
|
28
|
Ma Z, Lu Y, Yang Y, Wang J, Kang X. Research progress and prospects of tissue engineering scaffolds for spinal cord injury repair and protection. Regen Med 2019; 14:887-898. [PMID: 31436130 DOI: 10.2217/rme-2018-0156] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Accepted: 08/01/2019] [Indexed: 02/08/2023] Open
Abstract
Spinal cord injury (SCI) is one of the leading causes of global disability. However, there are currently no effective clinical treatments for SCI. Repair of SCI is essential but poses great challenges. As a comprehensive treatment program combining biological scaffolds, seed cells and drugs or biological factors, tissue engineering has gradually replaced the single transplantation approach to become a focus of research that brings new opportunities for the clinical treatment of SCI.
Collapse
Affiliation(s)
- Zhanjun Ma
- The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu 730000, PR China
| | - Yubao Lu
- The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu 730000, PR China
| | - Yang Yang
- The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu 730000, PR China
| | - Jing Wang
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou, Gansu 730000, PR China
- The International Cooperation Base of Gansu Province for The Pain Research in Spinal Disorders, Gansu 730000, PR China
| | - Xuewen Kang
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou, Gansu 730000, PR China
- The International Cooperation Base of Gansu Province for The Pain Research in Spinal Disorders, Gansu 730000, PR China
| |
Collapse
|
29
|
Hesari Z, Mottaghitalab F, Shafiee A, Soleymani M, Dinarvand R, Atyabi F. Application of microfluidic systems for neural differentiation of cells. PRECISION NANOMEDICINE 2019. [DOI: 10.33218/prnano2(4).181127.2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Neural differentiation of stem cells is an important issue in development of central nervous system. Different methods such as chemical stimulation with small molecules, scaffolds, and microRNA can be used for inducing the differentiation of neural stem cells. However, microfluidic systems with the potential to induce neuronal differentiation have established their reputation in the field of regenerative medicine. Organization of microfluidic system represents a novel model that mimic the physiologic microenvironment of cells among other two and three dimensional cell culture systems. Microfluidic system has patterned and well-organized structure that can be combined with other differentiation techniques to provide optimal conditions for neuronal differentiation of stem cells. In this review, different methods for effective differentiation of stem cells to neuronal cells are summarized. The efficacy of microfluidic systems in promoting neuronal differentiation is also addressed.
Collapse
Affiliation(s)
- Zahra Hesari
- Guilan University of Medical Sciences, Rasht, Iran
| | | | | | | | | | | |
Collapse
|
30
|
Csobonyeiova M, Polak S, Zamborsky R, Danisovic L. Recent Progress in the Regeneration of Spinal Cord Injuries by Induced Pluripotent Stem Cells. Int J Mol Sci 2019; 20:3838. [PMID: 31390782 PMCID: PMC6695701 DOI: 10.3390/ijms20153838] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 07/27/2019] [Accepted: 08/02/2019] [Indexed: 12/13/2022] Open
Abstract
Regeneration of injuries occurring in the central nervous system, particularly spinal cord injuries (SCIs), is extremely difficult. The complex pathological events following a SCI often restrict regeneration of nervous tissue at the injury site and frequently lead to irreversible loss of motor and sensory function. Neural stem/progenitor cells (NSCs/NPCs) possess neuroregenerative and neuroprotective features, and transplantation of such cells into the site of damaged tissue is a promising stem cell-based therapy for SCI. However, NSC/NPCs have mostly been induced from embryonic stem cells or fetal tissue, leading to ethical concerns. The pioneering work of Yamanaka and colleagues gave rise to the technology to induce pluripotent stem cells (iPSCs) from somatic cells, overcoming these ethical issues. The advent of iPSCs technology has meant significant progress in the therapy of neurodegenerative disease and nerve tissue damage. A number of published studies have described the successful differentiation of NSCs/NPCs from iPSCs and their subsequent engraftment into SCI animal models, followed by functional recovery of injury. The aim of this present review is to summarize various iPSC- NPCs differentiation methods, SCI modelling, and the current status of possible iPSC- NPCs- based therapy of SCI.
Collapse
Affiliation(s)
- Maria Csobonyeiova
- Institute of Histology and Embryology, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia
| | - Stefan Polak
- Institute of Histology and Embryology, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia
| | - Radoslav Zamborsky
- Department of Orthopaedics, Faculty of Medicine, Comenius University and National Institute of Children's Diseases, Limbova 1, 833 40 Bratislava, Slovakia
| | - Lubos Danisovic
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia.
- Regenmed Ltd., Medena 29, 811 01 Bratislava, Slovakia.
| |
Collapse
|
31
|
Zhang Q, Shi B, Ding J, Yan L, Thawani JP, Fu C, Chen X. Polymer scaffolds facilitate spinal cord injury repair. Acta Biomater 2019; 88:57-77. [PMID: 30710714 DOI: 10.1016/j.actbio.2019.01.056] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 01/10/2019] [Accepted: 01/28/2019] [Indexed: 12/23/2022]
Abstract
During the past decades, improving patient neurological recovery following spinal cord injury (SCI) has remained a challenge. An effective treatment for SCI would not only reduce fractured elements and isolate developing local glial scars to promote axonal regeneration but also ameliorate secondary effects, including inflammation, apoptosis, and necrosis. Three-dimensional (3D) scaffolds provide a platform in which these mechanisms can be addressed in a controlled manner. Polymer scaffolds with favorable biocompatibility and appropriate mechanical properties have been engineered to minimize cicatrization, customize drug release, and ensure an unobstructed space to promote cell growth and differentiation. These properties make polymer scaffolds an important potential therapeutic platform. This review highlights the recent developments in polymer scaffolds for SCI engineering. STATEMENT OF SIGNIFICANCE: How to improve the efficacy of neurological recovery after spinal cord injury (SCI) is always a challenge. Tissue engineering provides a promising strategy for SCI repair, and scaffolds are one of the most important elements in addition to cells and inducing factors. The review highlights recent development and future prospects in polymer scaffolds for SCI therapy. The review will guide future studies by outlining the requirements and characteristics of polymer scaffold technologies employed against SCI. Additionally, the peculiar properties of polymer materials used in the therapeutic process of SCI also have guiding significance to other tissue engineering approaches.
Collapse
|
32
|
Willerth SM, Sakiyama-Elbert SE. Combining Stem Cells and Biomaterial Scaffolds for Constructing Tissues and Cell Delivery. ACTA ACUST UNITED AC 2019. [DOI: 10.3233/stj-180001] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Combining stem cells with biomaterial scaffolds serves as a promising strategy for engineering tissues for both in vitro and in vivo applications. This updated review details commonly used biomaterial scaffolds for engineering tissues from stem cells. We first define the different types of stem cells and their relevant properties and commonly used scaffold formulations. Next, we discuss natural and synthetic scaffold materials typically used when engineering tissues, along with their associated advantages and drawbacks and gives examples of target applications. New approaches to engineering tissues, such as 3D bioprinting, are described as they provide exciting opportunities for future work along with current challenges that must be addressed. Thus, this review provides an overview of the available biomaterials for directing stem cell differentiation as a means of producing replacements for diseased or damaged tissues.
Collapse
Affiliation(s)
- Stephanie M. Willerth
- Department of Mechanical Engineering, University of Victoria, VIC, Canada
- Division of Medical Sciences, University of Victoria, VIC, Canada
- International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, Canada
| | | |
Collapse
|
33
|
Stem cell paracrine effect and delivery strategies for spinal cord injury regeneration. J Control Release 2019; 300:141-153. [PMID: 30851286 DOI: 10.1016/j.jconrel.2019.02.038] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Revised: 02/22/2019] [Accepted: 02/25/2019] [Indexed: 12/29/2022]
Abstract
Spinal cord injury (SCI) is a complicated neuropathological condition that results in functional dysfunction and paralysis. Various treatments have been proposed including drugs, biological factors and cells administered in several ways. Stem cell therapy offers a potentially revolutionary mode to repair the damaged spinal cord after injury. Initially, stem cells were considered promising for replacing cells and tissue lost after SCI. Many studies looked at their differentiation to replace neuronal and glial cells for a better functional outcome. However, it is becoming clear that different functional improvements recognized to stem cells are due to biomolecular activities by the transplanted stem cells rather than cell replacement. This review aimed to discuss the paracrine mechanisms for tissue repair and regeneration after stem cell transplantation in SCI. It focuses on stem cell factor production, effect in tissue restoration, and novel delivery strategies to use them for SCI therapy.
Collapse
|
34
|
Dumont CM, Carlson MA, Munsell MK, Ciciriello AJ, Strnadova K, Park J, Cummings BJ, Anderson AJ, Shea LD. Aligned hydrogel tubes guide regeneration following spinal cord injury. Acta Biomater 2019; 86:312-322. [PMID: 30610918 DOI: 10.1016/j.actbio.2018.12.052] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 12/22/2018] [Accepted: 12/31/2018] [Indexed: 12/21/2022]
Abstract
Directing the organization of cells into a tissue with defined architectures is one use of biomaterials for regenerative medicine. To this end, hydrogels are widely investigated as they have mechanical properties similar to native soft tissues and can be formed in situ to conform to a defect. Herein, we describe the development of porous hydrogel tubes fabricated through a two-step polymerization process with an intermediate microsphere phase that provides macroscale porosity (66.5%) for cell infiltration. These tubes were investigated in a spinal cord injury model, with the tubes assembled to conform to the injury and to provide an orientation that guides axons through the injury. Implanted tubes had good apposition and were integrated with the host tissue due to cell infiltration, with a transient increase in immune cell infiltration at 1 week that resolved by 2 weeks post injury compared to a gelfoam control. The glial scar was significantly reduced relative to control, which enabled robust axon growth along the inner and outer surface of the tubes. Axon density within the hydrogel tubes (1744 axons/mm2) was significantly increased more than 3-fold compared to the control (456 axons/mm2), with approximately 30% of axons within the tube myelinated. Furthermore, implantation of hydrogel tubes enhanced functional recovery relative to control. This modular assembly of porous tubes to fill a defect and directionally orient tissue growth could be extended beyond spinal cord injury to other tissues, such as vascular or musculoskeletal tissue. STATEMENT OF SIGNIFICANCE: Tissue engineering approaches that mimic the native architecture of healthy tissue are needed following injury. Traditionally, pre-molded scaffolds have been implemented but require a priori knowledge of wound geometries. Conversely, hydrogels can conform to any injury, but do not guide bi-directional regeneration. In this work, we investigate the feasibility of a system of modular hydrogel tubes to promote bi-directional regeneration after spinal cord injury. This system allows for tubes to be cut to size during surgery and implanted one-by-one to fill any injury, while providing bi-directional guidance. Moreover, this system of tubes can be broadly applied to tissue engineering approaches that require a modular guidance system, such as repair to vascular or musculoskeletal tissues.
Collapse
|
35
|
Martínez-Ramos C, Doblado LR, Mocholi EL, Alastrue-Agudo A, Petidier MS, Giraldo E, Pradas MM, Moreno-Manzano V. Biohybrids for spinal cord injury repair. J Tissue Eng Regen Med 2019; 13:509-521. [PMID: 30726582 DOI: 10.1002/term.2816] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 11/08/2018] [Accepted: 01/14/2019] [Indexed: 01/05/2023]
Abstract
Spinal cord injuries (SCIs) result in the loss of sensory and motor function with massive cell death and axon degeneration. We have previously shown that transplantation of spinal cord-derived ependymal progenitor cells (epSPC) significantly improves functional recovery after acute and chronic SCI in experimental models, via neuronal differentiation and trophic glial cell support. Here, we propose an improved procedure based on transplantation of epSPC in a tubular conduit of hyaluronic acid containing poly (lactic acid) fibres creating a biohybrid scaffold. In vitro analysis showed that the poly (lactic acid) fibres included in the conduit induce a preferential neuronal fate of the epSPC rather than glial differentiation, favouring elongation of cellular processes. The safety and efficacy of the biohybrid implantation was evaluated in a complete SCI rat model. The conduits allowed efficient epSPC transfer into the spinal cord, improving the preservation of the neuronal tissue by increasing the presence of neuronal fibres at the injury site and by reducing cavities and cyst formation. The biohybrid-implanted animals presented diminished astrocytic reactivity surrounding the scar area, an increased number of preserved neuronal fibres with a horizontal directional pattern, and enhanced coexpression of the growth cone marker GAP43. The biohybrids offer an improved method for cell transplantation with potential capabilities for neuronal tissue regeneration, opening a promising avenue for cell therapies and SCI treatment.
Collapse
Affiliation(s)
- Cristina Martínez-Ramos
- Center for Biomaterials and Tissue Engineering, Universitat Politècnica de València, Valencia, Spain
| | - Laura Rodríguez Doblado
- Center for Biomaterials and Tissue Engineering, Universitat Politècnica de València, Valencia, Spain
| | - Eric López Mocholi
- Neuronal and Tissue Regeneration Laboratory, Prince Felipe Research Center, Valencia, Spain
| | - Ana Alastrue-Agudo
- Neuronal and Tissue Regeneration Laboratory, Prince Felipe Research Center, Valencia, Spain
| | | | - Esther Giraldo
- Neuronal and Tissue Regeneration Laboratory, Prince Felipe Research Center, Valencia, Spain
| | - Manuel Monleón Pradas
- Center for Biomaterials and Tissue Engineering, Universitat Politècnica de València, Valencia, Spain.,Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Valencia, Spain
| | | |
Collapse
|
36
|
Wang J, Zou W, Ma J, Liu J. Biomaterials and Gene Manipulation in Stem Cell-Based Therapies for Spinal Cord Injury. Stem Cells Dev 2019; 28:239-257. [PMID: 30489226 DOI: 10.1089/scd.2018.0169] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Spinal cord injury (SCI), a prominent health issue, represents a substantial portion of the global health care burden. Stem cell-based therapies provide novel solutions for SCI treatment, yet obstacles remain in the form of low survival rate, uncontrolled differentiation, and functional recovery. The application of engineered biomaterials in stem cell therapy provides a physicochemical microenvironment that mimics the stem cell niche, facilitating self-renewal, stem cell differentiation, and tissue reorganization. Nonetheless, external microenvironment support is inadequate, and some obstacles persist, for example, limited sources, gradual aging, and immunogenicity of stem cells. Targeted stem cell gene manipulation could eliminate many of these drawbacks, allowing safer, more effective use under regulation of intrinsic mechanisms. Additionally, through genetic labeling of stem cells, their role in tissue engineering may be elucidated. Therefore, combining stem cell therapy, materials science, and genetic modification technologies may shed light on SCI treatment. Herein, recent advances and advantages of biomaterials and gene manipulation, especially with respect to stem cell-based therapies, are highlighted, and their joint performance in SCI is evaluated. Current technological limitations and perspectives on future directions are then discussed. Although this combination is still in the early stages of development, it is highly likely to substantially contribute to stem cell-based therapies in the foreseeable future.
Collapse
Affiliation(s)
- Jiayi Wang
- 1 Regenerative Medicine Center, the First Affiliated Hospital of Dalian Medical University, Dalian, China.,2 Stem Cell Clinical Research Center, the First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Wei Zou
- 3 College of Life Sciences, Liaoning Normal University, Dalian, China.,4 Liaoning Key Laboratories of Biotechnology and Molecular Drug Research & Development, Dalian, China
| | - Jingyun Ma
- 1 Regenerative Medicine Center, the First Affiliated Hospital of Dalian Medical University, Dalian, China.,2 Stem Cell Clinical Research Center, the First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jing Liu
- 1 Regenerative Medicine Center, the First Affiliated Hospital of Dalian Medical University, Dalian, China.,2 Stem Cell Clinical Research Center, the First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
37
|
Restoring electrical connection using a conductive biomaterial provides a new therapeutic strategy for rats with spinal cord injury. Neurosci Lett 2019; 692:33-40. [DOI: 10.1016/j.neulet.2018.10.031] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Revised: 09/28/2018] [Accepted: 10/15/2018] [Indexed: 11/22/2022]
|
38
|
Kong W, Qi Z, Xia P, Chang Y, Li H, Qu Y, Pan S, Yang X. Local delivery of FTY720 and NSCs on electrospun PLGA scaffolds improves functional recovery after spinal cord injury. RSC Adv 2019; 9:17801-17811. [PMID: 35520542 PMCID: PMC9064641 DOI: 10.1039/c9ra01717h] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 05/27/2019] [Indexed: 12/23/2022] Open
Abstract
Spinal cord injury (SCI) is a common issue in the clinic that causes severe motor and sensory dysfunction below the lesion level. FTY720, also known as fingolimod, has recently been reported to exert a positive effect on the recovery from a spinal cord injury. Through local delivery to the lesion site, FTY720 effectively integrates with biomaterials, and the systemic adverse effects are alleviated. However, the effects of the proper mass ratio of FTY720 in biomaterials on neural stem cell (NSC) proliferation and differentiation, as well as functional recovery after SCI, have not been thoroughly investigated. In our study, we fabricated electrospun poly (lactide-co-glycolide) (PLGA)/FTY720 scaffolds at different mass ratios (0.1%, 1%, and 10%) and characterized these scaffolds. The effects of electrospun PLGA/FTY720 scaffolds on NSC proliferation and differentiation were measured. Then, a rat model of spinal transection was established to investigate the effects of PLGA/FTY720 scaffolds loaded with NSCs. Notably, 1% PLGA/FTY720 scaffolds exerted the best effects on the proliferation and differentiation of NSCs and 10% PLGA/FTY720 was cytotoxic to NSCs. Based on the Basso, Beattie, and Bresnahan (BBB) score, HE staining and immunofluorescence staining, the PLGA/FTY720 scaffold loaded with NSCs effectively promoted the recovery of spinal cord function. Thus, FTY720 properly integrated with electrospun PLGA scaffolds, and electrospun PLGA/FTY720 scaffolds loaded with NSCs may have potential applications for SCI as a nerve implant. Spinal cord injury (SCI) is a common issue in the clinic that causes severe motor and sensory dysfunction below the lesion level.![]()
Collapse
Affiliation(s)
- Weijian Kong
- Department of Orthopedic Surgery
- The Second Hospital of Jilin University
- Changchun
- PR China
| | - Zhiping Qi
- Department of Orthopedic Surgery
- The Second Hospital of Jilin University
- Changchun
- PR China
| | - Peng Xia
- Department of Orthopedic Surgery
- The Second Hospital of Jilin University
- Changchun
- PR China
| | - Yuxin Chang
- Department of Orthopedic Surgery
- The Second Hospital of Jilin University
- Changchun
- PR China
| | - Hongru Li
- Department of Orthopedic Surgery
- The Second Hospital of Jilin University
- Changchun
- PR China
| | - Yunpeng Qu
- Department of Cardiovascular Medicine
- The Second Hospital of Jilin University
- Changchun
- PR China
| | - Su Pan
- Department of Orthopedic Surgery
- The Second Hospital of Jilin University
- Changchun
- PR China
| | - Xiaoyu Yang
- Department of Orthopedic Surgery
- The Second Hospital of Jilin University
- Changchun
- PR China
| |
Collapse
|
39
|
Babaloo H, Ebrahimi-Barough S, Derakhshan MA, Yazdankhah M, Lotfibakhshaiesh N, Soleimani M, Joghataei MT, Ai J. PCL/gelatin nanofibrous scaffolds with human endometrial stem cells/Schwann cells facilitate axon regeneration in spinal cord injury. J Cell Physiol 2018; 234:11060-11069. [PMID: 30584656 DOI: 10.1002/jcp.27936] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 10/25/2018] [Indexed: 12/28/2022]
Abstract
The significant consequences of spinal cord injury (SCI) include sensory and motor disability resulting from the death of neuronal cells and axon degeneration. In this respect, overcoming the consequences of SCI including the recovery of sensory and motor functions is considered to be a difficult tasks that requires attention to multiple aspects of treatment. The breakthrough in tissue engineering through the integration of biomaterial scaffolds and stem cells has brought a new hope for the treatment of SCI. In the present study, human endometrial stem cells (hEnSCs) were cultured with human Schwann cells (hSC) in transwells, their differentiation into nerve-like cells was confirmed by quantitative real-time reverse transcription polymerase chain reaction (qRT-PCR) and immunocytochemistry techniques. The differentiated cells (co-hEnSC) were then seeded on the poly ε-caprolactone (PCL)/gelatin scaffolds. The SEM images displayed the favorable seeding and survival of the cells on the scaffolds. The seeded scaffolds were then transplanted into hemisected SCI rats. The growth of neuronal cells was confirmed with immunohistochemical study using NF-H as a neuronal marker. Finally, the Basso, Beattie, and Bresnahan (BBB) test confirmed the recovery of sensory and motor functions. The results suggested that combination therapy using the differentiated hEnSC seeded on PCL/gelatin scaffolds has the potential to heal the injured spinal cord and to limit the secondary damage.
Collapse
Affiliation(s)
- Hamideh Babaloo
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Somayeh Ebrahimi-Barough
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Ali Derakhshan
- Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Meysam Yazdankhah
- Department of Ophthalmology, Glia Research Laboratory, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Nasrin Lotfibakhshaiesh
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Masoud Soleimani
- Department of Hematology, Faculty of Medical Science, Tarbiat Modares University, Tehran, Iran
| | - Mohammad-Taghi Joghataei
- Department of Anatomical Sciences, Neuroscience Research Center & Cellular and Molecular Research Center, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Jafar Ai
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
40
|
Boni R, Ali A, Shavandi A, Clarkson AN. Current and novel polymeric biomaterials for neural tissue engineering. J Biomed Sci 2018; 25:90. [PMID: 30572957 PMCID: PMC6300901 DOI: 10.1186/s12929-018-0491-8] [Citation(s) in RCA: 214] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 11/27/2018] [Indexed: 12/12/2022] Open
Abstract
The nervous system is a crucial component of the body and damages to this system, either by of injury or disease, can result in serious or potentially lethal consequences. Restoring the damaged nervous system is a great challenge due to the complex physiology system and limited regenerative capacity.Polymers, either synthetic or natural in origin, have been extensively evaluated as a solution for restoring functions in damaged neural tissues. Polymers offer a wide range of versatility, in particular regarding shape and mechanical characteristics, and their biocompatibility is unmatched by other biomaterials, such as metals and ceramics. Several studies have shown that polymers can be shaped into suitable support structures, including nerve conduits, scaffolds, and electrospun matrices, capable of improving the regeneration of damaged neural tissues. In general, natural polymers offer the advantage of better biocompatibility and bioactivity, while synthetic or non-natural polymers have better mechanical properties and structural stability. Often, combinations of the two allow for the development of polymeric conduits able to mimic the native physiological environment of healthy neural tissues and, consequently, regulate cell behaviour and support the regeneration of injured nervous tissues.Currently, most of neural tissue engineering applications are in pre-clinical study, in particular for use in the central nervous system, however collagen polymer conduits aimed at regeneration of peripheral nerves have already been successfully tested in clinical trials.This review highlights different types of natural and synthetic polymers used in neural tissue engineering and their advantages and disadvantages for neural regeneration.
Collapse
Affiliation(s)
- Rossana Boni
- Bioengineering Research Team, Centre for Bioengineering and Nanomedicine, Department of Food Science, University of Otago, PO Box 56, Dunedin, 9054 New Zealand
| | - Azam Ali
- Bioengineering Research Team, Centre for Bioengineering and Nanomedicine, Department of Food Science, University of Otago, PO Box 56, Dunedin, 9054 New Zealand
| | - Amin Shavandi
- Bioengineering Research Team, Centre for Bioengineering and Nanomedicine, Department of Food Science, University of Otago, PO Box 56, Dunedin, 9054 New Zealand
- BioMatter-Biomass Transformation Lab (BTL), École interfacultaire de Bioingénieurs (EIB), École polytechnique de Bruxelles, Université Libre de Bruxelles, Avenue F.D. Roosevelt, 50 - CP 165/61, 1050 Brussels, Belgium
| | - Andrew N. Clarkson
- Department of Anatomy, Brain Health Research Centre and Brain Research New Zealand, University of Otago, PO Box 56, Dunedin, 9054 New Zealand
| |
Collapse
|
41
|
Zhu W, Zhang H, Chen X, Jin K, Ning L. Numerical characterization of regenerative axons growing along a spherical multifunctional scaffold after spinal cord injury. PLoS One 2018; 13:e0205961. [PMID: 30365562 PMCID: PMC6203361 DOI: 10.1371/journal.pone.0205961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 10/04/2018] [Indexed: 11/18/2022] Open
Abstract
Spinal cord injury (SCI) followed by extensive cell loss, inflammation, and scarring, often permanently damages neurological function. Biomaterial scaffolds are promising but currently have limited applicability in SCI because after entering the scaffold, regenerating axons tend to become trapped and rarelyre-enter the host tissue, the reasons for which remain to be completely explored. Here, we propose a mathematical model and computer simulation for characterizing regenerative axons growing along a scaffold following SCI, and how their growth may be guided. The model assumed a solid, spherical, multifunctional, biomaterial scaffold, that would bridge the rostral and caudal stumps of a completely transected spinal cord in a rat model and would guide the rostral regenerative axons toward the caudal tissue. Other assumptions include the whole scaffold being coated with extracellular matrix components, and the caudal area being additionally seeded with chemoattractants. The chemical factors on and around the scaffold were formulated to several coupled variables, and the parameter values were derived fromexisting experimental data. Special attention was given to the effects of coating strength, seeding location, and seeding density, as well as the ramp slope of the scaffold, on axonal regeneration. In numerical simulations, a slimmer scaffold provided a small slope at the entry "on-ramp" area that improved the success rate of axonal regeneration. If success rates are high, an increased number of regenerative axons traverse through the narrow channels, causing congestion and lowering the growth rate. An increase in the number of severed axons (300-12000) did not significantly affect the growth rate, but it reduced the success rate of axonal regeneration. However, an increase in the seeding densities of the complexes on the whole scaffold, and that in the seeding densities of the chemoattractants on the caudal area, improved both the success and growth rates. However, an increase in the density of thecomplexes on the whole scaffold risks an over-eutrophic surface that harms axonal regeneration.Although theoretical predictions are yet to be validated directly by experiments, this theoretical tool can advance the treatment of SCI, and is also applicable to scaffolds with other architectures.
Collapse
Affiliation(s)
- Weiping Zhu
- Shanghai Institute of Applied Mathematics and Mechanics, Shanghai University, Shanghai, People's Republic of China
- * E-mail:
| | - Han Zhang
- Shanghai Institute of Applied Mathematics and Mechanics, Shanghai University, Shanghai, People's Republic of China
| | - Xuning Chen
- Shanghai Institute of Applied Mathematics and Mechanics, Shanghai University, Shanghai, People's Republic of China
| | - Kan Jin
- Shanghai Institute of Applied Mathematics and Mechanics, Shanghai University, Shanghai, People's Republic of China
| | - Le Ning
- Shanghai Institute of Applied Mathematics and Mechanics, Shanghai University, Shanghai, People's Republic of China
| |
Collapse
|
42
|
Zhou X, Shi G, Fan B, Cheng X, Zhang X, Wang X, Liu S, Hao Y, Wei Z, Wang L, Feng S. Polycaprolactone electrospun fiber scaffold loaded with iPSCs-NSCs and ASCs as a novel tissue engineering scaffold for the treatment of spinal cord injury. Int J Nanomedicine 2018; 13:6265-6277. [PMID: 30349249 PMCID: PMC6186894 DOI: 10.2147/ijn.s175914] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background Spinal cord injury (SCI) is a traumatic disease of the central nervous system, accompanied with high incidence and high disability rate. Tissue engineering scaffold can be used as therapeutic systems to provide effective repair for SCI. Purpose In this study, a novel tissue engineering scaffold has been synthesized in order to explore the effect of nerve repair on SCI. Patients and methods Polycaprolactone (PCL) scaffolds loaded with actived Schwann cells (ASCs) and induced pluripotent stem cells -derived neural stem cells (iPSC-NSCs), a combined cell transplantation strategy, were prepared and characterized. The cell-loaded PCL scaffolds were further utilized for the treatment of SCI in vivo. Histological observation, behavioral evaluation, Western-blot and qRT-PCR were used to investigate the nerve repair of Wistar rats after scaffold transplantation. Results The iPSCs displayed similar characteristics to embryonic stem cells and were efficiently differentiated into neural stem cells in vitro. The obtained PCL scaffolds werê0.5 mm in thickness with biocompatibility and biodegradability. SEM results indicated that the ASCs and (or) iPS-NSCs grew well on PCL scaffolds. Moreover, transplantation reduced the volume of lesion cavity and improved locomotor recovery of rats. In addition, the degree of spinal cord recovery and remodeling maybe closely related to nerve growth factor and glial cell-derived neurotrophic factor. In summary, our results demonstrated that tissue engineering scaffold treatment could increase tissue remodeling and could promote motor function recovery in a transection SCI model. Conclusion This study provides preliminary evidence for using tissue engineering scaffold as a clinically viable treatment for SCI in the future.
Collapse
Affiliation(s)
- XianHu Zhou
- International Science and Technology Cooperation Base of Spinal Cord Injury, Department of Orthopedic Surgery, Tianjin Medical University General Hospital, Tianjin, People's Republic of China,
| | - GuiDong Shi
- International Science and Technology Cooperation Base of Spinal Cord Injury, Department of Orthopedic Surgery, Tianjin Medical University General Hospital, Tianjin, People's Republic of China,
| | - BaoYou Fan
- International Science and Technology Cooperation Base of Spinal Cord Injury, Department of Orthopedic Surgery, Tianjin Medical University General Hospital, Tianjin, People's Republic of China, .,Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, People's Republic of China,
| | - Xin Cheng
- International Science and Technology Cooperation Base of Spinal Cord Injury, Department of Orthopedic Surgery, Tianjin Medical University General Hospital, Tianjin, People's Republic of China, .,Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, People's Republic of China,
| | - XiaoLei Zhang
- International Science and Technology Cooperation Base of Spinal Cord Injury, Department of Orthopedic Surgery, Tianjin Medical University General Hospital, Tianjin, People's Republic of China, .,Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, People's Republic of China,
| | - Xu Wang
- International Science and Technology Cooperation Base of Spinal Cord Injury, Department of Orthopedic Surgery, Tianjin Medical University General Hospital, Tianjin, People's Republic of China, .,Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, People's Republic of China,
| | - Shen Liu
- International Science and Technology Cooperation Base of Spinal Cord Injury, Department of Orthopedic Surgery, Tianjin Medical University General Hospital, Tianjin, People's Republic of China, .,Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, People's Republic of China,
| | - Yan Hao
- International Science and Technology Cooperation Base of Spinal Cord Injury, Department of Orthopedic Surgery, Tianjin Medical University General Hospital, Tianjin, People's Republic of China, .,Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, People's Republic of China,
| | - ZhiJian Wei
- International Science and Technology Cooperation Base of Spinal Cord Injury, Department of Orthopedic Surgery, Tianjin Medical University General Hospital, Tianjin, People's Republic of China, .,Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, People's Republic of China,
| | - LianYong Wang
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Nankai University, Tianjin, People's Republic of China,
| | - ShiQing Feng
- International Science and Technology Cooperation Base of Spinal Cord Injury, Department of Orthopedic Surgery, Tianjin Medical University General Hospital, Tianjin, People's Republic of China, .,Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, People's Republic of China,
| |
Collapse
|
43
|
Miller RJ, Chan CY, Rastogi A, Grant AM, White CM, Bette N, Schaub NJ, Corey JM. Combining electrospun nanofibers with cell-encapsulating hydrogel fibers for neural tissue engineering. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2018; 29:1625-1642. [PMID: 29862935 PMCID: PMC7446748 DOI: 10.1080/09205063.2018.1479084] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 05/17/2018] [Indexed: 10/14/2022]
Abstract
A promising component of biomaterial constructs for neural tissue engineering are electrospun fibers, which differentiate stem cells and neurons as well as direct neurite growth. However, means of protecting neurons, glia, and stem cells seeded on electrospun fibers between lab and surgical suite have yet to be developed. Here we report an effort to accomplish this using cell-encapsulating hydrogel fibers made by interfacial polyelectrolyte complexation (IPC). IPC-hydrogel fibers were created by interfacing acid-soluble chitosan (AsC) and cell-containing alginate and spinning them on bundles of aligned electrospun fibers. Primary spinal astrocytes, cortical neurons, or L929 fibroblasts were mixed into alginate hydrogels prior to IPC-fiber spinning. The viability of each cell type was assessed at 30 min, 4 h, 1 d, and 7 d after encapsulation in IPC hydrogels. Some neurons were encapsulated in IPC-hydrogel fibers made from water-soluble chitosan (WsC). Neurons were also stained with Tuj1 and assessed for neurite extension. Neuron survival in AsC-fibers was worse than astrocytes in AsC-fibers (p < 0.05) and neurons in WsC-fibers (p < 0.05). As expected, neuron and glia survival was worse than L929 fibroblasts (p < 0.05). Neurons in IPC-hydrogel fibers fabricated with WsC extended neurites robustly, while none in AsC fibers did. Neurons remaining inside IPC-hydrogel fibers extended neurites inside them, while others de-encapsulated, extending neurites on electrospun fibers, which did not fully integrate with IPC-hydrogel fibers. This study demonstrates that primary neurons and astrocytes can be encapsulated in IPC-hydrogel fibers at good percentages of survival. IPC hydrogel technology may be a useful tool for encapsulating neural and other cells on electrospun fiber scaffolds.
Collapse
Affiliation(s)
- Ryan J. Miller
- Department of Research and Geriatric Research Education and Clinical Center (GRECC), VA Ann Arbor Healthcare System, Ann Arbor, Michigan 48105
| | - Cheook Y. Chan
- Department of Research and Geriatric Research Education and Clinical Center (GRECC), VA Ann Arbor Healthcare System, Ann Arbor, Michigan 48105
- Undergraduate Research Opportunity Program, The University of Michigan, Ann Arbor, MI 48109, USA
| | - Arjun Rastogi
- Department of Research and Geriatric Research Education and Clinical Center (GRECC), VA Ann Arbor Healthcare System, Ann Arbor, Michigan 48105
| | - Allison M. Grant
- Department of Research and Geriatric Research Education and Clinical Center (GRECC), VA Ann Arbor Healthcare System, Ann Arbor, Michigan 48105
- Undergraduate Research Opportunity Program, The University of Michigan, Ann Arbor, MI 48109, USA
| | - Christina M. White
- Department of Research and Geriatric Research Education and Clinical Center (GRECC), VA Ann Arbor Healthcare System, Ann Arbor, Michigan 48105
| | - Nicole Bette
- Department of Research and Geriatric Research Education and Clinical Center (GRECC), VA Ann Arbor Healthcare System, Ann Arbor, Michigan 48105
| | - Nicholas J. Schaub
- Department of Research and Geriatric Research Education and Clinical Center (GRECC), VA Ann Arbor Healthcare System, Ann Arbor, Michigan 48105
- Department of Neurology, The University of Michigan, Ann Arbor, MI 48109, USA
| | - Joseph M. Corey
- Department of Research and Geriatric Research Education and Clinical Center (GRECC), VA Ann Arbor Healthcare System, Ann Arbor, Michigan 48105
- Department of Neurology, The University of Michigan, Ann Arbor, MI 48109, USA
- Department of Biomedical Engineering, The University of Michigan, Ann Arbor, MI 48109, USA
- Macromolecular Science and Engineering Program, The University of Michigan, Ann Arbor, MI 48109, USA
- Neuroscience Graduate Program, The University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
44
|
Lu X, Perera TH, Aria AB, Callahan LAS. Polyethylene glycol in spinal cord injury repair: a critical review. J Exp Pharmacol 2018; 10:37-49. [PMID: 30100766 PMCID: PMC6067622 DOI: 10.2147/jep.s148944] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Polyethylene glycol (PEG) is a synthetic biocompatible polymer with many useful properties for developing therapeutics to treat spinal cord injury. Direct application of PEG as a fusogen to the injury site can repair cell membranes, mitigate oxidative stress, and promote axonal regeneration to restore motor function. PEG can be covalently or noncovalently conjugated to proteins, peptides, and nanoparticles to limit their clearance by the reticuloendothelial system, reduce their immunogenicity, and facilitate crossing the blood-brain barrier. Cross-linking PEG produces hydrogels that can act as delivery vehicles for bioactive molecules including growth factors and cells such as bone marrow stromal cells, which can modulate the inflammatory response and support neural tissue regeneration. PEG hydrogels can be cross-linked in vitro or delivered as an injectable formulation that can gel in situ at the site of injury. Chemical and mechanical properties of PEG hydrogels are tunable and must be optimized for creating the most favorable delivery environment. Peptides mimicking extracellular matrix protein such as laminin and n-cadherin can be incorporated into PEG hydrogels to promote neural differentiation and axonal extensions. Different hydrogel cross-linking densities and stiffness will also affect the differentiation process. PEG hydrogels with a gradient of peptide concentrations or Young's modulus have been developed to systematically study these factors. This review will describe these and other recent advancements of PEG in the field of spinal cord injury in greater detail.
Collapse
Affiliation(s)
- Xi Lu
- Department of Neurosurgery, Center for Stem Cells and Regenerative Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA,
| | - T Hiran Perera
- Department of Neurosurgery, Center for Stem Cells and Regenerative Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA,
| | - Alexander B Aria
- Department of Neurosurgery, Center for Stem Cells and Regenerative Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA,
| | - Laura A Smith Callahan
- Department of Neurosurgery, Center for Stem Cells and Regenerative Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA,
| |
Collapse
|
45
|
Bhere D, Khajuria RK, Hendriks WT, Bandyopadhyay A, Bagci-Onder T, Shah K. Stem Cells Engineered During Different Stages of Reprogramming Reveal Varying Therapeutic Efficacies. Stem Cells 2018; 36:932-942. [PMID: 29451340 PMCID: PMC5992036 DOI: 10.1002/stem.2805] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 01/31/2018] [Accepted: 02/05/2018] [Indexed: 12/11/2022]
Abstract
Stem cells are emerging as promising treatment strategies for several brain disorders and pathologies. In this study, we explored the potential of creating induced pluripotent stem cell-derived neural stem cells (ipNSC) by using either unmodified or gene-modified somatic cells and tested their fate and therapeutic efficacies in vitro and in vivo. We show that cells engineered in somatic state lose transgene-expression during the neural induction process, which is partially restored by histone deacetylase inhibitor treatment whereas cells engineered at the ipNSC state have sustained expression of transgenes. In vivo, bimodal mouse and human ipNSCs engineered to express tumor specific death-receptor ligand and suicide-inducing therapeutic proteins have profound anti-tumor efficacy when encapsulated in synthetic extracellular matrix and transplanted in mouse models of resected-glioblastoma. This study provides insights into using somatic cells for treating CNS disorders and presents a receptor-targeted cancer therapeutic approach for brain tumors. Stem Cells 2018;36:932-942.
Collapse
Affiliation(s)
- Deepak Bhere
- Center for Stem Cell Therapeutics and Imaging, Harvard Medical School, Boston, MA 02114 USA
- Department of Radiology, Harvard Medical School, Boston, MA 02114 USA
| | - Rajiv Kumar Khajuria
- Center for Stem Cell Therapeutics and Imaging, Harvard Medical School, Boston, MA 02114 USA
- Department of Radiology, Harvard Medical School, Boston, MA 02114 USA
| | - William T. Hendriks
- The Collaborative Center for X-Linked Dystonia-Parkinsonism, Harvard Medical School, Boston, MA 02114 USA
- Harvard Brain Science Initiative, Harvard Medical School, Boston MA 02114 USA
| | - Antara Bandyopadhyay
- Center for Stem Cell Therapeutics and Imaging, Harvard Medical School, Boston, MA 02114 USA
- Department of Radiology, Harvard Medical School, Boston, MA 02114 USA
| | - Tugba Bagci-Onder
- Center for Stem Cell Therapeutics and Imaging, Harvard Medical School, Boston, MA 02114 USA
- Department of Radiology, Harvard Medical School, Boston, MA 02114 USA
| | - Khalid Shah
- Center for Stem Cell Therapeutics and Imaging, Harvard Medical School, Boston, MA 02114 USA
- Department of Radiology, Harvard Medical School, Boston, MA 02114 USA
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114 USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA
| |
Collapse
|
46
|
Biomaterial Scaffolds in Regenerative Therapy of the Central Nervous System. BIOMED RESEARCH INTERNATIONAL 2018; 2018:7848901. [PMID: 29805977 PMCID: PMC5899851 DOI: 10.1155/2018/7848901] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Revised: 02/18/2018] [Accepted: 02/21/2018] [Indexed: 02/08/2023]
Abstract
The central nervous system (CNS) is the most important section of the nervous system as it regulates the function of various organs. Injury to the CNS causes impairment of neurological functions in corresponding sites and further leads to long-term patient disability. CNS regeneration is difficult because of its poor response to treatment and, to date, no effective therapies have been found to rectify CNS injuries. Biomaterial scaffolds have been applied with promising results in regeneration medicine. They also show great potential in CNS regeneration for tissue repair and functional recovery. Biomaterial scaffolds are applied in CNS regeneration predominantly as hydrogels and biodegradable scaffolds. They can act as cellular supportive scaffolds to facilitate cell infiltration and proliferation. They can also be combined with cell therapy to repair CNS injury. This review discusses the categories and progression of the biomaterial scaffolds that are applied in CNS regeneration.
Collapse
|
47
|
Huang Y, Jiang Y, Wu Q, Wu X, An X, Chubykin AA, Cheng JX, Xu XM, Yang C. Nanoladders Facilitate Directional Axonal Outgrowth and Regeneration. ACS Biomater Sci Eng 2018; 4:1037-1045. [DOI: 10.1021/acsbiomaterials.7b00981] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Affiliation(s)
- Yimin Huang
- Department of Chemistry, Boston University, 590 Commonwealth Avenue, Boston, Massachusetts 02215, United States
| | - Ying Jiang
- Department of Biomedical Engineering, Boston University, 44 Cummington Mall, Boston, Massachusetts 02215, United States
| | - Qiuyu Wu
- Department of Biological Sciences, Purdue University, 915 W. State Street, West Lafayette, Indiana 47907, United States
| | - Xiangbing Wu
- Stark Neurosciences Research Institute, Indiana University School of Medicine, 320 W. 15th Street, Indianapolis, Indiana 46202, United States
| | - Xingda An
- Department of Chemistry, Boston University, 590 Commonwealth Avenue, Boston, Massachusetts 02215, United States
| | - Alexander A. Chubykin
- Department of Biological Sciences, Purdue University, 915 W. State Street, West Lafayette, Indiana 47907, United States
| | - Ji-Xin Cheng
- Department of Chemistry, Boston University, 590 Commonwealth Avenue, Boston, Massachusetts 02215, United States
- Department of Electrical and Computer Engineering, Boston University, 8 St Mary’s Street, Boston, Massachusetts 02215, United States
- Department of Biomedical Engineering, Boston University, 44 Cummington Mall, Boston, Massachusetts 02215, United States
| | - Xiao-Ming Xu
- Stark Neurosciences Research Institute, Indiana University School of Medicine, 320 W. 15th Street, Indianapolis, Indiana 46202, United States
| | - Chen Yang
- Department of Chemistry, Boston University, 590 Commonwealth Avenue, Boston, Massachusetts 02215, United States
- Department of Electrical and Computer Engineering, Boston University, 8 St Mary’s Street, Boston, Massachusetts 02215, United States
| |
Collapse
|
48
|
Lin C, Liu C, Zhang L, Huang Z, Zhao P, Chen R, Pang M, Chen Z, He L, Luo C, Rong L, Liu B. Interaction of iPSC-derived neural stem cells on poly(L-lactic acid) nanofibrous scaffolds for possible use in neural tissue engineering. Int J Mol Med 2017; 41:697-708. [PMID: 29207038 PMCID: PMC5752187 DOI: 10.3892/ijmm.2017.3299] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 10/27/2017] [Indexed: 12/12/2022] Open
Abstract
Tissue engineering is a rapidly growing technological area for the regeneration and reconstruction of damage to the central nervous system. By combining seed cells with appropriate biomaterial scaffolds, tissue engineering has the ability to improve nerve regeneration and functional recovery. In the present study, mouse induced pluripotent stem cells (iPSCs) were generated from mouse embryonic fibroblasts (MEFs) with the non-integrating episomal vectors pCEP4-EO2S-ET2K and pCEP4-miR-302-367 cluster, and differentiated into neural stem cells (NSCs) as transplanting cells. Electrospinning was then used to fabricate randomly oriented poly(L-lactic acid) (PLLA) nanofibers and aligned PLLA nanofibers and assessed their cytocompatibility and neurite guidance effect with iPSC-derived NSCs (iNSCs). The results demonstrated that non-integrated iPSCs were effectively generated and differentiated into iNSCs. PLLA nanofiber scaffolds were able to promote the adhesion, growth, survival and proliferation of the iNSCs. Furthermore, compared with randomly oriented PLLA nanofibers, the aligned PLLA nanofibers greatly directed neurite outgrowth from the iNSCs and significantly promoted neurite growth along the nanofibrous alignment. Overall, these findings indicate the feasibility of using PLLA nanofiber scaffolds in combination with iNSCs in vitro and support their potential for use in nerve tissue engineering.
Collapse
Affiliation(s)
- Chengkai Lin
- Department of Spine Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Chang Liu
- Department of Spine Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Liangming Zhang
- Department of Spine Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Zhi Huang
- Department of Spine Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Peipei Zhao
- Department of Biomedical Engineering, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong 510632, P.R. China
| | - Ruiqiang Chen
- Department of Spine Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Mao Pang
- Department of Spine Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Zhenxiang Chen
- Department of Spine Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Liumin He
- Department of Biomedical Engineering, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong 510632, P.R. China
| | - Chunxiao Luo
- Department of Spine Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Limin Rong
- Department of Spine Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Bin Liu
- Department of Spine Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510630, P.R. China
| |
Collapse
|
49
|
Functional Test Scales for Evaluating Cell-Based Therapies in Animal Models of Spinal Cord Injury. Stem Cells Int 2017; 2017:5160261. [PMID: 29109741 PMCID: PMC5646345 DOI: 10.1155/2017/5160261] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 06/28/2017] [Accepted: 08/01/2017] [Indexed: 01/22/2023] Open
Abstract
Recently, spinal cord researchers have focused on multifaceted approaches for the treatment of spinal cord injury (SCI). However, as there is no cure for the deficits produced by SCI, various therapeutic strategies have been examined using animal models. Due to the lack of standardized functional assessment tools for use in such models, it is important to choose a suitable animal model and precise behavioral test when evaluating the efficacy of potential SCI treatments. In the present review, we discuss recent evidence regarding functional recovery in various animal models of SCI, summarize the representative models currently used, evaluate recent cell-based therapeutic approaches, and aim to identify the most precise and appropriate scales for functional assessment in such research.
Collapse
|
50
|
Sensharma P, Madhumathi G, Jayant RD, Jaiswal AK. Biomaterials and cells for neural tissue engineering: Current choices. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2017; 77:1302-1315. [PMID: 28532008 DOI: 10.1016/j.msec.2017.03.264] [Citation(s) in RCA: 121] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Accepted: 03/28/2017] [Indexed: 02/06/2023]
Abstract
The treatment of nerve injuries has taken a new dimension with the development of tissue engineering techniques. Prior to tissue engineering, suturing and surgery were the only options for effective treatment. With the advent of tissue engineering, it is now possible to design a scaffold that matches the exact biological and mechanical properties of the tissue. This has led to substantial reduction in the complications posed by surgeries and suturing to the patients. New synthetic and natural polymers are being applied to test their efficiency in generating an ideal scaffold. Along with these, cells and growth factors are also being incorporated to increase the efficiency of a scaffold. Efforts are being made to devise a scaffold that is biodegradable, biocompatible, conducting and immunologically inert. The ultimate goal is to exactly mimic the extracellular matrix in our body, and to elicit a combination of biochemical, topographical and electrical cues via various polymers, cells and growth factors, using which nerve regeneration can efficiently occur.
Collapse
Affiliation(s)
- Prerana Sensharma
- School of Biosciences and Technology, VIT University, Vellore 632014, Tamilnadu, India
| | - G Madhumathi
- School of Biosciences and Technology, VIT University, Vellore 632014, Tamilnadu, India
| | - Rahul D Jayant
- Center for Personalized Nanomedicine, Institute of Neuro-Immune Pharmacology, Department of Immunology, Herbert Wertheim College of Medicine, Florida International University (FIU), Miami, FL 33199, USA
| | - Amit K Jaiswal
- Centre for Biomaterials, Cellular and Molecular Theranostics, VIT University, Vellore 632014, Tamilnadu, India.
| |
Collapse
|