1
|
Ma D, Fu C, Li F, Ruan R, Lin Y, Li X, Li M, Zhang J. Functional biomaterials for modulating the dysfunctional pathological microenvironment of spinal cord injury. Bioact Mater 2024; 39:521-543. [PMID: 38883317 PMCID: PMC11179178 DOI: 10.1016/j.bioactmat.2024.04.015] [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: 01/13/2024] [Revised: 04/13/2024] [Accepted: 04/14/2024] [Indexed: 06/18/2024] Open
Abstract
Spinal cord injury (SCI) often results in irreversible loss of sensory and motor functions, and most SCIs are incurable with current medical practice. One of the hardest challenges in treating SCI is the development of a dysfunctional pathological microenvironment, which mainly comprises excessive inflammation, deposition of inhibitory molecules, neurotrophic factor deprivation, glial scar formation, and imbalance of vascular function. To overcome this challenge, implantation of functional biomaterials at the injury site has been regarded as a potential treatment for modulating the dysfunctional microenvironment to support axon regeneration, remyelination at injury site, and functional recovery after SCI. This review summarizes characteristics of dysfunctional pathological microenvironment and recent advances in biomaterials as well as the technologies used to modulate inflammatory microenvironment, regulate inhibitory microenvironment, and reshape revascularization microenvironment. Moreover, technological limitations, challenges, and future prospects of functional biomaterials to promote efficient repair of SCI are also discussed. This review will aid further understanding and development of functional biomaterials to regulate pathological SCI microenvironment.
Collapse
Affiliation(s)
- Dezun Ma
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, 1 Qiuyang Road, Fuzhou, Fujian, 350122, PR China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, PR China
| | - Changlong Fu
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, 1 Qiuyang Road, Fuzhou, Fujian, 350122, PR China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, PR China
| | - Fenglu Li
- College of Chemical Engineering, Fuzhou University, 2 Xueyuan Road, Fuzhou, 350108, PR China
- Qingyuan Innovation Laboratory, 1 Xueyuan Road, Quanzhou, 362801, PR China
| | - Renjie Ruan
- College of Chemical Engineering, Fuzhou University, 2 Xueyuan Road, Fuzhou, 350108, PR China
- Qingyuan Innovation Laboratory, 1 Xueyuan Road, Quanzhou, 362801, PR China
| | - Yanming Lin
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, 1 Qiuyang Road, Fuzhou, Fujian, 350122, PR China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, PR China
| | - Xihai Li
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, 1 Qiuyang Road, Fuzhou, Fujian, 350122, PR China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, PR China
| | - Min Li
- Fujian Children's Hospital, Fujian Branch of Shanghai Children's Medical Center, 966 Hengyu Road, Fuzhou, 350014, PR China
- Fujian Maternity and Child Health Hospital, 111 Daoshan Road, Fuzhou, 350005, PR China
- College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, 111 Daoshan Road, Fuzhou, 350005, PR China
| | - Jin Zhang
- College of Chemical Engineering, Fuzhou University, 2 Xueyuan Road, Fuzhou, 350108, PR China
- Qingyuan Innovation Laboratory, 1 Xueyuan Road, Quanzhou, 362801, PR China
| |
Collapse
|
2
|
Baroudi M, Rezk A, Daher M, Balmaceno-Criss M, Gregoryczyk JG, Sharma Y, McDonald CL, Diebo BG, Daniels AH. Management of traumatic spinal cord injury: A current concepts review of contemporary and future treatment. Injury 2024; 55:111472. [PMID: 38460480 DOI: 10.1016/j.injury.2024.111472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 02/03/2024] [Accepted: 02/25/2024] [Indexed: 03/11/2024]
Abstract
Spinal Cord Injury (SCI) is a condition leading to inflammation, edema, and dysfunction of the spinal cord, most commonly due to trauma, tumor, infection, or vascular disturbance. Symptoms include sensory and motor loss starting at the level of injury; the extent of damage depends on injury severity as detailed in the ASIA score. In the acute setting, maintaining mean arterial pressure (MAP) higher than 85 mmHg for up to 7 days following injury is preferred; although caution must be exercised when using vasopressors such as phenylephrine due to serious side effects such as pulmonary edema and death. Decompression surgery (DS) may theoretically relieve edema and reduce intraspinal pressure, although timing of surgery remains a matter of debate. Methylprednisolone (MP) is currently used due to its ability to reduce inflammation but more recent studies question its clinical benefits, especially with inconsistency in recommending it nationally and internationally. The choice of MP is further complicated by conflicting evidence for optimal timing to initiate treatment, and by the reported observation that higher doses are correlated with increased risk of complications. Thyrotropin-releasing hormone may be beneficial in less severe injuries. Finally, this review discusses many options currently being researched and have shown promising pre-clinical results.
Collapse
Affiliation(s)
- Makeen Baroudi
- Department of Orthopedic Surgery, The Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Anna Rezk
- Department of Orthopedic Surgery, The Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Mohammad Daher
- Department of Orthopedic Surgery, The Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Mariah Balmaceno-Criss
- Department of Orthopedic Surgery, The Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Jerzy George Gregoryczyk
- Department of Orthopedic Surgery, The Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Yatharth Sharma
- Department of Orthopedic Surgery, The Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Christopher L McDonald
- Department of Orthopedic Surgery, The Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Bassel G Diebo
- Department of Orthopedic Surgery, The Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Alan H Daniels
- Department of Orthopedic Surgery, The Warren Alpert Medical School of Brown University, Providence, RI, USA.
| |
Collapse
|
3
|
Agosti E, Zeppieri M, Pagnoni A, Fontanella MM, Fiorindi A, Ius T, Panciani PP. Current status and future perspectives on stem cell transplantation for spinal cord injury. World J Transplant 2024; 14:89674. [PMID: 38576751 PMCID: PMC10989472 DOI: 10.5500/wjt.v14.i1.89674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 12/04/2023] [Accepted: 12/29/2023] [Indexed: 03/15/2024] Open
Abstract
BACKGROUND Previous assessments of stem cell therapy for spinal cord injuries (SCI) have encountered challenges and constraints. Current research primarily emphasizes safety in early-phase clinical trials, while systematic reviews prioritize effectiveness, often overlooking safety and translational feasibility. This situation prompts inquiries regarding the readiness for clinical adoption. AIM To offer an up-to-date systematic literature review of clinical trial results con cerning stem cell therapy for SCI. METHODS A systematic search was conducted across major medical databases [PubMed, Embase, Reference Citation Analysis (RCA), and Cochrane Library] up to October 14, 2023. The search strategy utilized relevant Medical Subject Heading (MeSH) terms and keywords related to "spinal cord", "injury", "clinical trials", "stem cells", "functional outcomes", and "adverse events". Studies included in this review consisted of randomized controlled trials and non-randomized controlled trials reporting on the use of stem cell therapies for the treatment of SCI. RESULTS In a comprehensive review of 66 studies on stem cell therapies for SCI, 496 papers were initially identified, with 237 chosen for full-text analysis. Among them, 236 were deemed eligible after excluding 170 for various reasons. These studies encompassed 1086 patients with varying SCI levels, with cervical injuries being the most common (42.2%). Bone marrow stem cells were the predominant stem cell type used (71.1%), with various administration methods. Follow-up durations averaged around 84.4 months. The 32.7% of patients showed functional impro vement from American spinal injury association Impairment Scale (AIS) A to B, 40.8% from AIS A to C, 5.3% from AIS A to D, and 2.1% from AIS B to C. Sensory improvements were observed in 30.9% of patients. A relatively small number of adverse events were recorded, including fever (15.1%), headaches (4.3%), muscle tension (3.1%), and dizziness (2.6%), highlighting the potential for SCI recovery with stem cell therapy. CONCLUSION In the realm of SCI treatment, stem cell-based therapies show promise, but clinical trials reveal potential adverse events and limitations, underscoring the need for meticulous optimization of transplantation conditions and parameters, caution against swift clinical implementation, a deeper understanding of SCI pathophysiology, and addressing ethical, tumorigenicity, immunogenicity, and immunotoxicity concerns before gradual and careful adoption in clinical practice.
Collapse
Affiliation(s)
- Edoardo Agosti
- Division of Neurosurgery, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia 25123, Italy
| | - Marco Zeppieri
- Department of Ophthalmology, University Hospital of Udine, Udine 33100, Italy
| | - Andrea Pagnoni
- Division of Neurosurgery, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia 25123, Italy
| | - Marco Maria Fontanella
- Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia, Brescia 25123, BS, Italy
| | - Alessandro Fiorindi
- Division of Neurosurgery, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia 25123, Italy
| | - Tamara Ius
- Neurosurgery Unit, Head-Neck and NeuroScience Department, University Hospital of Udine, Udine 33100, Italy
| | - Pier Paolo Panciani
- Division of Neurosurgery, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia 25123, Italy
| |
Collapse
|
4
|
Cong M, Wu X, Zhu L, Gu G, Ding F, Li G, Shi H. Anisotropic microtopography surface of chitosan scaffold regulating skin precursor-derived Schwann cells towards repair phenotype promotes neural regeneration. Regen Biomater 2024; 11:rbae005. [PMID: 38414797 PMCID: PMC10898340 DOI: 10.1093/rb/rbae005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 01/12/2024] [Accepted: 01/16/2024] [Indexed: 02/29/2024] Open
Abstract
For repairing peripheral nerve and spinal cord defects, biomaterial scaffold-based cell-therapy was emerged as an effective strategy, requiring the positive response of seed cells to biomaterial substrate and environment signals. Previous work highlighted that the imposed surface properties of scaffold could provide important guidance cues to adhered cells for polarization. However, the insufficiency of native Schwann cells and unclear cellular response mechanisms remained to be addressed. Given that, this study aimed to illuminate the micropatterned chitosan-film action on the rat skin precursor-derived Schwann cells (SKP-SCs). Chitosan-film with different ridge/groove size was fabricated and applied for the SKP-SCs induction. Results indicated that SKP-SCs cultured on 30 μm size microgroove surface showed better oriented alignment phenotype. Induced SKP-SCs presented similar genic phenotype as repair Schwann cells, increasing expression of c-Jun, neural cell adhesion molecule, and neurotrophic receptor p75. Moreover, SKP-SC-secretome was subjected to cytokine array GS67 assay, data indicated the regulation of paracrine phenotype, a panel of cytokines was verified up-regulated at secreted level and gene expression level in induced SKP-SCs. These up-regulated cytokines exhibit a series of promotive neural regeneration functions, including cell survival, cell migration, cell proliferation, angiogenesis, axon growth, and cellular organization etc. through bioinformatics analysis. Furthermore, the effectively polarized SKP-SCs-sourced secretome, promoted the proliferation and migration capacity of the primarily cultured native rat Schwann cells, and augmented neurites growth of the cultured motoneurons, as well as boosted axonal regrowth of the axotomy-injured motoneurons. Taken together, SKP-SCs obtained pro-neuroregeneration phenotype in adaptive response to the anisotropic topography surface of chitosan-film, displayed the oriented parallel growth, the transition towards repair Schwann cell genic phenotype, and the enhanced paracrine effect on neural regeneration. This study provided novel insights into the potency of anisotropic microtopography surface to Schwann-like cells phenotype regulation, that facilitating to provide promising engineered cell-scaffold in neural injury therapies.
Collapse
Affiliation(s)
- Meng Cong
- Key Laboratory of Neuroregenration of Jiangsu and Ministry of Education and Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China
| | - Xia Wu
- Key Laboratory of Neuroregenration of Jiangsu and Ministry of Education and Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China
| | - Lingjie Zhu
- Department of Pathophysiology, School of Medicine, Nantong University, Nantong 226001, China
| | - Guohao Gu
- Key Laboratory of Neuroregenration of Jiangsu and Ministry of Education and Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China
| | - Fei Ding
- Key Laboratory of Neuroregenration of Jiangsu and Ministry of Education and Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China
| | - Guicai Li
- Key Laboratory of Neuroregenration of Jiangsu and Ministry of Education and Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China
| | - Haiyan Shi
- Key Laboratory of Neuroregenration of Jiangsu and Ministry of Education and Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China
- Department of Pathophysiology, School of Medicine, Nantong University, Nantong 226001, China
| |
Collapse
|
5
|
Aderinto N, Abdulbasit MO, Olatunji D. Stem cell-based combinatorial therapies for spinal cord injury: a narrative review of current research and future directions. Ann Med Surg (Lond) 2023; 85:3943-3954. [PMID: 37554849 PMCID: PMC10406006 DOI: 10.1097/ms9.0000000000001034] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 06/22/2023] [Indexed: 08/10/2023] Open
Abstract
Spinal cord injury (SCI) is a devastating condition that can result in lifelong disability. Despite significant progress in SCI research, current treatments only offer limited functional recovery. Stem cell-based combinatorial therapies have emerged promising to enhance neural repair and regeneration after SCI. Combining stem cells with growth factors, biomaterials, and other therapeutic agents can improve outcomes by providing a multifaceted approach to neural repair. However, several challenges must be addressed before these therapies can be widely adopted in clinical practice. Standardisation of stem cell isolation, characterisation, and production protocols ensures consistency and safety in clinical trials. Developing appropriate animal models that accurately mimic human SCI is crucial for successfully translating these therapies. Additionally, optimal delivery methods and biomaterials that support the survival and integration of stem cells into injured tissue must be identified. Despite these challenges, stem cell-based combinatorial therapies for SCI hold great promise. Innovative approaches such as gene editing and the use of neural tissue engineering may further enhance the efficacy of these therapies. Further research and development in this area are critical to advancing the field and providing effective therapies for SCI patients. This paper discusses the current evidence and challenges from the literature on the potential of stem cell-based combinatorial therapies for SCI.
Collapse
Affiliation(s)
- Nicholas Aderinto
- Department of Medicine and Surgery, Ladoke Akintola University of Technology, Ogbomoso
| | | | - Deji Olatunji
- Department of Medicine and Surgery, University of Ilorin, Ilorin, Nigeria
| |
Collapse
|
6
|
Zhang C, Morozova AY, Abakumov MA, Mel'nikov PA, Gabashvili AN, Chekhonin VP. Evaluation of the Optimal Number of Implanted Mesenchymal Stem Cells for the Treatment of Post-Traumatic Syrinx and Recovery of Motor Activity after Chronic Spinal Cord Injury. Bull Exp Biol Med 2023; 175:557-568. [PMID: 37773573 DOI: 10.1007/s10517-023-05904-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Indexed: 10/01/2023]
Abstract
The present work aims at determining the most effective dose (number) of mesenchymal stem cells (MSC) for its transplantation in order to treat chronic spinal cord injury (SCI) in mature Sprague-Dawley rats (n=24). MSC were obtained from bone marrow of 4-6-month-old Sprague-Dawley rats. Four weeks after SCI, MSC suspension (4 μl) was injected to experimental animals into the injured area in doses of 4×105, 8×105, or 106. Using MRI, diffusion tensor imaging (DTI), diffusion tensor tractography (DTT), immunohistochemistry, histological staining, and behavioral tests, we studied the effect of transplantation of MSC in different doses on the following parameters in rats with SCI: the size of lesion cavity and post-traumatic syrinx (PTS), glial scar formation, neuronal fibers remodeling, axonal regeneration and sprouting, vascularization, expression of neuronal factors, and motor functions. MSC administration improved motor function in rats after SCI due to stimulation of regeneration and sprouting of the axons, enhanced recovery of locomotor functions, reduction of PTS and the glial scar, and stimulation of vascularization and expression of the neurotrophic factors. The effects of MSC were dose-dependent; the most effective dose was 106 cells.
Collapse
Affiliation(s)
- C Zhang
- Department of Medicinal Nanobiotechnology, Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, Moscow, Russia.
- Department of Basic and Applied Neurobiology, V. P. Serbsky National Medical Research Center for Psychiatry and Narcology, Ministry of Health of the Russian Federation, Moscow, Russia.
| | - A Yu Morozova
- Department of Basic and Applied Neurobiology, V. P. Serbsky National Medical Research Center for Psychiatry and Narcology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - M A Abakumov
- Department of Medicinal Nanobiotechnology, Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, Moscow, Russia
| | - P A Mel'nikov
- Department of Medicinal Nanobiotechnology, Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, Moscow, Russia
| | - A N Gabashvili
- Department of Medicinal Nanobiotechnology, Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, Moscow, Russia
| | - V P Chekhonin
- Department of Medicinal Nanobiotechnology, Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, Moscow, Russia
- Department of Basic and Applied Neurobiology, V. P. Serbsky National Medical Research Center for Psychiatry and Narcology, Ministry of Health of the Russian Federation, Moscow, Russia
| |
Collapse
|
7
|
Xiang W, Cao H, Tao H, Jin L, Luo Y, Tao F, Jiang T. Applications of chitosan-based biomaterials: From preparation to spinal cord injury neuroprosthetic treatment. Int J Biol Macromol 2023; 230:123447. [PMID: 36708903 DOI: 10.1016/j.ijbiomac.2023.123447] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 12/04/2022] [Accepted: 01/24/2023] [Indexed: 01/27/2023]
Abstract
Spinal cord injury (SCI)-related disabilities are a serious problem in the modern society. Further, the treatment of SCI is highly challenging and is urgently required in clinical practice. Research on nerve tissue engineering is an emerging approach for improving the treatment outcomes of SCI. Chitosan (CS) is a cationic polysaccharide derived from natural biomaterials. Chitosan has been found to exhibit excellent biological properties, such as nontoxicity, biocompatibility, biodegradation, and antibacterial activity. Recently, chitosan-based biomaterials have attracted significant attention for SCI repair in nerve tissue engineering applications. These studies revealed that chitosan-based biomaterials have various functions and mechanisms to promote SCI repair, such as promoting neural cell growth, guiding nerve tissue regeneration, delivering nerve growth factors, and as a vector for gene therapy. Chitosan-based biomaterials have proven to have excellent potential for the treatment of SCI. This review aims to introduce the recent advances in chitosan-based biomaterials for SCI treatment and to highlight the prospects for further application.
Collapse
Affiliation(s)
- Wei Xiang
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan University, Wuhan 430060, China
| | - Hui Cao
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan University, Wuhan 430060, China
| | - Hai Tao
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan University, Wuhan 430060, China
| | - Lin Jin
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan University, Wuhan 430060, China
| | - Yue Luo
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan University, Wuhan 430060, China
| | - Fenghua Tao
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan University, Wuhan 430060, China.
| | - Ting Jiang
- Department of Neurological Rehabilitation, Zhongnan Hospital of Wuhan University, Wuhan 430071, China.
| |
Collapse
|
8
|
Pourkhodadad S, Hosseinkazemi H, Bonakdar S, Nekounam H. Biomimetic engineered approaches for neural tissue engineering: Spinal cord injury. J Biomed Mater Res B Appl Biomater 2023; 111:701-716. [PMID: 36214332 DOI: 10.1002/jbm.b.35171] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 07/16/2022] [Accepted: 09/03/2022] [Indexed: 01/21/2023]
Abstract
The healing process for spinal cord injuries is complex and presents many challenges. Current advances in nerve regeneration are based on promising tissue engineering techniques, However, the chances of success depend on better mimicking the extracellular matrix (ECM) of neural tissue and better supporting neurons in a three-dimensional environment. The ECM provides excellent biological conditions, including desirable morphological features, electrical conductivity, and chemical compositions for neuron attachment, proliferation and function. This review outlines the rationale for developing a construct for neuron regrowth in spinal cord injury using appropriate biomaterials and scaffolding techniques.
Collapse
Affiliation(s)
| | - Hessam Hosseinkazemi
- Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| | - Shahin Bonakdar
- National Cell Bank Department, Pasteur Institute of Iran, Tehran, Iran
| | - Houra Nekounam
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
9
|
Khaing ZZ, Chen JY, Safarians G, Ezubeik S, Pedroncelli N, Duquette RD, Prasse T, Seidlits SK. Clinical Trials Targeting Secondary Damage after Traumatic Spinal Cord Injury. Int J Mol Sci 2023; 24:3824. [PMID: 36835233 PMCID: PMC9960771 DOI: 10.3390/ijms24043824] [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: 01/17/2023] [Revised: 02/06/2023] [Accepted: 02/09/2023] [Indexed: 02/17/2023] Open
Abstract
Spinal cord injury (SCI) often causes loss of sensory and motor function resulting in a significant reduction in quality of life for patients. Currently, no therapies are available that can repair spinal cord tissue. After the primary SCI, an acute inflammatory response induces further tissue damage in a process known as secondary injury. Targeting secondary injury to prevent additional tissue damage during the acute and subacute phases of SCI represents a promising strategy to improve patient outcomes. Here, we review clinical trials of neuroprotective therapeutics expected to mitigate secondary injury, focusing primarily on those in the last decade. The strategies discussed are broadly categorized as acute-phase procedural/surgical interventions, systemically delivered pharmacological agents, and cell-based therapies. In addition, we summarize the potential for combinatorial therapies and considerations.
Collapse
Affiliation(s)
- Zin Z. Khaing
- Department of Neurological Surgery, University of Washington, Seattle, WA 98195, USA
| | - Jessica Y. Chen
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Gevick Safarians
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Sohib Ezubeik
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Nicolas Pedroncelli
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Rebecca D. Duquette
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095, USA
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX 78712, USA
| | - Tobias Prasse
- Department of Neurological Surgery, University of Washington, Seattle, WA 98195, USA
- Department of Orthopedics and Trauma Surgery, University of Cologne, 50931 Cologne, Germany
| | - Stephanie K. Seidlits
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095, USA
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX 78712, USA
| |
Collapse
|
10
|
Suzuki H, Imajo Y, Funaba M, Ikeda H, Nishida N, Sakai T. Current Concepts of Biomaterial Scaffolds and Regenerative Therapy for Spinal Cord Injury. Int J Mol Sci 2023; 24:ijms24032528. [PMID: 36768846 PMCID: PMC9917245 DOI: 10.3390/ijms24032528] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/05/2023] [Accepted: 01/11/2023] [Indexed: 02/03/2023] Open
Abstract
Spinal cord injury (SCI) is a catastrophic condition associated with significant neurological deficit and social and financial burdens. It is currently being managed symptomatically, with no real therapeutic strategies available. In recent years, a number of innovative regenerative strategies have emerged and have been continuously investigated in preclinical research and clinical trials. In the near future, several more are expected to come down the translational pipeline. Among ongoing and completed trials are those reporting the use of biomaterial scaffolds. The advancements in biomaterial technology, combined with stem cell therapy or other regenerative therapy, can now accelerate the progress of promising novel therapeutic strategies from bench to bedside. Various types of approaches to regeneration therapy for SCI have been combined with the use of supportive biomaterial scaffolds as a drug and cell delivery system to facilitate favorable cell-material interactions and the supportive effect of neuroprotection. In this review, we summarize some of the most recent insights of preclinical and clinical studies using biomaterial scaffolds in regenerative therapy for SCI and summarized the biomaterial strategies for treatment with simplified results data. One hundred and sixty-eight articles were selected in the present review, in which we focused on biomaterial scaffolds. We conducted our search of articles using PubMed and Medline, a medical database. We used a combination of "Spinal cord injury" and ["Biomaterial", or "Scaffold"] as search terms and searched articles published up until 30 April 2022. Successful future therapies will require these biomaterial scaffolds and other synergistic approaches to address the persistent barriers to regeneration, including glial scarring, the loss of a structural framework, and biocompatibility. This database could serve as a benchmark to progress in future clinical trials for SCI using biomaterial scaffolds.
Collapse
|
11
|
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
|
12
|
Multiple strategies enhance the efficacy of MSCs transplantation for spinal cord injury. Biomed Pharmacother 2023; 157:114011. [PMID: 36410123 DOI: 10.1016/j.biopha.2022.114011] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 11/05/2022] [Accepted: 11/11/2022] [Indexed: 11/19/2022] Open
Abstract
Spinal cord injury (SCI) is a serious complication of the central nervous system (CNS) after spine injury, often resulting in severe sensory, motor, and autonomic dysfunction below the level of injury. To date, there is no effective treatment strategy for SCI. Recently, stem cell therapy has brought hope to patients with neurological diseases. Mesenchymal stem cells (MSCs) are considered to be the most promising source of cellular therapy after SCI due to their immunomodulatory, neuroprotective and angiogenic potential. Considering the limited therapeutic effect of MSCs due to the complex pathophysiological environment following SCI, this paper not only reviews the specific mechanism of MSCs to facilitate SCI repair, but also further discusses the research status of these pluripotent stem cells combined with other therapeutic approaches to promote anatomical and functional recovery post-SCI.
Collapse
|
13
|
Shang Z, Wang M, Zhang B, Wang X, Wanyan P. Clinical translation of stem cell therapy for spinal cord injury still premature: results from a single-arm meta-analysis based on 62 clinical trials. BMC Med 2022; 20:284. [PMID: 36058903 PMCID: PMC9442938 DOI: 10.1186/s12916-022-02482-2] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 07/14/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND How much scientific evidence is there to show that stem cell therapy is sufficient in preclinical and clinical studies of spinal cord injury before it is translated into clinical practice? This is a complicated problem. A single, small-sample clinical trial is difficult to answer, and accurate insights into this question can only be given by systematically evaluating all the existing evidence. METHODS The PubMed, Ovid-Embase, Web of Science, and Cochrane databases were searched from inception to February 10, 2022. Two independent reviewers performed the literature search, identified and screened the studies, and performed a quality assessment and data extraction. RESULTS In total, 62 studies involving 2439 patients were included in the analysis. Of these, 42 were single-arm studies, and 20 were controlled studies. The meta-analysis showed that stem cells improved the ASIA impairment scale score by at least one grade in 48.9% [40.8%, 56.9%] of patients with spinal cord injury. Moreover, the rate of improvement in urinary and gastrointestinal system function was 42.1% [27.6%, 57.2%] and 52.0% [23.6%, 79.8%], respectively. However, 28 types of adverse effects were observed to occur due to stem cells and transplantation procedures. Of these, neuropathic pain, abnormal feeling, muscle spasms, vomiting, and urinary tract infection were the most common, with an incidence of > 20%. While no serious adverse effects such as tumorigenesis were reported, this could be due to the insufficient follow-up period. CONCLUSIONS Overall, the results demonstrated that although the efficacy of stem cell therapy is encouraging, the subsequent adverse effects remain concerning. In addition, the clinical trials had problems such as small sample sizes, poor design, and lack of prospective registration, control, and blinding. Therefore, the current evidence is not sufficiently strong to support the clinical translation of stem cell therapy for spinal cord injury, and several problems remain. Additional well-designed animal experiments and high-quality clinical studies are warranted to address these issues.
Collapse
Affiliation(s)
- Zhizhong Shang
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, China
| | - Mingchuan Wang
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, China
| | - Baolin Zhang
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, China
| | - Xin Wang
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, China.
- Chengren Institute of Traditional Chinese Medicine, Lanzhou, 730000, Gansu Province, China.
- Department of Spine, Changzheng Hospital, Naval Medical University, Shanghai, 200003, China.
| | - Pingping Wanyan
- Gansu University of Chinese Medicine, Lanzhou, 730000, China
- The Second Hospital of Lanzhou University, Lanzhou, 730000, China
| |
Collapse
|
14
|
Van den Bos J, Ouaamari YE, Wouters K, Cools N, Wens I. Are Cell-Based Therapies Safe and Effective in the Treatment of Neurodegenerative Diseases? A Systematic Review with Meta-Analysis. Biomolecules 2022; 12:340. [PMID: 35204840 PMCID: PMC8869169 DOI: 10.3390/biom12020340] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 02/14/2022] [Accepted: 02/17/2022] [Indexed: 12/13/2022] Open
Abstract
Over the past two decades, significant advances have been made in the field of regenerative medicine. However, despite being of the utmost clinical urgency, there remains a paucity of therapeutic strategies for conditions with substantial neurodegeneration such as (progressive) multiple sclerosis (MS), spinal cord injury (SCI), Parkinson's disease (PD) and Alzheimer's disease (AD). Different cell types, such as mesenchymal stromal cells (MSC), neuronal stem cells (NSC), olfactory ensheathing cells (OEC), neurons and a variety of others, already demonstrated safety and regenerative or neuroprotective properties in the central nervous system during the preclinical phase. As a result of these promising findings, in recent years, these necessary types of cell therapies have been intensively tested in clinical trials to establish whether these results could be confirmed in patients. However, extensive research is still needed regarding elucidating the exact mechanism of action, possible immune rejection, functionality and survival of the administered cells, dose, frequency and administration route. To summarize the current state of knowledge, we conducted a systematic review with meta-analysis. A total of 27,043 records were reviewed by two independent assessors and 71 records were included in the final quantitative analysis. These results show that the overall frequency of serious adverse events was low: 0.03 (95% CI: 0.01-0.08). In addition, several trials in MS and SCI reported efficacy data, demonstrating some promising results on clinical outcomes. All randomized controlled studies were at a low risk of bias due to appropriate blinding of the treatment, including assessors and patients. In conclusion, cell-based therapies in neurodegenerative disease are safe and feasible while showing promising clinical improvements. Nevertheless, given their high heterogeneity, the results require a cautious approach. We advocate for the harmonization of study protocols of trials investigating cell-based therapies in neurodegenerative diseases, adverse event reporting and investigation of clinical outcomes.
Collapse
Affiliation(s)
- Jasper Van den Bos
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Universiteitsplein 1, B-2610 Antwerpen, Belgium; (Y.E.O.); (N.C.); (I.W.)
| | - Yousra El Ouaamari
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Universiteitsplein 1, B-2610 Antwerpen, Belgium; (Y.E.O.); (N.C.); (I.W.)
| | - Kristien Wouters
- Clinical Trial Center (CTC), CRC Antwerp, Antwerp University Hospital, University of Antwerp, Drie Eikenstraat 655, B-2650 Edegem, Belgium;
| | - Nathalie Cools
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Universiteitsplein 1, B-2610 Antwerpen, Belgium; (Y.E.O.); (N.C.); (I.W.)
- Center for Cell Therapy and Regenerative Medicine (CCRG), Antwerp University Hospital, Drie Eikenstraat 655, B-2650 Edegem, Belgium
| | - Inez Wens
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Universiteitsplein 1, B-2610 Antwerpen, Belgium; (Y.E.O.); (N.C.); (I.W.)
| |
Collapse
|
15
|
Suzuki H, Imajo Y, Funaba M, Nishida N, Sakamoto T, Sakai T. Current Concepts of Neural Stem/Progenitor Cell Therapy for Chronic Spinal Cord Injury. Front Cell Neurosci 2022; 15:794692. [PMID: 35185471 PMCID: PMC8850278 DOI: 10.3389/fncel.2021.794692] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 12/20/2021] [Indexed: 11/13/2022] Open
Abstract
Chronic spinal cord injury (SCI) is a devastating condition that results in major neurological deficits and social burden. It continues to be managed symptomatically, and no real therapeutic strategies have been devised for its treatment. Neural stem/neural progenitor cells (NSCs/NPCs) being used for the treatment of chronic SCI in experimental SCI models can not only replace the lost cells and remyelinate axons in the injury site but also support their growth and provide neuroprotective factors. Currently, several clinical studies using NSCs/NPCs are underway worldwide. NSCs/NPCs also have the potential to differentiate into all three neuroglial lineages to regenerate neural circuits, demyelinate denuded axons, and provide trophic support to endogenous cells. This article explains the challenging pathophysiology of chronic SCI and discusses key NSC/NPC-based techniques having the greatest potential for translation over the next decade.
Collapse
|
16
|
Kiyotake EA, Martin MD, Detamore MS. Regenerative rehabilitation with conductive biomaterials for spinal cord injury. Acta Biomater 2022; 139:43-64. [PMID: 33326879 DOI: 10.1016/j.actbio.2020.12.021] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 11/24/2020] [Accepted: 12/09/2020] [Indexed: 02/07/2023]
Abstract
The individual approaches of regenerative medicine efforts alone and rehabilitation efforts alone have not yet fully restored function after severe spinal cord injury (SCI). Regenerative rehabilitation may be leveraged to promote regeneration of the spinal cord tissue, and promote reorganization of the regenerated neural pathways and intact spinal circuits for better functional recovery for SCI. Conductive biomaterials may be a linchpin that empowers the synergy between regenerative medicine and rehabilitation approaches, as electrical stimulation applied to the spinal cord could facilitate neural reorganization. In this review, we discuss current regenerative medicine approaches in clinical trials and the rehabilitation, or neuromodulation, approaches for SCI, along with their respective translational limitations. Furthermore, we review the translational potential, in a surgical context, of conductive biomaterials (e.g., conductive polymers, carbon-based materials, metallic nanoparticle-based materials) as they pertain to SCI. While pre-formed scaffolds may be difficult to translate to human contusion SCIs, injectable composites that contain blended conductive components and can form within the injury may be more translational. However, given that there are currently no in vivo SCI studies that evaluated conductive materials combined with rehabilitation approaches, we discuss several limitations of conductive biomaterials, including demonstrating safety and efficacy, that will need to be addressed in the future for conductive biomaterials to become SCI therapeutics. Even so, the use of conductive biomaterials creates a synergistic opportunity to merge the fields of regenerative medicine and rehabilitation and redefine what regenerative rehabilitation means for the spinal cord. STATEMENT OF SIGNIFICANCE: For spinal cord injury (SCI), the individual approaches of regenerative medicine and rehabilitation are insufficient to fully restore functional recovery; however, the goal of regenerative rehabilitation is to combine these two disparate fields to maximize the functional outcomes. Concepts similar to regenerative rehabilitation for SCI have been discussed in several reviews, but for the first time, this review considers how conductive biomaterials may synergize the two approaches. We cover current regenerative medicine and rehabilitation approaches for SCI, and the translational advantages and disadvantages, in a surgical context, of conductive biomaterials used in biomedical applications that may be additionally applied to SCI. Furthermore, we identify the current limitations and translational challenges for conductive biomaterials before they may become therapeutics for SCI.
Collapse
|
17
|
Numerous nanoparticles as drug delivery system to control secondary immune response and promote spinal cord injury regeneration. Process Biochem 2022. [DOI: 10.1016/j.procbio.2021.11.025] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
18
|
Suzuki H, Sakai T. Current Concepts of Stem Cell Therapy for Chronic Spinal Cord Injury. Int J Mol Sci 2021; 22:ijms22147435. [PMID: 34299053 PMCID: PMC8308009 DOI: 10.3390/ijms22147435] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 07/08/2021] [Accepted: 07/09/2021] [Indexed: 12/14/2022] Open
Abstract
Chronic spinal cord injury (SCI) is a catastrophic condition associated with significant neurological deficit and social and financial burdens. It is currently being managed symptomatically with no real therapeutic strategies available. In recent years, a number of innovative regenerative strategies have emerged and have been continuously investigated in clinical trials. In addition, several more are coming down the translational pipeline. Among ongoing and completed trials are those reporting the use of mesenchymal stem cells, neural stem/progenitor cells, induced pluripotent stem cells, olfactory ensheathing cells, and Schwann cells. The advancements in stem cell technology, combined with the powerful neuroimaging modalities, can now accelerate the pathway of promising novel therapeutic strategies from bench to bedside. Various combinations of different molecular therapies have been combined with supportive scaffolds to facilitate favorable cell–material interactions. In this review, we summarized some of the most recent insights into the preclinical and clinical studies using stem cells and other supportive drugs to unlock the microenvironment in chronic SCI to treat patients with this condition. Successful future therapies will require these stem cells and other synergistic approaches to address the persistent barriers to regeneration, including glial scarring, loss of structural framework, and immunorejection.
Collapse
|
19
|
Monje PV, Deng L, Xu XM. Human Schwann Cell Transplantation for Spinal Cord Injury: Prospects and Challenges in Translational Medicine. Front Cell Neurosci 2021; 15:690894. [PMID: 34220455 PMCID: PMC8249939 DOI: 10.3389/fncel.2021.690894] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Accepted: 05/21/2021] [Indexed: 01/18/2023] Open
Abstract
The benefits of transplanting cultured Schwann cells (SCs) for the treatment of spinal cord injury (SCI) have been systematically investigated in experimental animals since the early 1990s. Importantly, human SC (hSC) transplantation for SCI has advanced to clinical testing and safety has been established via clinical trials conducted in the USA and abroad. However, multiple barriers must be overcome to enable accessible and effective treatments for SCI patients. This review presents available information on hSC transplantation for SCI with the intention to uncover gaps in our knowledge and discuss areas for future development. To this end, we introduce the historical progression of the work that supports existing and prospective clinical initiatives and explain the reasons for the choice of hSCs while also addressing their limitations as cell therapy products. A search of the relevant literature revealed that rat SCs have served as a preclinical model of reference since the onset of investigations, and that hSC transplants are relatively understudied, possibly due to the sophisticated resources and expertise needed for the traditional processing of hSC cultures from human nerves. In turn, we reason that additional experimentation and a reexamination of the available data are needed to understand the therapeutic value of hSC transplants taking into consideration that the manufacturing of the hSCs themselves may require further development for extended uses in basic research and clinical settings.
Collapse
Affiliation(s)
- Paula V. Monje
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Lingxiao Deng
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Xiao-Ming Xu
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
20
|
Yang Y, Cao TT, Tian ZM, Gao H, Wen HQ, Pang M, He WJ, Wang NX, Chen YY, Wang Y, Li H, Lin JW, Kang Z, Li MM, Liu B, Rong LM. Subarachnoid transplantation of human umbilical cord mesenchymal stem cell in rodent model with subacute incomplete spinal cord injury: Preclinical safety and efficacy study. Exp Cell Res 2020; 395:112184. [PMID: 32707134 DOI: 10.1016/j.yexcr.2020.112184] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 07/13/2020] [Accepted: 07/17/2020] [Indexed: 12/22/2022]
Abstract
Functional multipotency renders human umbilical cord mesenchymal stem cell (hUC-MSC) a promising candidate for the treatment of spinal cord injury (SCI). However, its safety and efficacy have not been fully understood for clinical translation. In this study, we performed cellular, kinematic, physiological, and anatomical analyses, either in vitro or in vivo, to comprehensively evaluate the safety and efficacy associated with subarachnoid transplantation of hUC-MSCs in rats with subacute incomplete SCI. Concerning safety, hUC-MSCs were shown to have normal morphology, excellent viability, steady proliferation, typical biomarkers, stable karyotype in vitro, and no tumorigenicity both in vitro and in vivo. Following subarachnoid transplantation of hUC-MSCs in the subject rodents, the biodistribution of hUC-MSCs was restricted to the spinal cord, and no toxicity to immune system or organ function was observed. Body weight, organ weight, and the ratio of the latter upon the former between stem cell-transplanted rats and placebo-injected rats revealed no statistical differences. Regarding efficacy, hUC-MSCs could differentiate into osteoblasts, chondrocytes, adipocytes and neural progenitor cells in vitro. While in vivo studies revealed that subarachnoid transplantation of stem cells resulted in significant improvement in locomotion, earlier automatic micturition recovery and reduced lesion size, which correlated with increased regeneration of tracking fiber and reduced parenchymal inflammation. In vivo luminescence imaging showed that a few of the transplanted luciferase-labeled hUC-MSCs tended to migrate towards the lesion epicenter. Shortened latency and enhanced amplitude were also observed in both motor and sensory evoked potentials, indicating improved signal conduction in the damaged site. Immunofluorescent staining confirmed that a few of the administrated hUC-MSCs integrated into the spinal cord parenchyma and differentiated into astrocytes and oligodendrocytes, but not neurons. Moreover, decreased astrogliosis, increased remyelination, and neuron regeneration could be observed. To the best of our knowledge, this preclinical study provides detailed safety and efficacy evidence regarding intrathecal transplantation of hUC-MSCs in treating SCI for the first time and thus, supports its initiation in the following clinical trial.
Collapse
Affiliation(s)
- Yang Yang
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong Province, People's Republic of China
| | - Ting-Ting Cao
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, 1023 Shatai South Road, Baiyun District, Guangzhou, Guangdong Province, People's Republic of China
| | - Zhen-Ming Tian
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong Province, People's Republic of China
| | - Han Gao
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong Province, People's Republic of China
| | - Hui-Quan Wen
- Department of Radiology, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong Province, People's Republic of China
| | - Mao Pang
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong Province, People's Republic of China
| | - Wei-Jie He
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong Province, People's Republic of China
| | - Nan-Xiang Wang
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong Province, People's Republic of China
| | - Yu-Yong Chen
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong Province, People's Republic of China
| | - Yang Wang
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong Province, People's Republic of China
| | - He Li
- Department of Physiology, School of Basic Medical Sciences, Southern Medical University, 1023 Shatai South Road, Baiyun District, Guangzhou, Guangdong Province, People's Republic of China
| | - Jun-Wei Lin
- Department of Obstetrics, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong Province, People's Republic of China
| | - Zhuang Kang
- Department of Radiology, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong Province, People's Republic of China
| | - Mang-Mang Li
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, 1023 Shatai South Road, Baiyun District, Guangzhou, Guangdong Province, People's Republic of China.
| | - Bin Liu
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong Province, People's Republic of China.
| | - Li-Min Rong
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong Province, People's Republic of China.
| |
Collapse
|
21
|
Hosseinzadeh S, Lindsay SL, Gallagher AG, Wellings DA, Riehle MO, Riddell JS, Barnett SC. A novel poly-ε-lysine based implant, Proliferate®, for promotion of CNS repair following spinal cord injury. Biomater Sci 2020; 8:3611-3627. [PMID: 32515439 DOI: 10.1039/d0bm00097c] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The limited regenerative capacity of the CNS poses formidable challenges to the repair of spinal cord injury (SCI). Two key barriers to repair are (i) the physical gap left by the injury, and (ii) the inhibitory milieu surrounding the injury, the glial scar. Biomaterial implantation into the injury site can fill the cavity, provide a substrate for cell migration, and potentially attenuate the glial scar. We investigated the biological viability of a biocompatible and biodegradable poly-ε-lysine based biomaterial, Proliferate®, in low and high cross-linked forms and when coated with IKVAV peptide, for SCI implantation. We demonstrate altered astrocyte morphology and nestin expression on Proliferate® compared to conventional glass cell coverslips suggesting a less reactive phenotype. Moreover Proliferate® supported myelination in vitro, with myelination observed sooner on IKVAV-coated constructs compared with uncoated Proliferate®, and delayed overall compared with maintenance on glass coverslips. For in vivo implantation, parallel-aligned channels were fabricated into Proliferate® to provide cell guidance cues. Extensive vascularisation and cellular infiltration were observed in constructs implanted in vivo, along with an astrocyte border and microglial response. Axonal ingrowth was observed at the construct border and inside implants in intact channels. We conclude that Proliferate® is a promising biomaterial for implantation following SCI.
Collapse
Affiliation(s)
- Sara Hosseinzadeh
- Institute of Infection, Immunity and Inflammation, Sir Graeme Davies Building, 120 University Place, University of Glasgow, Glasgow G12 8TA, UK.
| | | | | | | | | | | | | |
Collapse
|
22
|
Papa S, Pizzetti F, Perale G, Veglianese P, Rossi F. Regenerative medicine for spinal cord injury: focus on stem cells and biomaterials. Expert Opin Biol Ther 2020; 20:1203-1213. [PMID: 32421405 DOI: 10.1080/14712598.2020.1770725] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Spinal cord injury (SCI) is a dramatic medical pathology consequence of a trauma (primary injury). However, most of the post-traumatic degeneration of the tissue is caused by the so-called secondary injury, which is known to be a multifactorial process. This, indeed, includes a wide spectrum of events: blood-brain barrier dysfunction, local inflammation, neuronal death, demyelination and disconnection of nerve pathways. AREAS COVERED Cell therapy represents a promising cure to target diseases and disorders at the cellular level, by restoring cell population or using cells as carriers of therapeutic cargo. In particular, regenerative medicine with stem cells represents the most appealing category to be used, thanks to their peculiar features. EXPERT OPINION Many preclinical research studies demonstrated that cell treatment can improve animal sensory/motor functions and so demonstrated to be very promising for clinical trials. In particular, recent advances have led to the development of biomaterials aiming to promote in situ cell delivery. This review digs into this topic discussing the possibility of cell treatment to improve medical chances in SCI repair.
Collapse
Affiliation(s)
- Simonetta Papa
- Department of Neuroscience, IRCCS Istituto Di Ricerche Farmacologiche "Mario Negri" , Milan, Italy
| | - Fabio Pizzetti
- Department of Neuroscience, IRCCS Istituto Di Ricerche Farmacologiche "Mario Negri" , Milan, Italy.,Department of Chemistry, Materials and Chemical Engineering "Giulio Natta" , Milan, Italy
| | - Giuseppe Perale
- Faculty of Biomedical Sciences, University of Southern Switzerland (USI) , Lugano, Switzerland.,Ludwig Boltzmann Institute for Experimental and Clinical Traumatology , Vienna, Austria
| | - Pietro Veglianese
- Department of Neuroscience, IRCCS Istituto Di Ricerche Farmacologiche "Mario Negri" , Milan, Italy
| | - Filippo Rossi
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta" , Milan, Italy
| |
Collapse
|
23
|
Invited review: Utilizing peripheral nerve regenerative elements to repair damage in the CNS. J Neurosci Methods 2020; 335:108623. [DOI: 10.1016/j.jneumeth.2020.108623] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 01/31/2020] [Accepted: 02/02/2020] [Indexed: 12/20/2022]
|
24
|
Li X, Zhan J, Hou Y, Hou Y, Chen S, Luo D, Luan J, Wang L, Lin D. Coenzyme Q10 Regulation of Apoptosis and Oxidative Stress in H 2O 2 Induced BMSC Death by Modulating the Nrf-2/NQO-1 Signaling Pathway and Its Application in a Model of Spinal Cord Injury. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:6493081. [PMID: 31915512 PMCID: PMC6930770 DOI: 10.1155/2019/6493081] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 11/16/2019] [Indexed: 12/23/2022]
Abstract
Spinal cord injury (SCI) has always been considered to be a devastating problem that results in catastrophic dysfunction, high disability rate, low mortality rate, and huge cost for the patient. Stem cell-based therapy, especially using bone marrow mesenchymal stem cells (BMSCs), is a promising strategy for the treatment of SCI. However, SCI results in low rates of cell survival and a poor microenvironment, which limits the therapeutic efficiency of BMSC transplantation. Coenzyme Q10 (CoQ10) is known as a powerful antioxidant, which inhibits lipid peroxidation and scavenges free radicals, and its combined effect with BMSC transplantation has been shown to have a powerful impact on protecting the vitality of cells, as well as antioxidant and antiapoptotic compounds in SCI. Therefore, we aimed to evaluate whether CoQ10 could decrease oxidative stress against the apoptosis of BMSCs in vitro and explored its molecular mechanisms. Furthermore, we investigated the protective effect of CoQ10 combined with BMSCs transplanted into a SCI model to verify its ability. Our results demonstrate that CoQ10 treatment significantly decreases the expression of the proapoptotic proteins Bax and Caspase-3, as shown through TUNEL-positive staining and the products of oxidative stress (ROS), while increasing the expression of the antiapoptotic protein Bcl-2 and the products of antioxidation, such as glutathione (GSH), against apoptosis and oxidative stress, in a H2O2-induced model. We also identified consistent results from the CoQ10 treatment of BMSCs transplanted into SCI rats in vivo. Moreover, the Nrf-2 signaling pathway was also investigated in order to detail its molecular mechanism, and the results show that it plays an important role, both in vitro and in vivo. Thus, CoQ10 exerts an antiapoptotic and antioxidant effect, as well as improves the microenvironment in vitro and in vivo. It may also protect BMSCs from oxidative stress and enhance their therapeutic efficiency when transplanted for SCI treatment.
Collapse
Affiliation(s)
- Xing Li
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, No. 111 Dade Road, Guangzhou, Guangdong 510120, China
- Guangzhou University of Chinese Medicine, No. 12, Jichang Road, Baiyun District, Guangzhou 510405, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Jiheng Zhan
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, No. 111 Dade Road, Guangzhou, Guangdong 510120, China
- Guangzhou University of Chinese Medicine, No. 12, Jichang Road, Baiyun District, Guangzhou 510405, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Yu Hou
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, No. 111 Dade Road, Guangzhou, Guangdong 510120, China
- Guangzhou University of Chinese Medicine, No. 12, Jichang Road, Baiyun District, Guangzhou 510405, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Yonghui Hou
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, No. 111 Dade Road, Guangzhou, Guangdong 510120, China
- Guangzhou University of Chinese Medicine, No. 12, Jichang Road, Baiyun District, Guangzhou 510405, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Shudong Chen
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, No. 111 Dade Road, Guangzhou, Guangdong 510120, China
- Guangzhou University of Chinese Medicine, No. 12, Jichang Road, Baiyun District, Guangzhou 510405, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Dan Luo
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, No. 111 Dade Road, Guangzhou, Guangdong 510120, China
- Guangzhou University of Chinese Medicine, No. 12, Jichang Road, Baiyun District, Guangzhou 510405, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Jiyao Luan
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, No. 111 Dade Road, Guangzhou, Guangdong 510120, China
- Guangzhou University of Chinese Medicine, No. 12, Jichang Road, Baiyun District, Guangzhou 510405, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Le Wang
- Department of Spine Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Dingkun Lin
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, No. 111 Dade Road, Guangzhou, Guangdong 510120, China
- Guangzhou University of Chinese Medicine, No. 12, Jichang Road, Baiyun District, Guangzhou 510405, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| |
Collapse
|
25
|
Liu S, Xie YY, Wang B. Role and prospects of regenerative biomaterials in the repair of spinal cord injury. Neural Regen Res 2019; 14:1352-1363. [PMID: 30964053 PMCID: PMC6524500 DOI: 10.4103/1673-5374.253512] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 12/20/2018] [Indexed: 12/12/2022] Open
Abstract
Axonal junction defects and an inhibitory environment after spinal cord injury seriously hinder the regeneration of damaged tissues and neuronal functions. At the site of spinal cord injury, regenerative biomaterials can fill cavities, deliver curative drugs, and provide adsorption sites for transplanted or host cells. Some regenerative biomaterials can also inhibit apoptosis, inflammation and glial scar formation, or further promote neurogenesis, axonal growth and angiogenesis. This review summarized a variety of biomaterial scaffolds made of natural, synthetic, and combined materials applied to spinal cord injury repair. Although these biomaterial scaffolds have shown a certain therapeutic effect in spinal cord injury repair, there are still many problems to be resolved, such as product standards and material safety and effectiveness.
Collapse
Affiliation(s)
- Shuo Liu
- Clinical Stem Cell Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu Province, China
| | - Yuan-Yuan Xie
- Clinical Stem Cell Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu Province, China
| | - Bin Wang
- Clinical Stem Cell Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu Province, China
| |
Collapse
|
26
|
Yu Z, Xu N, Zhang N, Xiong Y, Wang Z, Liang S, Zhao D, Huang F, Zhang C. Repair of Peripheral Nerve Sensory Impairments via the Transplantation of Bone Marrow Neural Tissue-Committed Stem Cell-Derived Sensory Neurons. Cell Mol Neurobiol 2019; 39:341-353. [PMID: 30684112 PMCID: PMC11469867 DOI: 10.1007/s10571-019-00650-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 01/04/2019] [Indexed: 01/20/2023]
Abstract
The present study aimed to investigate the efficacy of transplantation of bone marrow neural tissue-committed stem cell-derived sensory neuron-like cells for the repair of peripheral nerve sensory impairments in rats. Bone marrow was isolated and cultured to obtain the neural tissue-committed stem cells (NTCSCs), and the differentiation of these cells into sensory neuron-like cells was induced. Bone marrow mesenchymal stem cells (BMSCs), bone marrow NTCSCs, and bone marrow NTCSC-derived sensory neurons (NTCSC-SNs) were transplanted by microinjection into the L4 and L5 dorsal root ganglions (DRGs) in an animal model of sensory defect. On the 2nd, 4th, 8th, and 12th week after the transplantation, the effects of the three types of stem cells on the repair of the sensory functional defect were analyzed via behavioral observation, sensory function evaluation, electrophysiological examination of the sciatic nerve, and morphological observation of the DRGs. The results revealed that the transplanted BMSCs, NTCSCs, and NTCSC-SNs were all able to repair the sensory nerves. In addition, the effect of the NTCSC-SNs was significantly better than that of the other two types of stem cells. The general posture and gait of the animals in the sensory defect model exhibited evident improvement over time. Plantar temperature sensitivity and pain sensitivity gradually recovered, and the sensation latency was reduced, with faster sensory nerve conduction velocity. Transplantation of NTCSC-SNs can improve the repair of peripheral nerve sensory defects in rats.
Collapse
Affiliation(s)
- Zhenhai Yu
- Department of Human Anatomy, College of Basic Medical Sciences, Binzhou Medical University, Yantai, 264003, People's Republic of China
- Department of Human Anatomy, College of Basic Medical Sciences, Second Military Medical University, Shanghai, 200433, People's Republic of China
| | - Ning Xu
- Department of Gastroenterology, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, 264100, People's Republic of China
| | - Naili Zhang
- Department of Human Anatomy, College of Basic Medical Sciences, Binzhou Medical University, Yantai, 264003, People's Republic of China
| | - Yanlian Xiong
- Department of Human Anatomy, College of Basic Medical Sciences, Binzhou Medical University, Yantai, 264003, People's Republic of China
| | - Zhiqiang Wang
- Department of Human Anatomy, College of Basic Medical Sciences, Binzhou Medical University, Yantai, 264003, People's Republic of China
| | - Shaohua Liang
- Department of Human Anatomy, College of Basic Medical Sciences, Binzhou Medical University, Yantai, 264003, People's Republic of China
| | - Dongmei Zhao
- Department of Human Anatomy, College of Basic Medical Sciences, Binzhou Medical University, Yantai, 264003, People's Republic of China
| | - Fei Huang
- Department of Human Anatomy, College of Basic Medical Sciences, Binzhou Medical University, Yantai, 264003, People's Republic of China.
| | - Chuansen Zhang
- Department of Human Anatomy, College of Basic Medical Sciences, Binzhou Medical University, Yantai, 264003, People's Republic of China.
- Department of Human Anatomy, College of Basic Medical Sciences, Second Military Medical University, Shanghai, 200433, People's Republic of China.
| |
Collapse
|
27
|
Cohen E, Merzendorfer H. Chitin/Chitosan: Versatile Ecological, Industrial, and Biomedical Applications. EXTRACELLULAR SUGAR-BASED BIOPOLYMERS MATRICES 2019; 12. [PMCID: PMC7115017 DOI: 10.1007/978-3-030-12919-4_14] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Chitin is a linear polysaccharide of N-acetylglucosamine, which is highly abundant in nature and mainly produced by marine crustaceans. Chitosan is obtained by hydrolytic deacetylation. Both polysaccharides are renewable resources, simply and cost-effectively extracted from waste material of fish industry, mainly crab and shrimp shells. Research over the past five decades has revealed that chitosan, in particular, possesses unique and useful characteristics such as chemical versatility, polyelectrolyte properties, gel- and film-forming ability, high adsorption capacity, antimicrobial and antioxidative properties, low toxicity, and biocompatibility and biodegradability features. A plethora of chemical chitosan derivatives have been synthesized yielding improved materials with suggested or effective applications in water treatment, biosensor engineering, agriculture, food processing and storage, textile additives, cosmetics fabrication, and in veterinary and human medicine. The number of studies in this research field has exploded particularly during the last two decades. Here, we review recent advances in utilizing chitosan and chitosan derivatives in different technical, agricultural, and biomedical fields.
Collapse
Affiliation(s)
- Ephraim Cohen
- Department of Entomology, The Robert H. Smith Faculty of Agriculture Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Hans Merzendorfer
- School of Science and Technology, Institute of Biology – Molecular Biology, University of Siegen, Siegen, Germany
| |
Collapse
|
28
|
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: 58] [Impact Index Per Article: 9.7] [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
|
29
|
Ruzicka J, Romanyuk N, Jirakova K, Hejcl A, Janouskova O, Machova LU, Bochin M, Pradny M, Vargova L, Jendelova P. The Effect of iPS-Derived Neural Progenitors Seeded on Laminin-Coated pHEMA-MOETACl Hydrogel with Dual Porosity in a Rat Model of Chronic Spinal Cord Injury. Cell Transplant 2019; 28:400-412. [PMID: 30654639 PMCID: PMC6628561 DOI: 10.1177/0963689718823705] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Spinal cord injury (SCI), is a devastating condition leading to the loss of locomotor and sensory function below the injured segment. Despite some progress in acute SCI treatment using stem cells and biomaterials, chronic SCI remains to be addressed. We have assessed the use of laminin-coated hydrogel with dual porosity, seeded with induced pluripotent stem cell-derived neural progenitors (iPSC-NPs), in a rat model of chronic SCI. iPSC-NPs cultured for 3 weeks in hydrogel in vitro were positive for nestin, glial fibrillary acidic protein (GFAP) and microtubule-associated protein 2 (MAP2). These cell-polymer constructs were implanted into a balloon compression lesion, 5 weeks after lesion induction. Animals were behaviorally tested, and spinal cord tissue was immunohistochemically analyzed 28 weeks after SCI. The implanted iPSC-NPs survived in the scaffold for the entire experimental period. Host axons, astrocytes and blood vessels grew into the implant and an increased sprouting of host TH+ fibers was observed in the lesion vicinity. The implantation of iPSC-NP-LHM cell-polymer construct into the chronic SCI led to the integration of material into the injured spinal cord, reduced cavitation and supported the iPSC-NPs survival, but did not result in a statistically significant improvement of locomotor recovery.
Collapse
Affiliation(s)
- Jiri Ruzicka
- 1 Department of Tissue Culture and Stem Cells, Institute of Experimental Medicine, CAS, Prague, Czech Republic
| | - Nataliya Romanyuk
- 1 Department of Tissue Culture and Stem Cells, Institute of Experimental Medicine, CAS, Prague, Czech Republic
| | - Klara Jirakova
- 1 Department of Tissue Culture and Stem Cells, Institute of Experimental Medicine, CAS, Prague, Czech Republic
| | - Ales Hejcl
- 1 Department of Tissue Culture and Stem Cells, Institute of Experimental Medicine, CAS, Prague, Czech Republic
| | - Olga Janouskova
- 2 Department of Polymer Networks and Gels, Institute of Macromolecular Chemistry, CAS, Prague, Czech Republic
| | - Lucia Urdzikova Machova
- 1 Department of Tissue Culture and Stem Cells, Institute of Experimental Medicine, CAS, Prague, Czech Republic
| | - Marcel Bochin
- 1 Department of Tissue Culture and Stem Cells, Institute of Experimental Medicine, CAS, Prague, Czech Republic.,3 Department of Neurosciences, 2nd Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Martin Pradny
- 2 Department of Polymer Networks and Gels, Institute of Macromolecular Chemistry, CAS, Prague, Czech Republic
| | - Lydia Vargova
- 1 Department of Tissue Culture and Stem Cells, Institute of Experimental Medicine, CAS, Prague, Czech Republic.,3 Department of Neurosciences, 2nd Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Pavla Jendelova
- 1 Department of Tissue Culture and Stem Cells, Institute of Experimental Medicine, CAS, Prague, Czech Republic.,3 Department of Neurosciences, 2nd Faculty of Medicine, Charles University, Prague, Czech Republic
| |
Collapse
|
30
|
Ko CC, Tu TH, Wu JC, Huang WC, Cheng H. Acidic Fibroblast Growth Factor in Spinal Cord Injury. Neurospine 2019; 16:728-738. [PMID: 30653905 PMCID: PMC6944993 DOI: 10.14245/ns.1836216.108] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Accepted: 12/26/2018] [Indexed: 12/30/2022] Open
Abstract
Spinal cord injury (SCI), with an incidence rate of 246 per million person-years among adults in Taiwan, remains a devastating disease in the modern day. Elderly men with lower socioeconomic status have an even higher risk for SCI. Despite advances made in medicine and technology to date, there are few effective treatments for SCI due to limitations in the regenerative capacity of the adult central nervous system. Experiments and clinical trials have explored neuro-regeneration in human SCI, encompassing cell- and molecule-based therapies. Furthermore, strategies have aimed at restoring connections, including autologous peripheral nerve grafts and biomaterial scaffolds that theoretically promote axonal growth. Most molecule-based therapies target the modulation of inhibitory molecules to promote axonal growth, degrade glial scarring obstacles, and stimulate intrinsic regenerative capacity. Among them, acidic fibroblast growth factor (aFGF) has been investigated for nerve repair; it is mitogenic and pluripotent in nature and could enhance axonal growth and mitigate glial scarring. For more than 2 decades, the authors have conducted multiple trials, including human and animal experiments, using aFGF to repair nerve injuries, including central and peripheral nerves. In these trials, aFGF has shown promise for neural regeneration, and in the future, more trials and applications should investigate aFGF as a neurotrophic factor. Focusing on aFGF, the current review aimed to summarize the historical evolution of the utilization of aFGF in SCI and nerve injuries, to present applications and trials, to summarize briefly its possible mechanisms, and to provide future perspectives.
Collapse
Affiliation(s)
- Chin-Chu Ko
- Jhong Jheng Spine & Orthopedic Hospital, Kaohsiung, Taiwan.,Institute of Pharmacology, National Yang-Ming University, Taipei, Taiwan.,School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Tsung-Hsi Tu
- School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan.,Taiwan International Graduate Program in Molecular Medicine, National Yang-Ming University and Academia Sinica, Taipei, Taiwan
| | - Jau-Ching Wu
- Institute of Pharmacology, National Yang-Ming University, Taipei, Taiwan.,School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Wen-Cheng Huang
- School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Henrich Cheng
- Institute of Pharmacology, National Yang-Ming University, Taipei, Taiwan.,School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan
| |
Collapse
|
31
|
Ashammakhi N, Ahadian S, Darabi MA, El Tahchi M, Lee J, Suthiwanich K, Sheikhi A, Dokmeci MR, Oklu R, Khademhosseini A. Minimally Invasive and Regenerative Therapeutics. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1804041. [PMID: 30565732 PMCID: PMC6709364 DOI: 10.1002/adma.201804041] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 08/20/2018] [Indexed: 05/03/2023]
Abstract
Advances in biomaterial synthesis and fabrication, stem cell biology, bioimaging, microsurgery procedures, and microscale technologies have made minimally invasive therapeutics a viable tool in regenerative medicine. Therapeutics, herein defined as cells, biomaterials, biomolecules, and their combinations, can be delivered in a minimally invasive way to regenerate different tissues in the body, such as bone, cartilage, pancreas, cardiac, skeletal muscle, liver, skin, and neural tissues. Sophisticated methods of tracking, sensing, and stimulation of therapeutics in vivo using nano-biomaterials and soft bioelectronic devices provide great opportunities to further develop minimally invasive and regenerative therapeutics (MIRET). In general, minimally invasive delivery methods offer high yield with low risk of complications and reduced costs compared to conventional delivery methods. Here, minimally invasive approaches for delivering regenerative therapeutics into the body are reviewed. The use of MIRET to treat different tissues and organs is described. Although some clinical trials have been performed using MIRET, it is hoped that such therapeutics find wider applications to treat patients. Finally, some future perspective and challenges for this emerging field are highlighted.
Collapse
Affiliation(s)
- Nureddin Ashammakhi
- Center for Minimally Invasive Therapeutics (C-MIT), University of California - Los Angeles, Los Angeles, California, USA
- California NanoSystems Institute (CNSI), University of California - Los Angeles, Los Angeles, California, USA
- Department of Bioengineering, University of California - Los Angeles, Los Angeles, California, USA
- Division of Plastic Surgery, Department of Surgery, Oulu University, Oulu, Finland
| | - Samad Ahadian
- Center for Minimally Invasive Therapeutics (C-MIT), University of California - Los Angeles, Los Angeles, California, USA
- California NanoSystems Institute (CNSI), University of California - Los Angeles, Los Angeles, California, USA
- Department of Bioengineering, University of California - Los Angeles, Los Angeles, California, USA
| | - Mohammad Ali Darabi
- Center for Minimally Invasive Therapeutics (C-MIT), University of California - Los Angeles, Los Angeles, California, USA
- California NanoSystems Institute (CNSI), University of California - Los Angeles, Los Angeles, California, USA
- Department of Bioengineering, University of California - Los Angeles, Los Angeles, California, USA
| | - Mario El Tahchi
- Center for Minimally Invasive Therapeutics (C-MIT), University of California - Los Angeles, Los Angeles, California, USA
- California NanoSystems Institute (CNSI), University of California - Los Angeles, Los Angeles, California, USA
- Department of Bioengineering, University of California - Los Angeles, Los Angeles, California, USA
- LBMI, Department of Physics, Lebanese University - Faculty of Sciences 2, PO Box 90656, Jdeidet, Lebanon
| | - Junmin Lee
- Center for Minimally Invasive Therapeutics (C-MIT), University of California - Los Angeles, Los Angeles, California, USA
- California NanoSystems Institute (CNSI), University of California - Los Angeles, Los Angeles, California, USA
- Department of Bioengineering, University of California - Los Angeles, Los Angeles, California, USA
| | - Kasinan Suthiwanich
- Center for Minimally Invasive Therapeutics (C-MIT), University of California - Los Angeles, Los Angeles, California, USA
- California NanoSystems Institute (CNSI), University of California - Los Angeles, Los Angeles, California, USA
- Department of Bioengineering, University of California - Los Angeles, Los Angeles, California, USA
- Department of Materials Science and Engineering, School of Materials and Chemical Technology, Tokyo Institute of Technology, Tokyo, Japan
| | - Amir Sheikhi
- Center for Minimally Invasive Therapeutics (C-MIT), University of California - Los Angeles, Los Angeles, California, USA
- California NanoSystems Institute (CNSI), University of California - Los Angeles, Los Angeles, California, USA
- Department of Bioengineering, University of California - Los Angeles, Los Angeles, California, USA
| | - Mehmet R. Dokmeci
- Center for Minimally Invasive Therapeutics (C-MIT), University of California - Los Angeles, Los Angeles, California, USA
- California NanoSystems Institute (CNSI), University of California - Los Angeles, Los Angeles, California, USA
- Department of Bioengineering, University of California - Los Angeles, Los Angeles, California, USA
| | - Rahmi Oklu
- Division of Interventional Radiology, Department of Radiology, Mayo Clinic, Scottsdale, USA
| | - Ali Khademhosseini
- Center for Minimally Invasive Therapeutics (C-MIT), University of California - Los Angeles, Los Angeles, California, USA
- California NanoSystems Institute (CNSI), University of California - Los Angeles, Los Angeles, California, USA
- Department of Bioengineering, University of California - Los Angeles, Los Angeles, California, USA
- Department of Radiological Sciences, University of California - Los Angeles, Los Angeles, California, USA
- Department of Chemical and Biomolecular Engineering, University of California - Los Angeles, Los Angeles, California, USA
- Center of Nanotechnology, Department of Physics, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Bioindustrial Technologies, College of Animal Bioscience and Technology, Konkuk University, Seoul, Republic of Korea
| |
Collapse
|
32
|
Mohrman AE, Farrag M, Grimm RK, Leipzig ND. Evaluation of in situ gelling chitosan-PEG copolymer for use in the spinal cord. J Biomater Appl 2018; 33:435-446. [PMID: 30111249 DOI: 10.1177/0885328218792824] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The goal of the present work was to characterize a hydrogel material for localized spinal cord delivery. To address spinal cord injuries, an injectable in situ gelling system was tested utilizing a simple, effective, and rapid cross-linking method via Michael addition. Thiolated chitosan material and maleimide-terminated polyethylene glycol material were mixed to form a hydrogel and evaluated in vitro and in vivo. Three distinct thiolated chitosan precursors were made by varying reaction conditions; a modification of chitosan with Traut's reagent (2-iminothiolane) displayed the most attractive hydrogel properties once mixed with polyethylene glycol. The final hydrogel chosen for animal testing had a swelling ratio (Q) of 57.5 ± 3.4 and elastic modulus of 378 ± 72 Pa. After confirming low cellular toxicity in vitro, the hydrogel was injected into the spinal cord of rats for 1 and 2 weeks to assess host reaction. The rats displayed no overt functional deficits due to injection following initial surgical recovery and throughout the 2-week period after for both the saline-injected sham group and hydrogel-injected group. The saline and hydrogel-injected animals both showed a similar response from ED1+ microglia and GFAP overexpression. No significant differences were found between saline-injected and hydrogel-injected groups for any of the measures studied, but there was a trend toward decreased affected area size from 1 to 2 weeks in both groups. Access to the central nervous system is limited by the blood-brain barrier for noninvasive therapies; further development of the current system for localized drug or cellular delivery has the potential to shape treatments of spinal cord injury.
Collapse
Affiliation(s)
- Ashley E Mohrman
- Department of Chemical and Biomolecular Engineering, University of Akron, Akron, OH, USA
| | - Mahmoud Farrag
- Department of Chemical and Biomolecular Engineering, University of Akron, Akron, OH, USA
| | - Rachel K Grimm
- Department of Chemical and Biomolecular Engineering, University of Akron, Akron, OH, USA
| | - Nic D Leipzig
- Department of Chemical and Biomolecular Engineering, University of Akron, Akron, OH, USA
| |
Collapse
|
33
|
Chen X, Zhao Y, Li X, Xiao Z, Yao Y, Chu Y, Farkas B, Romano I, Brandi F, Dai J. Functional Multichannel Poly(Propylene Fumarate)-Collagen Scaffold with Collagen-Binding Neurotrophic Factor 3 Promotes Neural Regeneration After Transected Spinal Cord Injury. Adv Healthc Mater 2018; 7:e1800315. [PMID: 29920990 DOI: 10.1002/adhm.201800315] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2018] [Revised: 05/11/2018] [Indexed: 01/12/2023]
Abstract
Many factors contribute to the poor axonal regrowth and ineffective functional recovery after spinal cord injury (SCI). Biomaterials have been used for SCI repair by promoting bridge formation and reconnecting the neural tissue at the lesion site. The mechanical properties of biomaterials are critical for successful design to ensure the stable support as soon as possible when compressed by the surrounding spine and musculature. Poly(propylene fumarate) (PPF) scaffolds with high mechanical strength have been shown to provide firm spatial maintenance and to promote repair of tissue defects. A multichannel PPF scaffold is combined with collagen biomaterial to build a novel biocompatible delivery system coated with neurotrophin-3 containing an engineered collagen-binding domain (CBD-NT3). The parallel-aligned multichannel structure of PPF scaffolds guide the direction of neural tissue regeneration across the lesion site and promote reestablishment of bridge connectivity. The combinatorial treatment consisting of PPF and collagen loaded with CBD-NT3 improves the inhibitory microenvironment, facilitates axonal and neuronal regeneration, survival of various types of functional neurons and remyelination and synapse formation of regenerated axons following SCI. This novel treatment strategy for SCI repair effectively promotes neural tissue regeneration after transected spinal injury by providing a regrowth-supportive microenvironment and eventually induces functional improvement.
Collapse
Affiliation(s)
- Xi Chen
- Institute of Combined Injury; State Key Laboratory of Trauma; Burns and Combined Injury; Chongqing Engineering Research Center for Nanomedicine; Chongqing Engineering Research Center for Biomaterials and Regenerative Medicine; College of Preventive Medicine; Army Medical University (Third Military Medical University); 30th Gaotanyan street Chongqing 400038 China
| | - Yannan Zhao
- State Key Laboratory of Molecular; Developmental Biology; Institute of Genetics and Developmental Biology; Chinese Academy of Sciences; Beijing 100101 China
| | - Xing Li
- State Key Laboratory of Molecular; Developmental Biology; Institute of Genetics and Developmental Biology; Chinese Academy of Sciences; Beijing 100101 China
| | - Zhifeng Xiao
- State Key Laboratory of Molecular; Developmental Biology; Institute of Genetics and Developmental Biology; Chinese Academy of Sciences; Beijing 100101 China
| | - Yuanjiang Yao
- Department of Neurobiology; Chongqing Key Laboratory of Neurobiology; Army Medical University (Third Military Medical University); 30th Gaotanyan street Chongqing 400038 China
| | - Yun Chu
- Division of Nanobiomedicine; Suzhou Institute of Nano-Tech and Nano-Bionics; Chinese Academy of Sciences; Suzhou 215123 China
| | - Balázs Farkas
- Istituto Italiano di Tecnologia; Via Morego 30 Genova 16163 Italy
| | - Ilaria Romano
- Istituto Italiano di Tecnologia; Via Morego 30 Genova 16163 Italy
| | - Fernando Brandi
- Istituto Italiano di Tecnologia; Via Morego 30 Genova 16163 Italy
- Istituto Nazionale di Ottica; Consiglio Nazionale delle Ricerche; Via Moruzzi 1 Pisa 56124 Italy
| | - Jianwu Dai
- Institute of Combined Injury; State Key Laboratory of Trauma; Burns and Combined Injury; Chongqing Engineering Research Center for Nanomedicine; Chongqing Engineering Research Center for Biomaterials and Regenerative Medicine; College of Preventive Medicine; Army Medical University (Third Military Medical University); 30th Gaotanyan street Chongqing 400038 China
- State Key Laboratory of Molecular; Developmental Biology; Institute of Genetics and Developmental Biology; Chinese Academy of Sciences; Beijing 100101 China
- Department of Neurobiology; Chongqing Key Laboratory of Neurobiology; Army Medical University (Third Military Medical University); 30th Gaotanyan street Chongqing 400038 China
| |
Collapse
|
34
|
Hejčl A, Růžička J, Proks V, Macková H, Kubinová Š, Tukmachev D, Cihlář J, Horák D, Jendelová P. Dynamics of tissue ingrowth in SIKVAV-modified highly superporous PHEMA scaffolds with oriented pores after bridging a spinal cord transection. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2018; 29:89. [PMID: 29938301 DOI: 10.1007/s10856-018-6100-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 06/05/2018] [Indexed: 06/08/2023]
Abstract
While many types of biomaterials have been evaluated in experimental spinal cord injury (SCI) research, little is known about the time-related dynamics of the tissue infiltration of these scaffolds. We analyzed the ingrowth of connective tissue, axons and blood vessels inside the superporous poly (2-hydroxyethyl methacrylate) hydrogel with oriented pores. The hydrogels, either plain or seeded with mesenchymal stem cells (MSCs), were implanted in spinal cord transection at the level of Th8. The animals were sacrificed at days 2, 7, 14, 28, 49 and 6 months after SCI and histologically evaluated. We found that within the first week, the hydrogels were already infiltrated with connective tissue and blood vessels, which remained stable for the next 6 weeks. Axons slowly and gradually infiltrated the hydrogel within the first month, after which the numbers became stable. Six months after SCI we observed rare axons crossing the hydrogel bridge and infiltrating the caudal stump. There was no difference in the tissue infiltration between the plain hydrogels and those seeded with MSCs. We conclude that while connective tissue and blood vessels quickly infiltrate the scaffold within the first week, axons show a rather gradual infiltration over the first month, and this is not facilitated by the presence of MSCs inside the hydrogel pores. Further research which is focused on the permissive micro-environment of the hydrogel scaffold is needed, to promote continuous and long-lasting tissue regeneration across the spinal cord lesion.
Collapse
Affiliation(s)
- Aleš Hejčl
- Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Vídeňská 1083, 142 20, Prague, Czech Republic.
- Department of Neurosurgery, J. E. Purkinje University, Masaryk Hospital, Sociální péče 12A, 401 13, Ústí nad Labem, Czech Republic.
| | - Jiří Růžička
- Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Vídeňská 1083, 142 20, Prague, Czech Republic
- Department of Neuroscience, 2nd Faculty of Medicine, Charles University, V Úvalu 84, 150 06, Prague 5, Czech Republic
| | - Vladimír Proks
- Institute of Macromolecular Chemistry, Academy of Sciences of the Czech Republic, Heyrovského nám.2, 162 06, Praha 6, Břevnov, Czech Republic
| | - Hana Macková
- Institute of Macromolecular Chemistry, Academy of Sciences of the Czech Republic, Heyrovského nám.2, 162 06, Praha 6, Břevnov, Czech Republic
| | - Šárka Kubinová
- Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Vídeňská 1083, 142 20, Prague, Czech Republic
| | - Dmitry Tukmachev
- Department of Neurosurgery, Motol University Hospital, V Úvalu 84, Prague 5, 150 06, Czech Republic
| | - Jiří Cihlář
- Department of Mathematics, Faculty of Science, J. E. Purkyně University, České mládeže 8, 400 96, Ústí nad Labem, Czech Republic
| | - Daniel Horák
- Institute of Macromolecular Chemistry, Academy of Sciences of the Czech Republic, Heyrovského nám.2, 162 06, Praha 6, Břevnov, Czech Republic
| | - Pavla Jendelová
- Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Vídeňská 1083, 142 20, Prague, Czech Republic
- Department of Neuroscience, 2nd Faculty of Medicine, Charles University, V Úvalu 84, 150 06, Prague 5, Czech Republic
| |
Collapse
|
35
|
Dalamagkas K, Tsintou M, Seifalian A, Seifalian AM. Translational Regenerative Therapies for Chronic Spinal Cord Injury. Int J Mol Sci 2018; 19:1776. [PMID: 29914060 PMCID: PMC6032191 DOI: 10.3390/ijms19061776] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 06/05/2018] [Accepted: 06/06/2018] [Indexed: 12/22/2022] Open
Abstract
Spinal cord injury is a chronic and debilitating neurological condition that is currently being managed symptomatically with no real therapeutic strategies available. Even though there is no consensus on the best time to start interventions, the chronic phase is definitely the most stable target in order to determine whether a therapy can effectively restore neurological function. The advancements of nanoscience and stem cell technology, combined with the powerful, novel neuroimaging modalities that have arisen can now accelerate the path of promising novel therapeutic strategies from bench to bedside. Several types of stem cells have reached up to clinical trials phase II, including adult neural stem cells, human spinal cord stem cells, olfactory ensheathing cells, autologous Schwann cells, umbilical cord blood-derived mononuclear cells, adult mesenchymal cells, and autologous bone-marrow-derived stem cells. There also have been combinations of different molecular therapies; these have been either alone or combined with supportive scaffolds with nanostructures to facilitate favorable cell⁻material interactions. The results already show promise but it will take some coordinated actions in order to develop a proper step-by-step approach to solve impactful problems with neural repair.
Collapse
Affiliation(s)
- Kyriakos Dalamagkas
- The Institute for Rehabilitation and Research, Memorial Hermann Texas Medical Centre, Houston, TX 77030, USA.
- Centre for Nanotechnology & Regenerative Medicine, Division of Surgery and Interventional Science, University College of London (UCL), London NW3 2QG, UK.
| | - Magdalini Tsintou
- Centre for Nanotechnology & Regenerative Medicine, Division of Surgery and Interventional Science, University College of London (UCL), London NW3 2QG, UK.
- Center for Neural Systems Investigations, Massachusetts General Hospital/HST Athinoula A., Martinos Centre for Biomedical Imaging, Harvard Medical School, Boston, MA 02129, USA.
| | - Amelia Seifalian
- Faculty of Medical Sciences, UCL Medical School, London WC1E 6BT, UK.
| | - Alexander M Seifalian
- NanoRegMed Ltd. (Nanotechnology & Regenerative Medicine Commercialization Centre), The London BioScience Innovation Centre, London NW1 0NH, UK.
| |
Collapse
|
36
|
Liu S, Schackel T, Weidner N, Puttagunta R. Biomaterial-Supported Cell Transplantation Treatments for Spinal Cord Injury: Challenges and Perspectives. Front Cell Neurosci 2018; 11:430. [PMID: 29375316 PMCID: PMC5768640 DOI: 10.3389/fncel.2017.00430] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 12/20/2017] [Indexed: 12/17/2022] Open
Abstract
Spinal cord injury (SCI), resulting in para- and tetraplegia caused by the partial or complete disruption of descending motor and ascending sensory neurons, represents a complex neurological condition that remains incurable. Following SCI, numerous obstacles comprising of the loss of neural tissue (neurons, astrocytes, and oligodendrocytes), formation of a cavity, inflammation, loss of neuronal circuitry and function must be overcome. Given the multifaceted primary and secondary injury events that occur with SCI treatment options are likely to require combinatorial therapies. While several methods have been explored, only the intersection of two, cell transplantation and biomaterial implantation, will be addressed in detail here. Owing to the constant advance of cell culture technologies, cell-based transplantation has come to the forefront of SCI treatment in order to replace/protect damaged tissue and provide physical as well as trophic support for axonal regrowth. Biomaterial scaffolds provide cells with a protected environment from the surrounding lesion, in addition to bridging extensive damage and providing physical and directional support for axonal regrowth. Moreover, in this combinatorial approach cell transplantation improves scaffold integration and therefore regenerative growth potential. Here, we review the advances in combinatorial therapies of Schwann cells (SCs), astrocytes, olfactory ensheathing cells (OECs), mesenchymal stem cells, as well as neural stem and progenitor cells (NSPCs) with various biomaterial scaffolds.
Collapse
Affiliation(s)
- Shengwen Liu
- Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Thomas Schackel
- Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| | - Norbert Weidner
- Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| | - Radhika Puttagunta
- Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
37
|
Chhabra HS, Sarda K. Clinical translation of stem cell based interventions for spinal cord injury - Are we there yet? Adv Drug Deliv Rev 2017; 120:41-49. [PMID: 28964881 DOI: 10.1016/j.addr.2017.09.021] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 09/14/2017] [Accepted: 09/20/2017] [Indexed: 01/01/2023]
Abstract
Recent advances in basic science in research related to spinal cord injury (SCI) and regeneration have led to a variety of novel experimental therapeutics designed to promote functionally effective axonal regrowth and sprouting. Stem cell and other cellular interventions have gained lot of attention due to their immense potential of regeneration. These interventions have been tested for their efficacy in case of SCI both at the pre-clinical and clinical level. In this review we critically discuss the published literature on the cellular interventions for SCI and their clinical applications with respect to the strength of evidence established by these studies. The need to curb unethical practice of offering unproven stem cell "therapies" for SCI at a global level is also discussed.
Collapse
|
38
|
Vismara I, Papa S, Rossi F, Forloni G, Veglianese P. Current Options for Cell Therapy in Spinal Cord Injury. Trends Mol Med 2017; 23:831-849. [PMID: 28811172 DOI: 10.1016/j.molmed.2017.07.005] [Citation(s) in RCA: 144] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2017] [Revised: 07/13/2017] [Accepted: 07/16/2017] [Indexed: 12/12/2022]
Abstract
Spinal cord injury (SCI) is a complex pathology that evolves after primary acute mechanical injury, causing further damage to the spinal cord tissue that exacerbates clinical outcomes. Based on encouraging results from preclinical experiments, some cell treatments being translated into clinical practice demonstrate promising and effective improvement in sensory/motor function. Combinatorial treatments of cell and drug/biological factors have been demonstrated to be more effective than cell treatments alone. Recent advances have led to the development of biomaterials aiming to promote in situ cell delivery for SCI, together with combinatorial strategies using drugs/biomolecules to achieve a maximized multitarget approach. This review provides an overview of single and combinatorial regenerative cell treatments as well as potential delivery options to treat SCI.
Collapse
Affiliation(s)
- Irma Vismara
- Dipartimento di Neuroscienze, Istituto Di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto di Ricerche Farmacologiche Mario Negri, via La Masa 19, 20156 Milano, Italy; These authors contributed equally to this work
| | - Simonetta Papa
- Dipartimento di Neuroscienze, Istituto Di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto di Ricerche Farmacologiche Mario Negri, via La Masa 19, 20156 Milano, Italy; These authors contributed equally to this work
| | - Filippo Rossi
- Dipartimento di Chimica, Materiali e Ingegneria Chimica 'Giulio Natta', Politecnico di Milano, via Mancinelli 7, 20131 Milano, Italy
| | - Gianluigi Forloni
- Dipartimento di Neuroscienze, Istituto Di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto di Ricerche Farmacologiche Mario Negri, via La Masa 19, 20156 Milano, Italy
| | - Pietro Veglianese
- Dipartimento di Neuroscienze, Istituto Di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto di Ricerche Farmacologiche Mario Negri, via La Masa 19, 20156 Milano, Italy.
| |
Collapse
|
39
|
Xiong LL, Liu F, Lu BT, Zhao WL, Dong XJ, Liu J, Zhang RP, Zhang P, Wang TH. Bone Marrow Mesenchymal Stem-Cell Transplantation Promotes Functional Improvement Associated with CNTF-STAT3 Activation after Hemi-Sectioned Spinal Cord Injury in Tree Shrews. Front Cell Neurosci 2017; 11:172. [PMID: 28701922 PMCID: PMC5487382 DOI: 10.3389/fncel.2017.00172] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 06/07/2017] [Indexed: 02/05/2023] Open
Abstract
Hemi-sectioned spinal cord injury (hSCI) can lead to spastic paralysis on the injured side, as well as flaccid paralysis on the contralateral side, which can negatively affect a patient’s daily life. Stem-cell therapy may offer an effective treatment option for individuals with hSCI. To examine the role of bone marrow mesenchymal stem cells (BMSCs) transplantation on hSCI and explore related mechanisms in the tree shrews, here, we created a model of hSCI by inducing injury at the tenth thoracic vertebra (T10). Hoechst 33342-labeled BMSCs derived from adult tree shrews were isolated, cultured, and implanted into the spinal cord around the injury site at 9 days after injury. The isolated BMSCs were able to survive, proliferate and release a variety of neurotrophic factors (NTFs) both in vitro and in vivo. At 28 days after injury, compared with the sham group, the hSCI group displayed scar formation and dramatic elevations in the mean interleukin 1 beta (IL-1β) density and cell apoptosis level, whereas the expression of signal transducer and activator of transcription 3 (STAT3) and ciliary neurotrophic factor (CNTF) mRNA was reduced. Following BMSC transplantation, motoneurons extent of shrinkage were reduced and the animals’ Basso, Beattie, and Bresnahan (BBB) locomotion scale scores were significantly higher at 21 and 28 days after injury when compared with the injured group. Moreover, the hSCI-induced elevations in scar formation, IL-1β, and cell apoptosis were reduced by BMSC transplantation to levels that were close to those of the sham group. Corresponding elevations in the expression of STAT3 and CNTF mRNA were observed in the hSCI + BMSCs group, and the levels were not significantly different from those observed in the sham group. Together, our results support that grafted BMSCs can significantly improve locomotor function in tree shrews subjected to hSCI and that this improvement is associated with the upregulation of CNTF and STAT3 signaling.
Collapse
Affiliation(s)
- Liu-Lin Xiong
- Institute of Neurological Disease and Department of Anesthesiology, Translational Neuroscience Center, West China Hospital, Sichuan UniversityChengdu, China.,Institute of Neuroscience, Animal Zoology Department, Kunming Medical UniversityKunming, China
| | - Fei Liu
- Institute of Neurological Disease and Department of Anesthesiology, Translational Neuroscience Center, West China Hospital, Sichuan UniversityChengdu, China
| | - Bing-Tuan Lu
- Institute of Neuroscience, Animal Zoology Department, Kunming Medical UniversityKunming, China
| | - Wen-Ling Zhao
- Institute of Neurological Disease and Department of Anesthesiology, Translational Neuroscience Center, West China Hospital, Sichuan UniversityChengdu, China
| | - Xiu-Juan Dong
- Key Laboratory of National Physical Fitness and Altitude Training Adaptation in Yunnan Province, Institute of Physical Education, Yunnan Normal UniversityKunming, China
| | - Jia Liu
- Institute of Neuroscience, Animal Zoology Department, Kunming Medical UniversityKunming, China
| | - Rong-Ping Zhang
- Biomedical Engineering Research Center, Kunming Medical UniversityKunming, China
| | - Piao Zhang
- Institute of Neuroscience, Animal Zoology Department, Kunming Medical UniversityKunming, China
| | - Ting-Hua Wang
- Institute of Neurological Disease and Department of Anesthesiology, Translational Neuroscience Center, West China Hospital, Sichuan UniversityChengdu, China.,Institute of Neuroscience, Animal Zoology Department, Kunming Medical UniversityKunming, China
| |
Collapse
|
40
|
Li G, Xiao Q, McNaughton R, Han L, Zhang L, Wang Y, Yang Y. Nanoengineered porous chitosan/CaTiO 3 hybrid scaffolds for accelerating Schwann cells growth in peripheral nerve regeneration. Colloids Surf B Biointerfaces 2017; 158:57-67. [PMID: 28672204 DOI: 10.1016/j.colsurfb.2017.06.026] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Revised: 06/19/2017] [Accepted: 06/20/2017] [Indexed: 01/11/2023]
Abstract
To further improve the property of promoting peripheral nerve regeneration of chitosan materials, CaTiO3 nanoparticles with various concentrations were synthesized in chitosan (CS) solution and formed to porous CS/CaTiO3 hybrid scaffolds. The properties including morphology, wettability, porosity, crystallization intensity and surface charges were characterized, respectively. The influence of the porous CS/CaTiO3 hybrid scaffolds on Schwann cells growth was evaluated. The results showed that the CaTiO3 hybridized CS scaffolds possessed homogeneous nanoparticles distribution with concentration-dependent effect. The hybridization of CaTiO3 nanoparticles could increase the hydrophobicity while reduce the porosity and surface charge density of the porous CS/CaTiO3 hybrid scaffolds The crystal structure of the hybridized scaffolds was mainly the orthorhombic structure of the calcium titanate accompanied by the amorphous phase of chitosan. Culture of Schwann cells indicated that the CS/CaTiO3 hybrid scaffolds with a suitable concentration of CaTiO3 nanoparticles could obviously promote the attachment, proliferation and biological function maintenance of Schwann cells, thus showing potentially great significance towards application in peripheral nerve regeneration.
Collapse
Affiliation(s)
- Guicai Li
- Key laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Nantong University, 226001, Nantong, PR China; The Neural Regeneration Co-innovation Center of Jiangsu Province, 226001 Nantong, PR China; Department of Mechanical Engineering, Boston University, Boston, MA 02215, USA.
| | - Qinzhi Xiao
- Department of Pediatrics, Affiliated Hospital of Nantong University, 226001, Nantong, PR China
| | - Ryan McNaughton
- Department of Mechanical Engineering, Boston University, Boston, MA 02215, USA
| | - Lei Han
- Key Laboratory of MEMS of the Ministry of Education, Southeast University, Nanjing 210096, PR China
| | - Luzhong Zhang
- Key laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Nantong University, 226001, Nantong, PR China; The Neural Regeneration Co-innovation Center of Jiangsu Province, 226001 Nantong, PR China
| | - Yaling Wang
- School of Chemical Engineering, Nantong University, 226001, Nantong, PR China
| | - Yumin Yang
- Key laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Nantong University, 226001, Nantong, PR China; The Neural Regeneration Co-innovation Center of Jiangsu Province, 226001 Nantong, PR China.
| |
Collapse
|
41
|
A combination of GDNF and hUCMSC transplantation loaded on SF/AGs composite scaffolds for spinal cord injury repair. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2017; 74:230-237. [DOI: 10.1016/j.msec.2016.12.017] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 10/26/2016] [Accepted: 12/04/2016] [Indexed: 12/14/2022]
|
42
|
Palejwala AH, Fridley JS, Mata JA, Samuel ELG, Luerssen TG, Perlaky L, Kent TA, Tour JM, Jea A. Biocompatibility of reduced graphene oxide nanoscaffolds following acute spinal cord injury in rats. Surg Neurol Int 2016; 7:75. [PMID: 27625885 PMCID: PMC5009578 DOI: 10.4103/2152-7806.188905] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 06/20/2016] [Indexed: 11/05/2022] Open
Abstract
Background: Graphene has unique electrical, physical, and chemical properties that may have great potential as a bioscaffold for neuronal regeneration after spinal cord injury. These nanoscaffolds have previously been shown to be biocompatible in vitro; in the present study, we wished to evaluate its biocompatibility in an in vivo spinal cord injury model. Methods: Graphene nanoscaffolds were prepared by the mild chemical reduction of graphene oxide. Twenty Wistar rats (19 male and 1 female) underwent hemispinal cord transection at approximately the T2 level. To bridge the lesion, graphene nanoscaffolds with a hydrogel were implanted immediately after spinal cord transection. Control animals were treated with hydrogel matrix alone. Histologic evaluation was performed 3 months after the spinal cord transection to assess in vivo biocompatibility of graphene and to measure the ingrowth of tissue elements adjacent to the graphene nanoscaffold. Results: The graphene nanoscaffolds adhered well to the spinal cord tissue. There was no area of pseudocyst around the scaffolds suggestive of cytotoxicity. Instead, histological evaluation showed an ingrowth of connective tissue elements, blood vessels, neurofilaments, and Schwann cells around the graphene nanoscaffolds. Conclusions: Graphene is a nanomaterial that is biocompatible with neurons and may have significant biomedical application. It may provide a scaffold for the ingrowth of regenerating axons after spinal cord injury.
Collapse
Affiliation(s)
- Ali H Palejwala
- Department of Neurosurgery, Baylor College of Medicine, Houston, Texas, USA; Division of Pediatric Neurosurgery, Texas Children's Hospital, Houston, Texas, USA
| | - Jared S Fridley
- Department of Neurosurgery, Baylor College of Medicine, Houston, Texas, USA; Division of Pediatric Neurosurgery, Texas Children's Hospital, Houston, Texas, USA
| | - Javier A Mata
- Department of Neurosurgery, Baylor College of Medicine, Houston, Texas, USA; Division of Pediatric Neurosurgery, Texas Children's Hospital, Houston, Texas, USA
| | | | - Thomas G Luerssen
- Department of Neurosurgery, Baylor College of Medicine, Houston, Texas, USA; Division of Pediatric Neurosurgery, Texas Children's Hospital, Houston, Texas, USA
| | - Laszlo Perlaky
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA; Research and Tissue Support Services Core Laboratory, Texas Children's Cancer and Hematology Services, Houston, Texas, USA
| | - Thomas A Kent
- Department of Neurology, Baylor College of Medicine, Houston, Texas, USA; Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, Texas, USA; Center for Translational Research in Inflammatory Diseases, Michael E. DeBakey VA Medical Center, Houston, Texas, USA
| | - James M Tour
- Department of Chemistry, Rice University, Houston, Texas, USA; Department of Chemistry and Materials Science and NanoEngineering, Rice University, Houston, Texas, USA
| | - Andrew Jea
- Department of Neurosurgery, Baylor College of Medicine, Houston, Texas, USA; Division of Pediatric Neurosurgery, Texas Children's Hospital, Houston, Texas, USA
| |
Collapse
|
43
|
Chhabra HS, Sarda K, Arora M, Sharawat R, Singh V, Nanda A, Sangodimath GM, Tandon V. Autologous bone marrow cell transplantation in acute spinal cord injury--an Indian pilot study. Spinal Cord 2015; 54:57-64. [PMID: 26282492 DOI: 10.1038/sc.2015.134] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Revised: 07/01/2015] [Accepted: 07/03/2015] [Indexed: 12/29/2022]
Abstract
STUDY DESIGN Phase- I/II, prospective, randomized, single-blind, controlled pilot study. PRIMARY OBJECTIVE To determine the safety and feasibility of autologous bone marrow transplantation in patients with acute spinal cord injury (SCI) via two routes of transplantation as compared with controls. SETTING Indian Spinal Injuries Center, New Delhi. METHODS Twenty-one subjects with acute, American Spinal Injury Association Impairment Scale (AIS) A (complete), traumatic SCI with neurological level T1-T12, were recruited and randomized into three groups of seven subjects each. Two groups underwent cell transplantation through the intrathecal or intralesional route, whereas the third served as control. Participants were assessed at baseline and followed up at 6 months and 12-months post enrollment. Safety and tolerability were evaluated by monitoring for any adverse events. Efficacy was assessed through neurological, functional and psychological evaluation, as well as through electrophysiological studies and urodynamics. RESULTS Surgery was tolerated well by all participants. There were no significant adverse events attributable to the procedure. There was no significant improvement in the neurological, electrophysiological or urodynamic efficacy variables. A statistically significant improvement in functional scores as evaluated by the Spinal Cord Independence Measure and International Spinal Cord Injury Scale was observed in all groups. CONCLUSIONS The procedure is safe and feasible in AIS A participants with thoracic-level injuries at 12-months follow-up. No efficacy could be demonstrated that could be attributed to the procedure.
Collapse
Affiliation(s)
- H S Chhabra
- Spine Service, Indian Spinal Injuries Center, New Delhi, India
| | - K Sarda
- Spine Service, Indian Spinal Injuries Center, New Delhi, India
| | - M Arora
- Spine Service, Indian Spinal Injuries Center, New Delhi, India
| | - R Sharawat
- Spine Service, Indian Spinal Injuries Center, New Delhi, India
| | - V Singh
- Spine Service, Indian Spinal Injuries Center, New Delhi, India
| | - A Nanda
- Spine Service, Indian Spinal Injuries Center, New Delhi, India
| | - G M Sangodimath
- Spine Service, Indian Spinal Injuries Center, New Delhi, India
| | - V Tandon
- Spine Service, Indian Spinal Injuries Center, New Delhi, India
| |
Collapse
|
44
|
Abstract
Stem cell-based interventions aim to use special regenerative cells (stem cells) to facilitate neuronal function beyond the site of the injury. Many studies involving animal models of spinal cord injury (SCI) suggest that certain stem cell-based therapies may restore function after SCI. Currently, in case of spinal cord injuries, new discoveries with clinical implications have been continuously made in basic stem cell research, and stem cell-based approaches are advancing rapidly toward application in patients. There is a huge base of preclinical evidence in vitro and in animal models which suggests the safety and clinical efficacy of cellular therapies after SCI. Despite this, data from clinical studies is not very encouraging and at times confounding. Here, we have attempted to cover preclinical and clinical evidence base dealing with safety, feasibility and efficacy of cell based interventions after SCI. The limitations of preclinical data and the reasons underlying its failure to translate in a clinical setting are also discussed. Based on the evidence base, it is suggested that a multifactorial approach is required to address this situation. Need for standardized, stringently designed multi-centric clinical trials for obtaining validated proof of evidence is also highlighted.
Collapse
Affiliation(s)
- Harvinder Singh Chhabra
- Spine Service, Indian Spinal Injuries Centre, Vasant Kunj, New Delhi, India,Address for correspondence: Dr. Harvinder Singh Chhabra, Indian Spinal Injuries Centre, Sector C, Vasant Kunj, New Delhi - 110 070, India. E-mail:
| | - Kanchan Sarda
- Spine Service, Indian Spinal Injuries Centre, Vasant Kunj, New Delhi, India
| |
Collapse
|