1
|
Dill-Macky AS, Lee EN, Wertheim JA, Koss KM. Glia in tissue engineering: From biomaterial tools to transplantation. Acta Biomater 2024; 190:24-49. [PMID: 39396630 DOI: 10.1016/j.actbio.2024.10.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 10/01/2024] [Accepted: 10/10/2024] [Indexed: 10/15/2024]
Abstract
Glia are imperative in nearly every function of the nervous system, including neurotransmission, neuronal repair, development, immunity, and myelination. Recently, the reparative roles of glia in the central and peripheral nervous systems have been elucidated, suggesting a tremendous potential for these cells as novel treatments to central nervous system disorders. Glial cells often behave as 'double-edged swords' in neuroinflammation, ultimately deciding the life or death of resident cells. Compared to glia, neuronal cells have limited mobility, lack the ability to divide and self-renew, and are generally more delicate. Glia have been candidates for therapeutic use in many successful grafting studies, which have been largely focused on restoring myelin with Schwann cells, olfactory ensheathing glia, and oligodendrocytes with support from astrocytes. However, few therapeutics of this class have succeeded past clinical trials. Several tools and materials are being developed to understand and re-engineer these grafting concepts for greater success, such as extra cellular matrix-based scaffolds, bioactive peptides, biomolecular delivery systems, biomolecular discovery for neuroinflammatory mediation, composite microstructures such as artificial channels for cell trafficking, and graft enhanced electrical stimulation. Furthermore, advances in stem cell-derived cortical/cerebral organoid differentiation protocols have allowed for the generation of patient-derived glia comparable to those acquired from tissues requiring highly invasive procedures or are otherwise inaccessible. However, research on bioengineered tools that manipulate glial cells is nowhere near as comprehensive as that for systems of neurons and neural stem cells. This article explores the therapeutic potential of glia in transplantation with an emphasis on novel bioengineered tools for enhancement of their reparative properties. STATEMENT OF SIGNIFICANCE: Neural glia are responsible for a host of developmental, homeostatic, and reparative roles in the central nervous system but are often a major cause of tissue damage and cellular loss in insults and degenerative pathologies. Most glial grafts have employed Schwann cells for remyelination, but other glial with novel biomaterials have been employed, emphasizing their diverse functionality. Promising strategies have emerged, including neuroimmune mediation of glial scar tissues and facilitated migration and differentiation of stem cells for neural replacement. Herein, a comprehensive review of biomaterial tools for glia in transplantation is presented, highlighting Schwann cells, astrocytes, olfactory ensheating glia, oligodendrocytes, microglia, and ependymal cells.
Collapse
Affiliation(s)
- A S Dill-Macky
- Department of Surgery, University of Arizona, 1501 N Campbell Ave, Tucson, AZ 85724, United States
| | - E N Lee
- Department of Surgery, University of Arizona, 1501 N Campbell Ave, Tucson, AZ 85724, United States
| | - J A Wertheim
- Department of Surgery, University of Arizona, 1501 N Campbell Ave, Tucson, AZ 85724, United States
| | - K M Koss
- Department of Neurobiology, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555-0625, United States; Sealy Institute for Drug Discovery, University of Texas Medical Branch, 105 11th Street Galveston, TX 77555-1110, United States.
| |
Collapse
|
2
|
Stankovic KM, Batts S, Welling DB, Vasilijic S. Immune Profiling of Secreted Factors from Human Vestibular Schwannoma Cells and Tumor-associated Macrophages. Laryngoscope 2024; 134 Suppl 5:S1-S14. [PMID: 37776249 DOI: 10.1002/lary.31067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 07/27/2023] [Accepted: 09/12/2023] [Indexed: 10/02/2023]
Abstract
OBJECTIVES This study compared the immune-related secretory capacity of human vestibular schwannoma (VS) and tumor-assisted macrophages (TAMs) with their normal counterparts (Schwann cells [SC] and peripheral blood monocyte-derived macrophages [Mo-MFs], respectively), and examined relationships with presurgical hearing and tumor size. METHODS VS tumors (n = 16), auditory nerve (n = 1), blood (n = 9), and great auricular nerves (n = 3) were used. SCs (S100B+ ) and TAMs (CD68+ ) were isolated from VS tissue for culture. The secreted levels of 65 immune-related factors were measured and compared using unpaired t-tests with Welch correction (schwannoma vs. SCs) or Mann-Whitney tests (TAMs and Mo-MFs). Associations between factor concentration and word recognition (WR), pure-tone average (PTA), and tumor size were evaluated with Spearman correlation. RESULTS Secreted factors with significantly higher concentrations in schwannoma versus SC supernatants included IL-2 and BAFF, whereas MMP-1, IL-6, FGF-2, VEGF-A, MIP-3α, and GRO-α concentrations were significantly higher in TAMs versus Mo-MFs (all p < 0.05). Worse WR was significantly associated with higher secretion of fractalkine, eotaxin-3, CD30, and IL-16 by VS cells; IP-10, eotaxin-3, multiple interleukins, GM-CSF, SCF, and CD30 by TAMs; and TNF-α and MIP-1α by Mo-MFs (all p < 0.05). Worse PTA was significantly correlated with higher secretion of IL-16 by VS cells (p < 0.05). Larger tumor size was significantly correlated with higher secretion of eotaxin by VS cells, and of IL-7, IL-21, and LIF by TAMs (all p = 0.017). CONCLUSIONS Differential secretion of immune-related factors was observed in schwannoma versus normal SCs and in TAMs versus Mo-MFs, some of which were correlated with worse hearing and larger VS tumors. LEVEL OF EVIDENCE N/A Laryngoscope, 134:S1-S14, 2024.
Collapse
Affiliation(s)
- Konstantina M Stankovic
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine, Palo Alto, California, U.S.A
- Department of Neurosurgery, Stanford University School of Medicine, Palo Alto, California, U.S.A
- Wu Tsai Neuroscience Institute, Stanford University, Palo Alto, California, U.S.A
- Department of Otolaryngology-Head and Neck Surgery at Massachusetts Eye and Ear and, Harvard Medical School, Boston, Massachusetts, U.S.A
| | - Shelley Batts
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine, Palo Alto, California, U.S.A
| | - D Bradley Welling
- Department of Otolaryngology-Head and Neck Surgery at Massachusetts Eye and Ear and, Harvard Medical School, Boston, Massachusetts, U.S.A
| | - Sasa Vasilijic
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine, Palo Alto, California, U.S.A
- Department of Otolaryngology-Head and Neck Surgery at Massachusetts Eye and Ear and, Harvard Medical School, Boston, Massachusetts, U.S.A
| |
Collapse
|
3
|
Stratton S, Wang S, Hashemi S, Pressman Y, Nanchanatt J, Oudega M, Arinzeh TL. A scaffold containing zinc oxide for Schwann cell-mediated axon growth. J Neural Eng 2023; 20:066009. [PMID: 37931311 DOI: 10.1088/1741-2552/ad0a00] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 11/06/2023] [Indexed: 11/08/2023]
Abstract
Objective.Schwann cells (SCs) transplanted in damaged nervous tissue promote axon growth, which may support the recovery of function lost after injury. However, SC transplant-mediated axon growth is often limited and lacks direction.Approach.We have developed a zinc oxide (ZnO) containing fibrous scaffold consisting of aligned fibers of polycaprolactone (PCL) with embedded ZnO nanoparticles as a biodegradable, bifunctional scaffold for promoting and guiding axon growth. This scaffold has bifunctional properties wherein zinc is released providing bioactivity and ZnO has well-known piezoelectric properties where piezoelectric materials generate electrical activity in response to minute deformations. In this study, SC growth, SC-mediated axon extension, and the presence of myelin basic protein (MBP), as an indicator of myelination, were evaluated on the scaffolds containing varying concentrations of ZnOin vitro. SCs and dorsal root ganglion (DRG) neurons were cultured, either alone or in co-culture, on the scaffolds.Main results.Findings demonstrated that scaffolds with 1 wt.% ZnO promoted the greatest SC growth and SC-mediated axon extension. The presence of brain-derived neurotrophic factor (BDNF) was also determined. BDNF increased in co-cultures for all scaffolds as compared to SCs or DRGs cultured alone on all scaffolds. For co-cultures, cells on scaffolds with low levels of ZnO (0.5 wt.% ZnO) had the highest amount of BDNF as compared to cells on higher ZnO-containing scaffolds (1 and 2 wt.%). MBP immunostaining was only detected in co-cultures on PCL control scaffolds (without ZnO).Significance.The results of this study demonstrate the potential of the ZnO-containing scaffolds for SC-mediated axon growth and its potential for use in nervous tissue repair.
Collapse
Affiliation(s)
- Scott Stratton
- Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, United States of America
| | - Shuo Wang
- Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, United States of America
| | - Sharareh Hashemi
- Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, United States of America
| | - Yelena Pressman
- The Miami Project, University of Miami, Miami, FL, United States of America
| | - James Nanchanatt
- Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, United States of America
| | - Martin Oudega
- Physical Therapy & Human Movement Sciences and Physiology, Northwestern University, Chicago, IL, United States of America
- Hines VA Hospital, Hines, IL, United States of America
- Shirley Ryan AbilityLab, Chicago, IL, United States of America
| | - Treena Livingston Arinzeh
- Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, United States of America
- Biomedical Engineering, Columbia University, New York, NY, United States of America
| |
Collapse
|
4
|
Takeya H, Itai S, Kimura H, Kurashina Y, Amemiya T, Nagoshi N, Iwamoto T, Sato K, Shibata S, Matsumoto M, Onoe H, Nakamura M. Schwann cell-encapsulated chitosan-collagen hydrogel nerve conduit promotes peripheral nerve regeneration in rodent sciatic nerve defect models. Sci Rep 2023; 13:11932. [PMID: 37488180 PMCID: PMC10366170 DOI: 10.1038/s41598-023-39141-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 07/20/2023] [Indexed: 07/26/2023] Open
Abstract
Chitosan has various tissue regeneration effects. This study was designed to investigate the nerve regeneration effect of Schwann cell (SC)-encapsulated chitosan-collagen hydrogel nerve conduit (CCN) transplanted into a rat model of sciatic nerve defect. We prepared a CCN consisting of an outer layer of chitosan hydrogel and an inner layer of collagen hydrogel to encapsulate the intended cells. Rats with a 10-mm sciatic nerve defect were treated with SCs encapsulated in CCN (CCN+), CCN without SCs (CCN-), SC-encapsulated silicone tube (silicone+), and autologous nerve transplanting (auto). Behavioral and histological analyses indicated that motor functional recovery, axonal regrowth, and myelination of the CCN+ group were superior to those of the CCN- and silicone+ groups. Meanwhile, the CCN- and silicone+ groups showed no significant differences in the recovery of motor function and nerve histological restoration. In conclusion, SC-encapsulated CCN has a synergistic effect on peripheral nerve regeneration, especially axonal regrowth and remyelination of host SCs. In the early phase after transplantation, SC-encapsulated CCNs have a positive effect on recovery. Therefore, using SC-encapsulated CCNs may be a promising approach for massive peripheral nerve defects.
Collapse
Affiliation(s)
- Hiroaki Takeya
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi Shinjuku-Ku, Tokyo, 160-8582, Japan
| | - Shun Itai
- Department of Mechanical Engineering, Faculty of Science and Technology, Keio University, 3-14-1 Hiyoshi, Kohoku-Ku, Yokohama-Shi, Kanagawa, 223-8522, Japan
- Division of Medical Science, Graduate School of Biomedical Engineering, Tohoku University, 1-1 Seiryomachi, Aoba-Ku, Sendai, Miyagi, 980-8574, Japan
| | - Hiroo Kimura
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi Shinjuku-Ku, Tokyo, 160-8582, Japan.
| | - Yuta Kurashina
- Division of Advanced Mechanical Systems Engineering, Institute of Engineering, Tokyo University of Agriculture and Technology, 2-24-16 Nakacho, Koganei-Shi, Tokyo, 184-8588, Japan
| | - Tsuyoshi Amemiya
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi Shinjuku-Ku, Tokyo, 160-8582, Japan
| | - Narihito Nagoshi
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi Shinjuku-Ku, Tokyo, 160-8582, Japan
| | - Takuji Iwamoto
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi Shinjuku-Ku, Tokyo, 160-8582, Japan
| | - Kazuki Sato
- Institute for Integrated Sports Medicine, Keio University School of Medicine, 35 Shinanomachi Shinjuku-Ku, Tokyo, Japan
| | - Shinsuke Shibata
- Division of Microscopic Anatomy, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-Dori, Chuo-Ku, Niigata, 951-8510, Japan
| | - Morio Matsumoto
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi Shinjuku-Ku, Tokyo, 160-8582, Japan
| | - Hiroaki Onoe
- Department of Mechanical Engineering, Faculty of Science and Technology, Keio University, 3-14-1 Hiyoshi, Kohoku-Ku, Yokohama-Shi, Kanagawa, 223-8522, Japan
| | - Masaya Nakamura
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi Shinjuku-Ku, Tokyo, 160-8582, Japan
| |
Collapse
|
5
|
Hu T, Chang S, Qi F, Zhang Z, Chen J, Jiang L, Wang D, Deng C, Nie K, Xu G, Wei Z. Neural grafts containing exosomes derived from Schwann cell-like cells promote peripheral nerve regeneration in rats. BURNS & TRAUMA 2023; 11:tkad013. [PMID: 37122841 PMCID: PMC10141455 DOI: 10.1093/burnst/tkad013] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 12/06/2022] [Accepted: 03/02/2023] [Indexed: 05/02/2023]
Abstract
Background Schwann cell-like cells (SCLCs), differentiated from mesenchymal stem cells, have shown promising outcomes in the treatment of peripheral nerve injuries in preclinical studies. However, certain clinical obstacles limit their application. Hence, the primary aim of this study was to investigate the role of exosomes derived from SCLCs (SCLCs-exo) in peripheral nerve regeneration. Methods SCLCs were differentiated from human amniotic mesenchymal stem cells (hAMSCs) in vitro and validated by immunofluorescence, real-time quantitative PCR and western blot analysis. Exosomes derived from hAMSCs (hAMSCs-exo) and SCLCs were isolated by ultracentrifugation and validated by nanoparticle tracking analysis, WB analysis and electron microscopy. A prefabricated nerve graft was used to deliver hAMSCs-exo or SCLCs-exo in an injured sciatic nerve rat model. The effects of hAMSCs-exo or SCLCs-exo on rat peripheral nerve injury (PNI) regeneration were determined based on the recovery of neurological function and histomorphometric variation. The effects of hAMSCs-exo or SCLCs-exo on Schwann cells were also determined via cell proliferation and migration assessment. Results SCLCs significantly expressed the Schwann cell markers glial fibrillary acidic protein and S100. Compared to hAMSCs-exo, SCLCs-exo significantly enhanced motor function recovery, attenuated gastrocnemius muscle atrophy and facilitated axonal regrowth, myelin formation and angiogenesis in the rat model. Furthermore, hAMSCs-exo and SCLCs-exo were efficiently absorbed by Schwann cells. However, compared to hAMSCs-exo, SCLCs-exo significantly promoted the proliferation and migration of Schwann cells. SCLCs-exo also significantly upregulated the expression of a glial cell-derived neurotrophic factor, myelin positive regulators (SRY-box transcription factor 10, early growth response protein 2 and organic cation/carnitine transporter 6) and myelin proteins (myelin basic protein and myelin protein zero) in Schwann cells. Conclusions These findings suggest that SCLCs-exo can more efficiently promote PNI regeneration than hAMSCs-exo and are a potentially novel therapeutic approach for treating PNI.
Collapse
Affiliation(s)
| | | | - Fang Qi
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, No. 149 Dalian Road, Huichuan District, Zunyi, Guizhou 563003, China
- The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi medical University, No. 6 West Xuefu Road, Xinpu District, Zunyi, Guizhou, 563003, China
| | - Zhonghui Zhang
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, No. 149 Dalian Road, Huichuan District, Zunyi, Guizhou 563003, China
- The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi medical University, No. 6 West Xuefu Road, Xinpu District, Zunyi, Guizhou, 563003, China
| | - Jiayin Chen
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, No. 149 Dalian Road, Huichuan District, Zunyi, Guizhou 563003, China
- The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi medical University, No. 6 West Xuefu Road, Xinpu District, Zunyi, Guizhou, 563003, China
| | - Lingli Jiang
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, No. 149 Dalian Road, Huichuan District, Zunyi, Guizhou 563003, China
- The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi medical University, No. 6 West Xuefu Road, Xinpu District, Zunyi, Guizhou, 563003, China
| | - Dali Wang
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, No. 149 Dalian Road, Huichuan District, Zunyi, Guizhou 563003, China
- The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi medical University, No. 6 West Xuefu Road, Xinpu District, Zunyi, Guizhou, 563003, China
| | - Chengliang Deng
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, No. 149 Dalian Road, Huichuan District, Zunyi, Guizhou 563003, China
- The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi medical University, No. 6 West Xuefu Road, Xinpu District, Zunyi, Guizhou, 563003, China
| | - Kaiyu Nie
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, No. 149 Dalian Road, Huichuan District, Zunyi, Guizhou 563003, China
- The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi medical University, No. 6 West Xuefu Road, Xinpu District, Zunyi, Guizhou, 563003, China
| | | | - Zairong Wei
- Correspondence. Guangchao Xu, ; Zairong Wei,
| |
Collapse
|
6
|
Muheremu A, Shu L, Liang J, Aili A, Jiang K. Sustained delivery of neurotrophic factors to treat spinal cord injury. Transl Neurosci 2021; 12:494-511. [PMID: 34900347 PMCID: PMC8633588 DOI: 10.1515/tnsci-2020-0200] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 11/04/2021] [Accepted: 11/05/2021] [Indexed: 12/16/2022] Open
Abstract
Acute spinal cord injury (SCI) is a devastating condition that results in tremendous physical and psychological harm and a series of socioeconomic problems. Although neurons in the spinal cord need neurotrophic factors for their survival and development to reestablish their connections with their original targets, endogenous neurotrophic factors are scarce and the sustainable delivery of exogeneous neurotrophic factors is challenging. The widely studied neurotrophic factors such as brain-derived neurotrophic factor, neurotrophin-3, nerve growth factor, ciliary neurotrophic factor, basic fibroblast growth factor, and glial cell-derived neurotrophic factor have a relatively short cycle that is not sufficient enough for functionally significant neural regeneration after SCI. In the past decades, scholars have tried a variety of cellular and viral vehicles as well as tissue engineering scaffolds to safely and sustainably deliver those necessary neurotrophic factors to the injury site, and achieved satisfactory neural repair and functional recovery on many occasions. Here, we review the neurotrophic factors that have been used in trials to treat SCI, and vehicles that were commonly used for their sustained delivery.
Collapse
Affiliation(s)
- Aikeremujiang Muheremu
- Department of Spine Surgery, Sixth Affiliated Hospital of Xinjiang Medical University, 39 Wuxing Nan Rd, Tianshan District, Urumqi, Xinjiang, 86830001, People’s Republic of China
| | - Li Shu
- Department of Orthopedics, Sixth Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 86830001, People’s Republic of China
| | - Jing Liang
- Department of Laboratory Medicine, Sixth Affiliated Hospital of Xinjiang Medical University, 39, Wuxing Nan Rd, Tianshan District, Urumqi, Xinjiang, 86830001, People’s Republic of China
| | - Abudunaibi Aili
- Department of Spine Surgery, Sixth Affiliated Hospital of Xinjiang Medical University, 39 Wuxing Nan Rd, Tianshan District, Urumqi, Xinjiang, 86830001, People’s Republic of China
| | - Kan Jiang
- Department of Orthopedics, Sixth Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 86830001, People’s Republic of China
| |
Collapse
|
7
|
Spinal Reflex Recovery after Dorsal Rhizotomy and Repair with Platelet-Rich Plasma (PRP) Gel Combined with Bioengineered Human Embryonic Stem Cells (hESCs). Stem Cells Int 2020; 2020:8834360. [PMID: 33178285 PMCID: PMC7647752 DOI: 10.1155/2020/8834360] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 09/20/2020] [Accepted: 09/30/2020] [Indexed: 02/07/2023] Open
Abstract
Dorsal root rhizotomy (DRZ) is currently considered an untreatable injury, resulting in the loss of sensitive function and usually leading to neuropathic pain. In this context, we recently proposed a new surgical approach to treat DRZ that uses platelet-rich plasma (PRP) gel to restore the spinal reflex. Success was correlated with the reentry of primary afferents into the spinal cord. Here, aiming to enhance previous results, cell therapy with bioengineered human embryonic stem cells (hESCs) to overexpress fibroblast growth factor 2 (FGF2) was combined with PRP. For these experiments, adult female rats were submitted to a unilateral rhizotomy of the lumbar spinal dorsal roots, which was followed by root repair with PRP gel with or without bioengineered hESCs. One week after DRZ, the spinal cords were processed to evaluate changes in the glial response (GFAP and Iba-1) and excitatory synaptic circuits (VGLUT1) by immunofluorescence. Eight weeks postsurgery, the lumbar intumescences were processed for analysis of the repaired microenvironment by transmission electron microscopy. Spinal reflex recovery was evaluated by the electronic Von Frey method for eight weeks. The transcript levels for human FGF2 were over 37-fold higher in the induced hESCs than in the noninduced and the wildtype counterparts. Altogether, the results indicate that the combination of hESCs with PRP gel promoted substantial and prominent axonal regeneration processes after DRZ. Thus, the repair of dorsal roots, if done appropriately, may be considered an approach to regain sensory-motor function after dorsal root axotomy.
Collapse
|
8
|
Yu Z, Li H, Xia P, Kong W, Chang Y, Fu C, Wang K, Yang X, Qi Z. Application of fibrin-based hydrogels for nerve protection and regeneration after spinal cord injury. J Biol Eng 2020; 14:22. [PMID: 32774454 PMCID: PMC7397605 DOI: 10.1186/s13036-020-00244-3] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 07/27/2020] [Indexed: 12/13/2022] Open
Abstract
Traffic accidents, falls, and many other events may cause traumatic spinal cord injuries (SCIs), resulting in nerve cells and extracellular matrix loss in the spinal cord, along with blood loss, inflammation, oxidative stress (OS), and others. The continuous development of neural tissue engineering has attracted increasing attention on the application of fibrin hydrogels in repairing SCIs. Except for excellent biocompatibility, flexibility, and plasticity, fibrin, a component of extracellular matrix (ECM), can be equipped with cells, ECM protein, and various growth factors to promote damage repair. This review will focus on the advantages and disadvantages of fibrin hydrogels from different sources, as well as the various modifications for internal topographical guidance during the polymerization. From the perspective of further improvement of cell function before and after the delivery of stem cell, cytokine, and drug, this review will also evaluate the application of fibrin hydrogels as a carrier to the therapy of nerve repair and regeneration, to mirror the recent development tendency and challenge.
Collapse
Affiliation(s)
- Ziyuan Yu
- Department of Orthopedic Surgery, The Second Hospital of Jilin University, Ziqiang Street No. 218, Changchun, TX 130041 PR China
| | - Hongru Li
- Department of Orthopedic Surgery, The Second Hospital of Jilin University, Ziqiang Street No. 218, Changchun, TX 130041 PR China
| | - Peng Xia
- Department of Orthopedic Surgery, The Second Hospital of Jilin University, Ziqiang Street No. 218, Changchun, TX 130041 PR China
| | - Weijian Kong
- Department of Orthopedic Surgery, The Second Hospital of Jilin University, Ziqiang Street No. 218, Changchun, TX 130041 PR China
| | - Yuxin Chang
- Department of Orthopedic Surgery, The Second Hospital of Jilin University, Ziqiang Street No. 218, Changchun, TX 130041 PR China
| | - Chuan Fu
- Department of Orthopedic Surgery, The Second Hospital of Jilin University, Ziqiang Street No. 218, Changchun, TX 130041 PR China
| | - Kai Wang
- Department of Orthopedic Surgery, The Second Hospital of Jilin University, Ziqiang Street No. 218, Changchun, TX 130041 PR China
| | - Xiaoyu Yang
- Department of Orthopedic Surgery, The Second Hospital of Jilin University, Ziqiang Street No. 218, Changchun, TX 130041 PR China
| | - Zhiping Qi
- Department of Orthopedic Surgery, The Second Hospital of Jilin University, Ziqiang Street No. 218, Changchun, TX 130041 PR China
| |
Collapse
|
9
|
Systemic treatment with a novel basic fibroblast growth factor mimic small-molecule compound boosts functional recovery after spinal cord injury. PLoS One 2020; 15:e0236050. [PMID: 32678832 PMCID: PMC7367485 DOI: 10.1371/journal.pone.0236050] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 06/26/2020] [Indexed: 11/19/2022] Open
Abstract
Neurotrophic factors have been regarded having promising potentials for neuronal protection and regeneration, and thus promoting beneficial effects of kinesiological functions. They can be suspected to play important roles in cell/tissue grafting for various neural diseases. The clinical applications of such trophic factors to the central nervous system (CNS), however, have caused problematic side effects on account of the distinctive bioactive properties. In the course of developing synthetic compounds reflecting beneficial properties of basic fibroblast growth factor (bFGF), we conducted screening candidates that stimulate to trigger the intracellular tyrosine phosphorylation of FGF receptor and lead to the subsequent intracellular signaling in neurons. A small synthetic molecule SUN13837 was characterized by mimicking the beneficial properties of bFGF, which have been known as its specific activities when applied to CNS. What is more remarkable is that SUN13837 is eliminated the bioactivity to induce cell proliferation of non-neuronal somatic cells. On the bases of studies of pharmacology, behavior, physiology and histology, the present study reports that SUN13837 is characterized as a promising synthetic compound for treatment of devastating damages onto the rat spinal cord.
Collapse
|
10
|
The effect of Matrigel as scaffold material for neural stem cell transplantation for treating spinal cord injury. Sci Rep 2020; 10:2576. [PMID: 32054865 PMCID: PMC7018993 DOI: 10.1038/s41598-020-59148-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 01/24/2020] [Indexed: 11/09/2022] Open
Abstract
Traumatic injury to the spinal cord causes permanent loss of function and major personal, social, and economic problems. Cell-based delivery strategies is a promising approach for treating spinal cord injury (SCI). However, the inhospitable microenvironment in the injured spinal cord results in poor cell survival and uncontrolled differentiation of the transplanted stem cells. The combination of a scaffold with cells has been developed with a tendency for achieving greater survival and integration with the host tissue. We investigated the effect of Matrigel combined with neural stem cells (NSCs) in vitro and in vivo. We compared the effect of different types of scaffold on the survival and differentiation of brain-derived NSCs in an in vitro culture. Subsequently, NSCs were transplanted subcutaneously into nude mice to detect graft survival and differentiation in vivo. Finally, phosphate-buffered saline (PBS), Matrigel alone, or Matrigel seeded with NSCs was injected into 48 subacute, clinically relevant rat models of SCI (16 rats per group). Matrigel supported cell survival and differentiation efficiently in vitro and in vivo. SCI rats transplanted with NSCs in Matrigel showed improved behavioral recovery and neuronal and reactive astrocyte marker expression levels compared to PBS- or Matrigel-transplanted rats. Functional repair and neuronal and reactive astrocyte marker expression was slightly improved in the Matrigel-alone group relative to the PBS group, but not statistically significantly. These data suggest that Matrigel is a promising scaffold material for cell transplantation to the injured spinal cord.
Collapse
|
11
|
Goldshmit Y, Tang JKKY, Siegel AL, Nguyen PD, Kaslin J, Currie PD, Jusuf PR. Different Fgfs have distinct roles in regulating neurogenesis after spinal cord injury in zebrafish. Neural Dev 2018; 13:24. [PMID: 30447699 PMCID: PMC6240426 DOI: 10.1186/s13064-018-0122-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 11/08/2018] [Indexed: 11/10/2022] Open
Abstract
Background Despite conserved developmental processes and organization of the vertebrate central nervous system, only some vertebrates including zebrafish can efficiently regenerate neural damage including after spinal cord injury. The mammalian spinal cord shows very limited regeneration and neurogenesis, resulting in permanent life-long functional impairment. Therefore, there is an urgent need to identify the cellular and molecular mechanisms that can drive efficient vertebrate neurogenesis following injury. A key pathway implicated in zebrafish neurogenesis is fibroblast growth factor signaling. Methods In the present study we investigated the roles of distinct fibroblast growth factor members and their receptors in facilitating different aspects of neural development and regeneration at different timepoints following spinal cord injury. After spinal cord injury in adults and during larval development, loss and/or gain of Fgf signaling was combined with immunohistochemistry, in situ hybridization and transgenes marking motor neuron populations in in vivo zebrafish and in vitro mammalian PC12 cell culture models. Results Fgf3 drives neurogenesis of Islet1 expressing motor neuron subtypes and mediate axonogenesis in cMet expressing motor neuron subtypes. We also demonstrate that the role of Fgf members are not necessarily simple recapitulating development. During development Fgf2, Fgf3 and Fgf8 mediate neurogenesis of Islet1 expressing neurons and neuronal sprouting of both, Islet1 and cMet expressing motor neurons. Strikingly in mammalian PC12 cells, all three Fgfs increased cell proliferation, however, only Fgf2 and to some extent Fgf8, but not Fgf3 facilitated neurite outgrowth. Conclusions This study demonstrates differential Fgf member roles during neural development and adult regeneration, including in driving neural proliferation and neurite outgrowth of distinct spinal cord neuron populations, suggesting that factors including Fgf type, age of the organism, timing of expression, requirements for different neuronal populations could be tailored to best drive all of the required regenerative processes.
Collapse
Affiliation(s)
- Yona Goldshmit
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC, 3800, Australia.,Steyer School of Health Professions, Sackler School of Medicine, Tel-Aviv University, P.O. Box 39040, 6997801, Tel Aviv, Israel
| | - Jean Kitty K Y Tang
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC, 3800, Australia
| | - Ashley L Siegel
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC, 3800, Australia
| | - Phong D Nguyen
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC, 3800, Australia
| | - Jan Kaslin
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC, 3800, Australia
| | - Peter D Currie
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC, 3800, Australia
| | - Patricia R Jusuf
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC, 3800, Australia. .,School of Biosciences, University of Melbourne, Parkville, VIC, 3010, Australia.
| |
Collapse
|
12
|
Rochkind S. Photobiomodulation in Neuroscience: A Summary of Personal Experience. Photomed Laser Surg 2018; 35:604-615. [PMID: 29099679 DOI: 10.1089/pho.2017.4381] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE This review summarizes personal experience with laser photobiomodulation and its potentials for the treatment of peripheral and central nerve system injuries. METHODS AND RESULTS Laser photobiomodulation was shown to induce nerve cell activation, have a positive effect on metabolism of the nerve cells, and to stimulate nerve sprouting processes. Studies investigating the effects of laser photobiomodulation on injured peripheral nerves in rats reported immediate protective effects which increase the functional activity of the nerve, decrease or prevent scar tissue formation at the injured site, prevent or decrease degeneration in corresponding motor neurons of the spinal cord, and significantly increase axonal growth and myelinization. A direct application of laser on the spinal cord had a positive impact on the corresponding injured peripheral nerve and promoted recovery. A 780-nm laser phototherapy was applied following peripheral nerve reconstruction using a guiding nerve tube. Results showed myelinated axons crossing through the nerve tube and the continuation of axonal sprouting through the tube toward the distal part of the nerve. In a double-blind, placebo-controlled randomized pilot clinical trial in patients with incomplete stable long-term peripheral nerve injury (PNI), 780-nm laser irradiation progressively improved peripheral nerve function and led to substantial functional recovery. Muscle atrophy represents a major challenge in restorative medicine. Laser phototherapy was shown to increase biochemical activity and improve morphological recovery in muscle and, thus, could have a direct therapeutic application, especially during progressive muscle atrophy resulting from PNI. The effectiveness of composite implants of cultured embryonal nerve cells and the role of laser irradiation on regeneration and repair of the completely transected rat spinal cord were examined. Results suggested that laser photobiomodulation treatment accelerates the axonal growth. CONCLUSIONS The significance of the performed experimental and clinical studies is in the provision of new laser technology in field of cell therapy and its therapeutic value for peripheral nerve and spinal cord injuries. Additional well-designed clinical studies are needed to evaluate the effectiveness and role of laser photobiomodulation treatment in a clinical setting.
Collapse
Affiliation(s)
- Shimon Rochkind
- Division of Peripheral Nerve Reconstruction, Department of Neurosurgery, Research Center for Nerve Reconstruction, Tel Aviv Sourasky Medical Center, Tel Aviv University , Tel Aviv, Israel
| |
Collapse
|
13
|
Wu S, Chen MS, Maurel P, Lee YS, Bunge MB, Arinzeh TL. Aligned fibrous PVDF-TrFE scaffolds with Schwann cells support neurite extension and myelination in vitro. J Neural Eng 2018; 15:056010. [PMID: 29794323 DOI: 10.1088/1741-2552/aac77f] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
OBJECTIVE Polyvinylidene fluoride-trifluoroethylene (PVDF-TrFE), which is a piezoelectric, biocompatible polymer, holds promise as a scaffold in combination with Schwann cells (SCs) for spinal cord repair. Piezoelectric materials can generate electrical activity in response to mechanical deformation, which could potentially stimulate spinal cord axon regeneration. Our goal in this study was to investigate PVDF-TrFE scaffolds consisting of aligned fibers in supporting SC growth and SC-supported neurite extension and myelination in vitro. APPROACH Aligned fibers of PVDF-TrFE were fabricated using the electrospinning technique. SCs and dorsal root ganglion (DRG) explants were co-cultured to evaluate SC-supported neurite extension and myelination on PVDF-TrFE scaffolds. MAIN RESULTS PVDF-TrFE scaffolds supported SC growth and neurite extension, which was further enhanced by coating the scaffolds with Matrigel. SCs were oriented and neurites extended along the length of the aligned fibers. SCs in co-culture with DRGs on PVDF-TrFE scaffolds promoted longer neurite extension as compared to scaffolds without SCs. In addition to promoting neurite extension, SCs also formed myelin around DRG neurites on PVDF-TrFE scaffolds. SIGNIFICANCE This study demonstrated PVDF-TrFE scaffolds containing aligned fibers supported SC-neurite extension and myelination. The combination of SCs and PVDF-TrFE scaffolds may be a promising tissue engineering strategy for spinal cord repair.
Collapse
Affiliation(s)
- Siliang Wu
- Materials Science and Engineering Program, New Jersey Institute of Technology, Newark, NJ 07102, United States of America
| | | | | | | | | | | |
Collapse
|
14
|
Lee YS, Funk LH, Lee JK, Bunge MB. Macrophage depletion and Schwann cell transplantation reduce cyst size after rat contusive spinal cord injury. Neural Regen Res 2018; 13:684-691. [PMID: 29722321 PMCID: PMC5950679 DOI: 10.4103/1673-5374.230295] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/22/2018] [Indexed: 01/07/2023] Open
Abstract
Schwann cell transplantation is a promising therapy for the treatment of spinal cord injury (SCI) and is currently in clinical trials. In our continuing efforts to improve Schwann cell transplantation strategies, we sought to determine the combined effects of Schwann cell transplantation with macrophage depletion. Since macrophages are major inflammatory contributors to the acute spinal cord injury, and are the major phagocytic cells, we hypothesized that transplanting Schwann cells after macrophage depletion will improve cell survival and integration with host tissue after SCI. To test this hypothesis, rat models of contusive SCI at thoracic level 8 were randomly subjected to macrophage depletion or not. In rat subjected to macrophage depletion, liposomes filled with clodronate were intraperitoneally injected at 1, 3, 6, 11, and 18 days post injury. Rats not subjected to macrophage depletion were intraperitoneally injected with liposomes filled with phosphate buffered saline. Schwann cells were transplanted 1 week post injury in all rats. Biotinylated dextran amine (BDA) was injected at thoracic level 5 to evalute axon regeneration. The Basso, Beattie, and Bresnahan locomotor test, Gridwalk test, and sensory test using von Frey filaments were performed to assess functional recovery. Immunohistochemistry was used to detect glial fibrillary acidic protein, neurofilament, and green fluorescent protein (GFP), and also to visulize BDA-labelled axons. The GFP labeled Schwann cell and cyst and lesion volumes were quantified using stained slides. The numbers of BDA-positive axons were also quantified. At 8 weeks after Schwann cell transplantation, there was a significant reduction in cyst and lesion volumes in the combined treatment group compared to Schwann cell transplantation alone. These changes were not associated, however, with improved Schwann cell survival, axon growth, or locomotor recovery. Although combining Schwann cell transplantation with macrophage depletion does improve histopathology of the injury site, the effect on axon growth and behavioral recovery appears no better than what can be achieved with Schwann cell transplants alone.
Collapse
Affiliation(s)
- Yee-Shuan Lee
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Lucy H. Funk
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Jae K. Lee
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Mary Bartlett Bunge
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Cell Biology, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
15
|
Abstract
Cell transplant-mediated tissue repair of the damaged spinal cord is being tested in several clinical trials. The current candidates are neural stem cells, stromal cells, and autologous Schwann cells (aSC). Due to their peripheral origin and limited penetration of astrocytic regions, aSC are transplanted intralesionally as compared to neural stem cells that are transplanted into intact spinal cord. Injections into either location can cause iatrogenic injury, and thus technical precision is important in the therapeutic risk-benefit equation. In this chapter, we discuss how we bridged from transplant studies in large animals to human application for two Phase 1 aSC transplant studies, one subacute and one chronic. Preclinical SC transplant studies conducted at the University of Miami in 2009-2012 in rodents, minipigs, and primates supported a successful Investigational New Drug (IND) submission for a Phase 1 trial in subacute complete spinal cord injury (SCI). Our studies optimized the safety and efficiency of intralesional cell delivery for subacute human SCI and led to the development of new simpler techniques for cell delivery into subjects with chronic SCI. Key parameters of delivery methodology include precision localization of the injury site, stereotaxic devices to control needle trajectory, method of entry into the spinal cord, spinal cord motion reduction, the volume and density of the cell suspension, rate of delivery, and control of shear stresses on cells.
Collapse
|
16
|
Lee YS, Wu S, Arinzeh TL, Bunge MB. Transplantation of Schwann Cells Inside PVDF-TrFE Conduits to Bridge Transected Rat Spinal Cord Stumps to Promote Axon Regeneration Across the Gap. J Vis Exp 2017. [PMID: 29155759 DOI: 10.3791/56077] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Among various models for spinal cord injury in rats, the contusion model is the most often used because it is the most common type of human spinal cord injury. The complete transection model, although not as clinically relevant as the contusion model, is the most rigorous method to evaluate axon regeneration. In the contusion model, it is difficult to distinguish regenerated from sprouted or spared axons due to the presence of remaining tissue post injury. In the complete transection model, a bridging method is necessary to fill the gap and create continuity from the rostral to the caudal stumps in order to evaluate the effectiveness of the treatments. A reliable bridging surgery is essential to test outcome measures by reducing the variability due to the surgical method. The protocols described here are used to prepare Schwann cells (SCs) and conduits prior to transplantation, complete transection of the spinal cord at thoracic level 8 (T8), insert the conduit, and transplant SCs into the conduit. This approach also uses in situ gelling of an injectable basement membrane matrix with SC transplantation that allows improved axon growth across the rostral and caudal interfaces with the host tissue.
Collapse
Affiliation(s)
- Yee-Shuan Lee
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine
| | - Siliang Wu
- Department of Materials Science and Engineering, New Jersey Institute of Technology
| | | | - Mary Bartlett Bunge
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine; Department of Cell Biology, University of Miami Miller School of Medicine; Department of Neurological Surgery, University of Miami Miller School of Medicine;
| |
Collapse
|
17
|
Schaal SM, Kitay BM, Cho KS, Lo TP, Barakat DJ, Marcillo AE, Sanchez AR, Andrade CM, Pearse DD. Schwann Cell Transplantation Improves Reticulospinal Axon Growth and Forelimb Strength after Severe Cervical Spinal Cord Contusion. Cell Transplant 2017; 16:207-28. [PMID: 17503734 DOI: 10.3727/000000007783464768] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Schwann cell (SC) implantation alone has been shown to promote the growth of propriospinal and sensory axons, but not long-tract descending axons, after thoracic spinal cord injury (SCI). In the current study, we examined if an axotomy close to the cell body of origin (so as to enhance the intrinsic growth response) could permit supraspinal axons to grow onto SC grafts. Adult female Fischer rats received a severe (C5) cervical contusion (1.1 mm displacement, 3 KDyn). At 1 week postinjury, 2 million SCs ex vivo transduced with lentiviral vector encoding enhanced green fluorescent protein (EGFP) were implanted within media into the injury epicenter; injury-only animals served as controls. Animals were tested weekly using the BBB score for 7 weeks postimplantation and received at end point tests for upper body strength: self-supported forelimb hanging, forearm grip force, and the incline plane. Following behavioral assessment, animals were anterogradely traced bilaterally from the reticular formation using BDA-Texas Red. Stereological quantification revealed a twofold increase in the numbers of preserved NeuN+ neurons rostral and caudal to the injury/graft site in SC implanted animals, corroborating previous reports of their neuroprotective efficacy. Examination of labeled reticulospinal axon growth revealed that while rarely an axon was present within the lesion site of injury-only controls, numerous reticulospinal axons had penetrated the SC implant/lesion milieu. This has not been observed following implantation of SCs alone into the injured thoracic spinal cord. Significant behavioral improvements over injury-only controls in upper limb strength, including an enhanced grip strength (a 296% increase) and an increased self-supported forelimb hanging, accompanied SC-mediated neuroprotection and reticulospinal axon growth. The current study further supports the neuroprotective efficacy of SC implants after SCI and demonstrates that SCs alone are capable of supporting modest supraspinal axon growth when the site of axon injury is closer to the cell body of the axotomized neuron.
Collapse
Affiliation(s)
- S M Schaal
- The Miami Project to Cure Paralysis, University of Miami School of Medicine, Miami, FL 33101, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Nandoe Tewarie RDS, Nandoe RDS, Hurtado A, Levi ADO, Grotenhuis JA, Grotenhuis A, Oudega M. Bone Marrow Stromal Cells for Repair of the Spinal Cord: Towards Clinical Application. Cell Transplant 2017; 15:563-77. [PMID: 17176609 DOI: 10.3727/000000006783981602] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Stem cells have been recognized and intensively studied for their potential use in restorative approaches for degenerative diseases and traumatic injuries. In the central nervous system (CNS), stem cell-based strategies have been proposed to replace lost neurons in degenerative diseases such as Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis (Lou Gehrig's disease), or to replace lost oligodendrocytes in demyelinating diseases such as multiple sclerosis. Stem cells have also been implicated in repair of the adult spinal cord. An impact to the spinal cord results in immediate damage to tissue including blood vessels, causing loss of neurons, astrocytes, and oligodendrocytes. In time, more tissue nearby or away from the injury site is lost due to secondary injury. In case of relatively minor damage to the cord some return of function can be observed, but in most cases the neurological loss is permanent. This review will focus on in vitro and in vivo studies on the use of bone marrow stromal cells (BMSCs), a heterogeneous cell population that includes mesenchymal stem cells, for repair of the spinal cord in experimental injury models and their potential for human application. To optimally benefit from BMSCs for repair of the spinal cord it is imperative to develop in vitro techniques that will generate the desired cell type and/or a large enough number for in vivo transplantation approaches. We will also assess the potential and possible pitfalls for use of BMSCs in humans and ongoing clinical trials.
Collapse
Affiliation(s)
- Rishi D S Nandoe Tewarie
- The Miami Project to Cure Paralysis, University of Miami, School of Medicine, Miami, FL 33136, USA
| | | | | | | | | | | | | |
Collapse
|
19
|
Abstract
During vertebrate embryonic development, the spinal cord is formed by the neural derivatives of a neuromesodermal population that is specified at early stages of development and which develops in concert with the caudal regression of the primitive streak. Several processes related to spinal cord specification and maturation are coupled to this caudal extension including neurogenesis, ventral patterning and neural crest specification and all of them seem to be crucially regulated by Fibroblast Growth Factor (FGF) signaling, which is prominently active in the neuromesodermal region and transiently in its derivatives. Here we review the role of FGF signaling in those processes, trying to separate its different functions and highlighting the interactions with other signaling pathways. Finally, these early functions of FGF signaling in spinal cord development may underlay partly its ability to promote regeneration in the lesioned spinal cord as well as its action promoting specific fates in neural stem cell cultures that may be used for therapeutical purposes.
Collapse
Affiliation(s)
- Ruth Diez Del Corral
- Department of Cellular, Molecular and Developmental Neurobiology, Cajal Institute, Consejo Superior de Investigaciones CientíficasMadrid, Spain.,Champalimaud Research, Champalimaud Centre for the UnknownLisbon, Portugal
| | - Aixa V Morales
- Department of Cellular, Molecular and Developmental Neurobiology, Cajal Institute, Consejo Superior de Investigaciones CientíficasMadrid, Spain
| |
Collapse
|
20
|
The Function of FGFR1 Signalling in the Spinal Cord: Therapeutic Approaches Using FGFR1 Ligands after Spinal Cord Injury. Neural Plast 2017; 2017:2740768. [PMID: 28197342 PMCID: PMC5286530 DOI: 10.1155/2017/2740768] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 12/25/2016] [Indexed: 11/24/2022] Open
Abstract
Extensive research is ongoing that concentrates on finding therapies to enhance CNS regeneration after spinal cord injury (SCI) and to cure paralysis. This review sheds light on the role of the FGFR pathway in the injured spinal cord and discusses various therapies that use FGFR activating ligands to promote regeneration after SCI. We discuss studies that use peripheral nerve grafts or Schwann cell grafts in combination with FGF1 or FGF2 supplementation. Most of these studies show evidence that these therapies successfully enhance axon regeneration into the graft. Further they provide evidence for partial recovery of sensory function shown by electrophysiology and motor activity evidenced by behavioural data. We also present one study that indicates that combination with additional, synergistic factors might further drive the system towards functional regeneration. In essence, this review summarises the potential of nerve and cell grafts combined with FGF1/2 supplementation to improve outcome even after severe spinal cord injury.
Collapse
|
21
|
Hodgetts SI, Harvey AR. Neurotrophic Factors Used to Treat Spinal Cord Injury. VITAMINS AND HORMONES 2016; 104:405-457. [PMID: 28215303 DOI: 10.1016/bs.vh.2016.11.007] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The application of neurotrophic factors as a therapy to improve morphological and behavioral outcomes after experimental spinal cord injury (SCI) has been the focus of many studies. These studies vary markedly in the type of neurotrophic factor that is delivered, the mode of administration, and the location, timing, and duration of the treatment. Generally, the majority of studies have had significant success if neurotrophic factors are applied in or close to the lesion site during the acute or the subacute phase after SCI. Comparatively fewer studies have administered neurotrophic factors in order to directly target the somata of injured neurons. The mode of delivery varies between acute injection of recombinant proteins, subacute or chronic delivery using a variety of strategies including osmotic minipumps, cell-mediated delivery, delivery using polymer release vehicles or supporting bridges of some sort, or the use of gene therapy to modify neurons, glial cells, or precursor/stem cells. In this brief review, we summarize the state of play of many of the therapies using these factors, most of which have been undertaken in rodent models of SCI.
Collapse
Affiliation(s)
- S I Hodgetts
- School of Anatomy, Physiology and Human Biology, The University of Western Australia, Perth, WA, Australia; Western Australian Neuroscience Research Institute, Perth, WA, Australia.
| | - A R Harvey
- School of Anatomy, Physiology and Human Biology, The University of Western Australia, Perth, WA, Australia; Western Australian Neuroscience Research Institute, Perth, WA, Australia
| |
Collapse
|
22
|
Lee YS, Wu S, Arinzeh TL, Bunge MB. Enhanced noradrenergic axon regeneration into schwann cell-filled PVDF-TrFE conduits after complete spinal cord transection. Biotechnol Bioeng 2016; 114:444-456. [PMID: 27570167 DOI: 10.1002/bit.26088] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 07/08/2016] [Accepted: 08/23/2016] [Indexed: 12/12/2022]
Abstract
Schwann cell (SC) transplantation has been utilized for spinal cord repair and demonstrated to be a promising therapeutic strategy. In this study, we investigated the feasibility of combining SC transplantation with novel conduits to bridge the completely transected adult rat spinal cord. This is the first and initial study to evaluate the potential of using a fibrous piezoelectric polyvinylidene fluoride trifluoroethylene (PVDF-TrFE) conduit with SCs for spinal cord repair. PVDF-TrFE has been shown to enhance neurite growth in vitro and peripheral nerve repair in vivo. In this study, SCs adhered and proliferated when seeded onto PVDF-TrFE scaffolds in vitro. SCs and PVDF-TrFE conduits, consisting of random or aligned fibrous inner walls, were transplanted into transected rat spinal cords for 3 weeks to examine early repair. Glial fibrillary acidic protein (GFAP)+ astrocyte processes and GFP (green fluorescent protein)-SCs were interdigitated at both rostral and caudal spinal cord/SC transplant interfaces in both types of conduits, indicative of permissivity to axon growth. More noradrenergic/DβH+ (dopamine-beta-hydroxylase) brainstem axons regenerated across the transplant when greater numbers of GFAP+ astrocyte processes were present. Aligned conduits promoted extension of DβH+ axons and GFAP+ processes farther into the transplant than random conduits. Sensory CGRP+ (calcitonin gene-related peptide) axons were present at the caudal interface. Blood vessels formed throughout the transplant in both conduits. This study demonstrates that PVDF-TrFE conduits harboring SCs are promising for spinal cord repair and deserve further investigation. Biotechnol. Bioeng. 2017;114: 444-456. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Yee-Shuan Lee
- The Miami Project to Cure Paralysis, Lois Pope LIFE Center, University of Miami Miller School of Medicine, P.O. Box 016960, Mail locator R-48, Miami, Florida 33101
| | - Siliang Wu
- Department of Material Science and Engineering, New Jersey Institute of Technology, Newark, New Jersey
| | | | - Mary Bartlett Bunge
- The Miami Project to Cure Paralysis, Lois Pope LIFE Center, University of Miami Miller School of Medicine, P.O. Box 016960, Mail locator R-48, Miami, Florida 33101.,Department of Cell Biology, University of Miami Miller School of Medicine, Miami, Florida 33101.,Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida 33101
| |
Collapse
|
23
|
Rao SNR, Pearse DD. Regulating Axonal Responses to Injury: The Intersection between Signaling Pathways Involved in Axon Myelination and The Inhibition of Axon Regeneration. Front Mol Neurosci 2016; 9:33. [PMID: 27375427 PMCID: PMC4896923 DOI: 10.3389/fnmol.2016.00033] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 05/02/2016] [Indexed: 01/06/2023] Open
Abstract
Following spinal cord injury (SCI), a multitude of intrinsic and extrinsic factors adversely affect the gene programs that govern the expression of regeneration-associated genes (RAGs) and the production of a diversity of extracellular matrix molecules (ECM). Insufficient RAG expression in the injured neuron and the presence of inhibitory ECM at the lesion, leads to structural alterations in the axon that perturb the growth machinery, or form an extraneous barrier to axonal regeneration, respectively. Here, the role of myelin, both intact and debris, in antagonizing axon regeneration has been the focus of numerous investigations. These studies have employed antagonizing antibodies and knockout animals to examine how the growth cone of the re-growing axon responds to the presence of myelin and myelin-associated inhibitors (MAIs) within the lesion environment and caudal spinal cord. However, less attention has been placed on how the myelination of the axon after SCI, whether by endogenous glia or exogenously implanted glia, may alter axon regeneration. Here, we examine the intersection between intracellular signaling pathways in neurons and glia that are involved in axon myelination and axon growth, to provide greater insight into how interrogating this complex network of molecular interactions may lead to new therapeutics targeting SCI.
Collapse
Affiliation(s)
- Sudheendra N R Rao
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine Miami, FL, USA
| | - Damien D Pearse
- The Miami Project to Cure Paralysis, University of Miami Miller School of MedicineMiami, FL, USA; The Department of Neurological Surgery, University of Miami Miller School of MedicineMiami, FL, USA; The Neuroscience Program, University of Miami Miller School of MedicineMiami, FL, USA; The Interdisciplinary Stem Cell Institute, University of Miami Miller School of MedicineMiami, FL, USA; Bruce W. Carter Department of Veterans Affairs Medical CenterMiami, FL, USA
| |
Collapse
|
24
|
Acute Putrescine Supplementation with Schwann Cell Implantation Improves Sensory and Serotonergic Axon Growth and Functional Recovery in Spinal Cord Injured Rats. Neural Plast 2015; 2015:186385. [PMID: 26550496 PMCID: PMC4621347 DOI: 10.1155/2015/186385] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Revised: 06/25/2015] [Accepted: 07/02/2015] [Indexed: 01/29/2023] Open
Abstract
Schwann cell (SC) transplantation exhibits significant potential for spinal cord injury (SCI) repair and its use as a therapeutic modality has now progressed to clinical trials for subacute and chronic human SCI. Although SC implants provide a receptive environment for axonal regrowth and support functional recovery in a number of experimental SCI models, axonal regeneration is largely limited to local systems and the behavioral improvements are modest without additional combinatory approaches. In the current study we investigated whether the concurrent delivery of the polyamine putrescine, started either 30 min or 1 week after SCI, could enhance the efficacy of SCs when implanted subacutely (1 week after injury) into the contused rat spinal cord. Polyamines are ubiquitous organic cations that play an important role in the regulation of the cell cycle, cell division, cytoskeletal organization, and cell differentiation. We show that the combination of putrescine with SCs provides a significant increase in implant size, an enhancement in axonal (sensory and serotonergic) sparing and/or growth, and improved open field locomotion after SCI, as compared to SC implantation alone. These findings demonstrate that polyamine supplementation can augment the effectiveness of SCs when used as a therapeutic approach for subacute SCI repair.
Collapse
|
25
|
Siddiqui AM, Khazaei M, Fehlings MG. Translating mechanisms of neuroprotection, regeneration, and repair to treatment of spinal cord injury. PROGRESS IN BRAIN RESEARCH 2015; 218:15-54. [PMID: 25890131 DOI: 10.1016/bs.pbr.2014.12.007] [Citation(s) in RCA: 101] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
One of the big challenges in neuroscience that remains to be understood is why the central nervous system is not able to regenerate to the extent that the peripheral nervous system does. This is especially problematic after traumatic injuries, like spinal cord injury (SCI), since the lack of regeneration leads to lifelong deficits and paralysis. Treatment of SCI has improved during the last several decades due to standardized protocols for emergency medical response teams and improved medical, surgical, and rehabilitative treatments. However, SCI continues to result in profound impairments for the individual. There are many processes that lead to the pathophysiology of SCI, such as ischemia, vascular disruption, neuroinflammation, oxidative stress, excitotoxicity, demyelination, and cell death. Current treatments include surgical decompression, hemodynamic control, and methylprednisolone. However, these early treatments are associated with modest functional recovery. Some treatments currently being investigated for use in SCI target neuroprotective (riluzole, minocycline, G-CSF, FGF-2, and polyethylene glycol) or neuroregenerative (chondroitinase ABC, self-assembling peptides, and rho inhibition) strategies, while many cell therapies (embryonic stem cells, neural stem cells, induced pluripotent stem cells, mesenchymal stromal cells, Schwann cells, olfactory ensheathing cells, and macrophages) have also shown promise. However, since SCI has multiple factors that determine the progress of the injury, a combinatorial therapeutic approach will most likely be required for the most effective treatment of SCI.
Collapse
Affiliation(s)
- Ahad M Siddiqui
- Department of Genetics and Development, Toronto Western Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Mohamad Khazaei
- Department of Genetics and Development, Toronto Western Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Michael G Fehlings
- Department of Genetics and Development, Toronto Western Research Institute, University Health Network, Toronto, Ontario, Canada; Department of Surgery, University of Toronto, Toronto, Ontario, Canada; Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
26
|
Li L, Yang M, Wang C, Zhao Q, Liu J, Zhan C, Liu Z, Li X, Wang W, Yang X. Effects of cytokines and chemokines on migration of mesenchymal stem cells following spinal cord injury. Neural Regen Res 2015; 7:1106-12. [PMID: 25722702 PMCID: PMC4340025 DOI: 10.3969/j.issn.1673-5374.2012.14.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2012] [Accepted: 03/20/2012] [Indexed: 01/16/2023] Open
Abstract
We investigated the effects of cytokines and chemokines and their associated signaling pathways on mesenchymal stem cell migration after spinal cord injury, to determine their roles in the curative effects of mesenchymal stem cells. This study reviewed the effects of tumor necrosis factor-α, vascular endothelial growth factor, hepatocyte growth factor, platelet-derived growth factor, basic fibroblast growth factor, insulin like growth factor-1, stromal cell-derived factor and monocyte chemoattractant protein-1, 3 during mesenchymal stem cell migration to damaged sites, and analyzed the signal transduction pathways involved in their effects on mesenchymal stem cell migration. The results confirmed that phosphatidylinositol 3-kinase/serine/threonine protein kinases and nuclear factor-κB play crucial roles in the migration of mesenchymal stem cells induced by cytokines and chemokines.
Collapse
Affiliation(s)
- Longyun Li
- Department of Orthopedics, China-Japan Friendship Hospital, Jilin University, Changchun 130033, Jilin Province, China
| | - Maoguang Yang
- Department of Orthopedics, China-Japan Friendship Hospital, Jilin University, Changchun 130033, Jilin Province, China
| | - Chunxin Wang
- Department of Orthopedics, China-Japan Friendship Hospital, Jilin University, Changchun 130033, Jilin Province, China
| | - Qiheng Zhao
- Department of Orthopedics, China-Japan Friendship Hospital, Jilin University, Changchun 130033, Jilin Province, China
| | - Jian Liu
- Department of Orthopedics, China-Japan Friendship Hospital, Jilin University, Changchun 130033, Jilin Province, China
| | - Chuanguo Zhan
- Department of Orthopedics, China-Japan Friendship Hospital, Jilin University, Changchun 130033, Jilin Province, China
| | - Zhi Liu
- Department of Orthopedics, China-Japan Friendship Hospital, Jilin University, Changchun 130033, Jilin Province, China
| | - Xuepeng Li
- Department of Orthopedics, China-Japan Friendship Hospital, Jilin University, Changchun 130033, Jilin Province, China
| | - Weihua Wang
- Department of Orthopedics, China-Japan Friendship Hospital, Jilin University, Changchun 130033, Jilin Province, China
| | - Xiaoyu Yang
- Department of Orthopedics, China-Japan Friendship Hospital, Jilin University, Changchun 130033, Jilin Province, China
| |
Collapse
|
27
|
Kaneko A, Matsushita A, Sankai Y. A 3D nanofibrous hydrogel and collagen sponge scaffold promotes locomotor functional recovery, spinal repair, and neuronal regeneration after complete transection of the spinal cord in adult rats. Biomed Mater 2015; 10:015008. [DOI: 10.1088/1748-6041/10/1/015008] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
28
|
Murayama N, Noshita T, Ogino R, Masuda T, Kadoshima T, Oka T, Ueno N, Takemoto N, Toba T, Ueno S, Schulze W, Igawa Y, Morita Y, Inoue T. SUN11602-induced hyperexpression of calbindin D-28k is pivotal for the survival of hippocampal neurons under neurotoxic conditions. Brain Res 2015; 1594:71-81. [DOI: 10.1016/j.brainres.2014.10.066] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Revised: 09/12/2014] [Accepted: 10/29/2014] [Indexed: 02/03/2023]
|
29
|
Liu J, Chen P, Wang Q, Chen Y, Yu H, Ma J, Guo M, Piao M, Ren W, Xiang L. Meta analysis of olfactory ensheathing cell transplantation promoting functional recovery of motor nerves in rats with complete spinal cord transection. Neural Regen Res 2014; 9:1850-8. [PMID: 25422649 PMCID: PMC4239777 DOI: 10.4103/1673-5374.143434] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/20/2014] [Indexed: 01/22/2023] Open
Abstract
OBJECTIVE: To evaluate the effects of olfactory ensheathing cell transplantation on functional recovery of rats with complete spinal cord transection. DATA SOURCES: A computer-based online search of Medline (1989–2013), Embase (1989–2013), Cochrane library (1989–2013), Chinese Biomedical Literature Database (1989–2013), China National Knowledge Infrastructure (1989–2013), VIP (1989–2013), Wanfang databases (1989–2013) and Chinese Clinical Trial Register was conducted to collect randomized controlled trial data regarding olfactory ensheathing cell transplantation for the treatment of complete spinal cord transection in rats. SELECTION CRITERIA: Randomized controlled trials investigating olfactory ensheathing cell transplantation and other transplantation methods for promoting neurological functional recovery of rats with complete spinal cord transection were included in the analysis. Meta analysis was conducted using RevMan 4.2.2 software. MAIN OUTCOME MEASURES: Basso, Beattie and Bresnahan scores of rats with complete spinal cord transection were evaluated in this study. RESULTS: Six randomized controlled trials with high quality methodology were included. Meta analysis showed that Basso, Beattie and Bresnahan scores were significantly higher in the olfactory ensheathing cell transplantation group compared with the control group (WMD = 3.16, 95% CI (1.68, 4.65); P < 0.00001). CONCLUSION: Experimental studies have shown that olfactory ensheathing cell transplantation can promote the functional recovery of motor nerves in rats with complete spinal cord transection.
Collapse
Affiliation(s)
- Jun Liu
- Department of Orthopedics, General Hospital of Shenyang Military Command Area of Chinese PLA, Shenyang, Liaoning Province, China
| | - Ping Chen
- Department of Gastroenterology, Taian Central Hospital, Taian, Shandong Province, China
| | - Qi Wang
- Department of Orthopedics, General Hospital of Shenyang Military Command Area of Chinese PLA, Shenyang, Liaoning Province, China
| | - Yu Chen
- Department of Orthopedics, General Hospital of Shenyang Military Command Area of Chinese PLA, Shenyang, Liaoning Province, China
| | - Haiong Yu
- Department of Orthopedics, General Hospital of Shenyang Military Command Area of Chinese PLA, Shenyang, Liaoning Province, China
| | - Junxiong Ma
- Department of Orthopedics, General Hospital of Shenyang Military Command Area of Chinese PLA, Shenyang, Liaoning Province, China
| | - Mingming Guo
- Department of Orthopedics, General Hospital of Shenyang Military Command Area of Chinese PLA, Shenyang, Liaoning Province, China
| | - Meihui Piao
- Department of Orthopedics, General Hospital of Shenyang Military Command Area of Chinese PLA, Shenyang, Liaoning Province, China
| | - Weijian Ren
- Department of Orthopedics, General Hospital of Shenyang Military Command Area of Chinese PLA, Shenyang, Liaoning Province, China
| | - Liangbi Xiang
- Department of Orthopedics, General Hospital of Shenyang Military Command Area of Chinese PLA, Shenyang, Liaoning Province, China
| |
Collapse
|
30
|
Harvey AR, Lovett SJ, Majda BT, Yoon JH, Wheeler LPG, Hodgetts SI. Neurotrophic factors for spinal cord repair: Which, where, how and when to apply, and for what period of time? Brain Res 2014; 1619:36-71. [PMID: 25451132 DOI: 10.1016/j.brainres.2014.10.049] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Revised: 10/20/2014] [Accepted: 10/23/2014] [Indexed: 12/22/2022]
Abstract
A variety of neurotrophic factors have been used in attempts to improve morphological and behavioural outcomes after experimental spinal cord injury (SCI). Here we review many of these factors, their cellular targets, and their therapeutic impact on spinal cord repair in different, primarily rodent, models of SCI. A majority of studies report favourable outcomes but results are by no means consistent, thus a major aim of this review is to consider how best to apply neurotrophic factors after SCI to optimize their therapeutic potential. In addition to which factors are chosen, many variables need be considered when delivering trophic support, including where and when to apply a given factor or factors, how such factors are administered, at what dose, and for how long. Overall, the majority of studies have applied neurotrophic support in or close to the spinal cord lesion site, in the acute or sub-acute phase (0-14 days post-injury). Far fewer chronic SCI studies have been undertaken. In addition, comparatively fewer studies have administered neurotrophic factors directly to the cell bodies of injured neurons; yet in other instructive rodent models of CNS injury, for example optic nerve crush or transection, therapies are targeted directly at the injured neurons themselves, the retinal ganglion cells. The mode of delivery of neurotrophic factors is also an important variable, whether delivered by acute injection of recombinant proteins, sub-acute or chronic delivery using osmotic minipumps, cell-mediated delivery, delivery using polymer release vehicles or supporting bridges of some sort, or the use of gene therapy to modify neurons, glial cells or precursor/stem cells. Neurotrophic factors are often used in combination with cell or tissue grafts and/or other pharmacotherapeutic agents. Finally, the dose and time-course of delivery of trophic support should ideally be tailored to suit specific biological requirements, whether they relate to neuronal survival, axonal sparing/sprouting, or the long-distance regeneration of axons ending in a different mode of growth associated with terminal arborization and renewed synaptogenesis. This article is part of a Special Issue entitled SI: Spinal cord injury.
Collapse
Affiliation(s)
- Alan R Harvey
- School of Anatomy, Physiology and Human Biology, The University of Western Australia, 35 Stirling Highway, Crawley, WA 6009, Australia.
| | - Sarah J Lovett
- School of Anatomy, Physiology and Human Biology, The University of Western Australia, 35 Stirling Highway, Crawley, WA 6009, Australia
| | - Bernadette T Majda
- School of Anatomy, Physiology and Human Biology, The University of Western Australia, 35 Stirling Highway, Crawley, WA 6009, Australia
| | - Jun H Yoon
- School of Anatomy, Physiology and Human Biology, The University of Western Australia, 35 Stirling Highway, Crawley, WA 6009, Australia
| | - Lachlan P G Wheeler
- School of Anatomy, Physiology and Human Biology, The University of Western Australia, 35 Stirling Highway, Crawley, WA 6009, Australia
| | - Stuart I Hodgetts
- School of Anatomy, Physiology and Human Biology, The University of Western Australia, 35 Stirling Highway, Crawley, WA 6009, Australia
| |
Collapse
|
31
|
Goldshmit Y, Frisca F, Pinto AR, Pébay A, Tang JKKY, Siegel AL, Kaslin J, Currie PD. Fgf2 improves functional recovery-decreasing gliosis and increasing radial glia and neural progenitor cells after spinal cord injury. Brain Behav 2014; 4:187-200. [PMID: 24683512 PMCID: PMC3967535 DOI: 10.1002/brb3.172] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Accepted: 07/30/2013] [Indexed: 01/26/2023] Open
Abstract
OBJECTIVES A major impediment for recovery after mammalian spinal cord injury (SCI) is the glial scar formed by proliferating reactive astrocytes. Finding factors that may reduce glial scarring, increase neuronal survival, and promote neurite outgrowth are of major importance for improving the outcome after SCI. Exogenous fibroblast growth factor (Fgf) has been shown to decrease injury volume and improve functional outcome; however, the mechanisms by which this is mediated are still largely unknown. METHODS In this study, Fgf2 was administered for 2 weeks in mice subcutaneously, starting 30 min after spinal cord hemisection. RESULTS Fgf2 treatment decreased the expression of TNF-a at the lesion site, decreased monocyte/macrophage infiltration, and decreased gliosis. Fgf2 induced astrocytes to adopt a polarized morphology and increased expression of radial markers such as Pax6 and nestin. In addition, the levels of chondroitin sulfate proteoglycans (CSPGs), expressed by glia, were markedly decreased. Furthermore, Fgf2 treatment promotes the formation of parallel glial processes, "bridges," at the lesion site that enable regenerating axons through the injury site. Additionally, Fgf2 treatment increased Sox2-expressing cells in the gray matter and neurogenesis around and at the lesion site. Importantly, these effects were correlated with enhanced functional recovery of the left paretic hind limb. CONCLUSIONS Thus, early pharmacological intervention with Fgf2 following SCI is neuroprotective and creates a proregenerative environment by the modulation of the glia response.
Collapse
Affiliation(s)
- Yona Goldshmit
- Australian Regenerative Medicine Institute East Melbourne, VIC, Australia ; Centre for Eye Research Australia & Royal Victorian Eye and Ear Hospital East Melbourne, VIC, Australia
| | - Frisca Frisca
- Department of Ophthalmology, The University of Melbourne East Melbourne, VIC, Australia
| | - Alexander R Pinto
- Australian Regenerative Medicine Institute East Melbourne, VIC, Australia
| | - Alice Pébay
- Centre for Eye Research Australia & Royal Victorian Eye and Ear Hospital East Melbourne, VIC, Australia ; Department of Ophthalmology, The University of Melbourne East Melbourne, VIC, Australia
| | | | - Ashley L Siegel
- Australian Regenerative Medicine Institute East Melbourne, VIC, Australia
| | - Jan Kaslin
- Australian Regenerative Medicine Institute East Melbourne, VIC, Australia
| | - Peter D Currie
- Australian Regenerative Medicine Institute East Melbourne, VIC, Australia
| |
Collapse
|
32
|
Co-transplantation of bone marrow-derived mesenchymal stem cells and nanospheres containing FGF-2 improve cell survival and neurological function in the injured rat spinal cord. Acta Neurochir (Wien) 2014; 156:297-303. [PMID: 24352373 DOI: 10.1007/s00701-013-1963-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Accepted: 11/28/2013] [Indexed: 12/11/2022]
Abstract
BACKGROUND Spinal cord injury (SCI) is a devastating and irreversible event, and much research using fibroblast growth factor-2 (FGF-2) has been performed to test its capacity to blunt the effects of SCI as well as to provide an environment conducive for SCI repair. METHODS We tested how the in vitro release of FGF-2 from heparin-conjugated poly(L-lactide-co-glycolide) (PLGA)-conjugated nanospheres (HCPNs) affected the growth of human bone marrow-derived mesenchymal stem cells (hBMSCs), as well as the effects of their co-transplantation in an animal model of SCI. RESULTS Our results showed that sustained, long-term release of FGF-2 from HCPNs significantly increased hBMSCs proliferation in vitro, and that their co-transplantation following rat SCI lead to increased functional improvement, a greater amount of hBMSCs surviving transplantation, and a greater density of neurofilament-positive cells in the injury epicenter. CONCLUSION These results suggest a proliferative, protective, and neural inductive potential of FGF-2 for transplanted hBMSCs, as well as a possible role for sustained FGF-2 delivery along with hBMSCs transplantation in the injured spinal cord. Future studies will be required to ascertain the safety FGF-2-containing HCPNs before clinical application.
Collapse
|
33
|
Kanno H, Pressman Y, Moody A, Berg R, Muir EM, Rogers JH, Ozawa H, Itoi E, Pearse DD, Bunge MB. Combination of engineered Schwann cell grafts to secrete neurotrophin and chondroitinase promotes axonal regeneration and locomotion after spinal cord injury. J Neurosci 2014; 34:1838-55. [PMID: 24478364 PMCID: PMC3905147 DOI: 10.1523/jneurosci.2661-13.2014] [Citation(s) in RCA: 126] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2013] [Revised: 11/14/2013] [Accepted: 12/19/2013] [Indexed: 11/21/2022] Open
Abstract
Transplantation of Schwann cells (SCs) is a promising therapeutic strategy for spinal cord repair. SCs introduced into lesions support axon regeneration, but because these axons do not exit the transplant, additional approaches with SCs are needed. Here, we transplanted SCs genetically modified to secrete a bifunctional neurotrophin (D15A) and chondroitinase ABC (ChABC) into a subacute contusion injury in rats. We examined the effects of these modifications on graft volume, SC number, degradation of chondroitin sulfate proteoglycans (CSPGs), astrogliosis, SC myelination of axons, propriospinal and supraspinal axon numbers, locomotor outcome (BBB scoring, CatWalk gait analysis), and mechanical and thermal sensitivity on the hind paws. D15A secreted from transplanted SCs increased graft volume and SC number and myelinated axon number. SCs secreting ChABC significantly decreased CSPGs, led to some egress of SCs from the graft, and increased propriospinal and 5-HT-positive axons in the graft. SCs secreting both D15A and ChABC yielded the best responses: (1) the largest number of SC myelinated axons, (2) more propriospinal axons in the graft and host tissue around and caudal to it, (3) more corticospinal axons closer to the graft and around and caudal to it, (4) more brainstem neurons projecting caudal to the transplant, (5) increased 5-HT-positive axons in the graft and caudal to it, (6) significant improvement in aspects of locomotion, and (7) improvement in mechanical and thermal allodynia. This is the first evidence that the combination of SC transplants engineered to secrete neurotrophin and chondroitinase further improves axonal regeneration and locomotor and sensory function.
Collapse
Affiliation(s)
- Haruo Kanno
- Miami Project to Cure Paralysis
- Department of Orthopaedic Surgery, Tohoku University School of Medicine, Sendai, Japan, 9808574
| | | | | | | | - Elizabeth M. Muir
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, CB2 3EG, United Kingdom, and
| | - John H. Rogers
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, CB2 3EG, United Kingdom, and
| | - Hiroshi Ozawa
- Department of Orthopaedic Surgery, Tohoku University School of Medicine, Sendai, Japan, 9808574
| | - Eiji Itoi
- Department of Orthopaedic Surgery, Tohoku University School of Medicine, Sendai, Japan, 9808574
| | - Damien D. Pearse
- Miami Project to Cure Paralysis
- Department of Neurological Surgery
- Neuroscience Program
- Interdisciplinary Stem Cell Institute, and
| | - Mary Bartlett Bunge
- Miami Project to Cure Paralysis
- Department of Neurological Surgery
- Neuroscience Program
- Interdisciplinary Stem Cell Institute, and
- Department of Cell Biology, University of Miami Miller School of Medicine, Miami, Florida 33136
| |
Collapse
|
34
|
Flora G, Joseph G, Patel S, Singh A, Bleicher D, Barakat DJ, Louro J, Fenton S, Garg M, Bunge MB, Pearse DD. Combining Neurotrophin-Transduced Schwann Cells and Rolipram to Promote Functional Recovery from Subacute Spinal Cord Injury. Cell Transplant 2013; 22:2203-17. [DOI: 10.3727/096368912x658872] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Following spinal cord injury (SCI), both an inhibitory environment and lack of intrinsic growth capacity impede axonal regeneration. In a previous study, prevention of cyclic adenosine monophosphate (AMP) hydrolysis by the phosphodiesterase-4 inhibitor rolipram, in combination with Schwann cell (SC) grafts, promoted significant supraspinal and proprioceptive fiber growth and/or sparing and improved locomotion. In another study, transplanted SCs transduced to generate a bifunctional neurotrophin (D15A) led to significant increases in graft SCs and axons, including supraspinal and myelinated axons. Here we studied the growth and myelination of local and supraspinal axons and functional outcome following the combination of rolipram administration and neurotrophin-transduced SC implantation after SCI. Rolipram was administered subcutaneously for 4 weeks immediately after contusion at vertebral T8 (25.0-mm weight drop, MASCIS impactor). GFP or GFP-D15A-transduced SCs were injected into the injury epicenter 1 week after SCI. GFP-D15A SC grafts and GFP SC grafts with rolipram contained significantly more serotonergic fibers compared to GFP SCs. SC myelinated axons were increased significantly in GFP SC with rolipram-treated animals compared to animals receiving SCI alone. Rolipram administered with either GFP or GFP-D15A SCs significantly increased numbers of brain stem-derived axons below the lesion/implant area and improved hindlimb function. Compared to the single treatments, the combination led to the largest SC grafts, the highest numbers of serotonergic fibers in the grafts, and increased numbers of axons from the reticular formation below the lesion/implant area and provided the greatest improvement in hindlimb function. These findings demonstrate the therapeutic potential for a combination therapy involving the maintenance of cyclic AMP levels and neurotrophin-transduced SCs to repair the subacutely injured spinal cord.
Collapse
Affiliation(s)
- Govinder Flora
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, USA
- The Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Gravil Joseph
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, USA
- The Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Samik Patel
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, USA
- The Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Amanpreet Singh
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, USA
- The Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Drew Bleicher
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, USA
- The Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL, USA
| | - David J. Barakat
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, USA
- The Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Jack Louro
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, USA
- The Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Stephanie Fenton
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, USA
- The Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Maneesh Garg
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, USA
- The Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Mary Bartlett Bunge
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, USA
- The Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Cell Biology, University of Miami Miller School of Medicine, Miami, FL, USA
- The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Damien D. Pearse
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, USA
- The Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
- The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
35
|
Williams RR, Henao M, Pearse DD, Bunge MB. Permissive Schwann cell graft/spinal cord interfaces for axon regeneration. Cell Transplant 2013; 24:115-31. [PMID: 24152553 DOI: 10.3727/096368913x674657] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
The transplantation of autologous Schwann cells (SCs) to repair the injured spinal cord is currently being evaluated in a clinical trial. In support, this study determined properties of spinal cord/SC bridge interfaces that enabled regenerated brainstem axons to cross them, possibly leading to improvement in rat hindlimb movement. Fluid bridges of SCs and Matrigel were placed in complete spinal cord transections. Compared to pregelled bridges of SCs and Matrigel, they improved regeneration of brainstem axons across the rostral interface. The regenerating brainstem axons formed synaptophysin(+) bouton-like terminals and contacted MAP2A(+) dendrites at the caudal interface. Brainstem axon regeneration was directly associated with glial fibrillary acidic protein (GFAP(+)) astrocyte processes that elongated into the SC bridge. Electron microscopy revealed that axons, SCs, and astrocytes were enclosed together within tunnels bounded by a continuous basal lamina. Neuroglycan (NG2) expression was associated with these tunnels. One week after injury, the GFAP(+) processes coexpressed nestin and brain lipid-binding protein, and the tips of GFAP(+)/NG2(+) processes extended into the bridges together with the regenerating brainstem axons. Both brainstem axon regeneration and number of GFAP(+) processes in the bridges correlated with improvement in hindlimb locomotion. Following SCI, astrocytes may enter a reactive state that prohibits axon regeneration. Elongation of astrocyte processes into SC bridges, however, and formation of NG2(+) tunnels enable brainstem axon regeneration and improvement in function. It is important for spinal cord repair to define conditions that favor elongation of astrocytes into lesions/transplants.
Collapse
Affiliation(s)
- Ryan R Williams
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, USA
| | | | | | | |
Collapse
|
36
|
Berrocal YA, Almeida VW, Gupta R, Levi AD. Transplantation of Schwann cells in a collagen tube for the repair of large, segmental peripheral nerve defects in rats. J Neurosurg 2013; 119:720-32. [DOI: 10.3171/2013.4.jns121189] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Object
Segmental nerve defects pose a daunting clinical challenge, as peripheral nerve injury studies have established that there is a critical nerve gap length for which the distance cannot be successfully bridged with current techniques. Construction of a neural prosthesis filled with Schwann cells (SCs) could provide an alternative treatment to successfully repair these long segmental gaps in the peripheral nervous system. The object of this study was to evaluate the ability of autologous SCs to increase the length at which segmental nerve defects can be bridged using a collagen tube.
Methods
The authors studied the use of absorbable collagen conduits in combination with autologous SCs (200,000 cells/μl) to promote axonal growth across a critical size defect (13 mm) in the sciatic nerve of male Fischer rats. Control groups were treated with serum only–filled conduits of reversed sciatic nerve autografts. Animals were assessed for survival of the transplanted SCs as well as the quantity of myelinated axons in the proximal, middle, and distal portions of the channel.
Results
Schwann cell survival was confirmed at 4 and 16 weeks postsurgery by the presence of prelabeled green fluorescent protein–positive SCs within the regenerated cable. The addition of SCs to the nerve guide significantly enhanced the regeneration of myelinated axons from the nerve stump into the proximal (p < 0.001) and middle points (p < 0.01) of the tube at 4 weeks. The regeneration of myelinated axons at 16 weeks was significantly enhanced throughout the entire length of the nerve guide (p < 0.001) as compared with their number in a serum–only filled tube and was similar in number compared with the reversed autograft. Autotomy scores were significantly lower in the animals whose sciatic nerve was repaired with a collagen conduit either without (p < 0.01) or with SCs (p < 0.001) when compared with a reversed autograft.
Conclusions
The technique of adding SCs to a guidance channel significantly enhanced the gap distance that can be repaired after peripheral nerve injury with long segmental defects and holds promise in humans. Most importantly, this study represents some of the first essential steps in bringing autologous SC-based therapies to the domain of peripheral nerve injuries with long segmental defects.
Collapse
Affiliation(s)
- Yerko A. Berrocal
- 1The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida; and
| | - Vania W. Almeida
- 1The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida; and
| | - Ranjan Gupta
- 2Department of Orthopedic Surgery, University of California–Irvine, California
| | - Allan D. Levi
- 1The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida; and
| |
Collapse
|
37
|
Sakiyama-Elbert S, Johnson PJ, Hodgetts SI, Plant GW, Harvey AR. Scaffolds to promote spinal cord regeneration. HANDBOOK OF CLINICAL NEUROLOGY 2013; 109:575-94. [PMID: 23098738 DOI: 10.1016/b978-0-444-52137-8.00036-x] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Substantial research effort in the spinal cord injury (SCI) field is directed towards reduction of secondary injury changes and enhancement of tissue sparing. However, pathway repair after complete transections, large lesions, or after chronic injury may require the implantation of some form of oriented bridging structure to restore tissue continuity across a trauma zone. These matrices or scaffolds should be biocompatible and create an environment that facilitates tissue growth and vascularization, and allow axons to regenerate through and beyond the implant in order to reconnect with "normal" tissue distal to the injury. The myelination of regrown axons is another important requirement. In this chapter, we describe recent advances in biomaterial technology designed to provide a terrain for regenerating axons to grow across the site of injury and/or create an environment for endogenous repair. Many different types of scaffold are under investigation; they can be biodegradable or nondegradable, natural or synthetic. Scaffolds can be designed to incorporate immobilized signaling molecules and/or used as devices for controlled release of therapeutic agents, including growth factors. These bridging structures can also be infiltrated with specific cell types deemed suitable for spinal cord repair.
Collapse
Affiliation(s)
- S Sakiyama-Elbert
- Department of Biomedical Engineering, Washington University, St. Louis, MO, USA
| | | | | | | | | |
Collapse
|
38
|
Krishna V, Konakondla S, Nicholas J, Varma A, Kindy M, Wen X. Biomaterial-based interventions for neuronal regeneration and functional recovery in rodent model of spinal cord injury: a systematic review. J Spinal Cord Med 2013; 36:174-90. [PMID: 23809587 PMCID: PMC3654443 DOI: 10.1179/2045772313y.0000000095] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
CONTEXT There is considerable interest in translating laboratory advances in neuronal regeneration following spinal cord injury (SCI). A multimodality approach has been advocated for successful functional neuronal regeneration. With this goal in mind several biomaterials have been employed as neuronal bridges either to support cellular transplants, to release neurotrophic factors, or to do both. A systematic review of this literature is lacking. Such a review may provide insight to strategies with a high potential for further investigation and potential clinical application. OBJECTIVE To systematically review the design strategies and outcomes after biomaterial-based multimodal interventions for neuronal regeneration in rodent SCI model. To analyse functional outcomes after implantation of biomaterial-based multimodal interventions and to identify predictors of functional outcomes. METHODS A broad PubMed, CINHAL, and a manual search of relevant literature databases yielded data from 24 publications; 14 of these articles included functional outcome information. Studies reporting behavioral data in rat model of SCI and employing biodegradable polymer-based multimodal intervention were included. For behavioral recovery, studies using severe injury models (transection or severe clip compression (>16.9 g) or contusion (50 g/cm)) were categorized separately from those investigating partial injury models (hemisection or moderate-to-severe clip compression or contusion). RESULTS The cumulative mean improvements in Basso, Beattie, and Bresnahan scores after biomaterial-based interventions are 5.93 (95% CI = 2.41 - 9.45) and 4.44 (95% CI = 2.65 - 6.24) for transection and hemisection models, respectively. Factors associated with improved outcomes include the type of polymer used and a follow-up period greater than 6 weeks. CONCLUSION The functional improvement after implantation of biopolymer-based multimodal implants is modest. The relationship with neuronal regeneration and functional outcome, the effects of inflammation at the site of injury, the prolonged survival of supporting cells, the differentiation of stem cells, the effective delivery of neurotrophic factors, and longer follow-up periods are all topics for future elucidation. Future investigations should strive to further define specific factors associated with improved functional outcomes in clinically relevant models.
Collapse
Affiliation(s)
- Vibhor Krishna
- Medical University of South Carolina, Charleston, SC, USA.
| | | | - Joyce Nicholas
- Medical University of South Carolina, Charleston, SC, USA
| | - Abhay Varma
- Medical University of South Carolina, Charleston, SC, USA
| | - Mark Kindy
- Medical University of South Carolina, Charleston, SC, USA
| | - Xuejun Wen
- Medical University of South Carolina, Charleston, SC, USA; and Department of Bioengineering, Clemson University, SC, USA
| |
Collapse
|
39
|
Hill CE, Brodak DM, Bartlett Bunge M. Dissociated predegenerated peripheral nerve transplants for spinal cord injury repair: a comprehensive assessment of their effects on regeneration and functional recovery compared to Schwann cell transplants. J Neurotrauma 2012; 29:2226-43. [PMID: 22655857 DOI: 10.1089/neu.2012.2377] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Several recent studies suggest that predegenerated nerves (PDNs) or dissociated PDNs (dPDNs) can improve behavioral and histological outcomes following transplantation into the injured rat spinal cord. In the current study we tested the efficacy of dPDN transplantation by grafting cells isolated from the sciatic nerve 7 days after crush. We did not replicate one study, but rather assessed what appeared, based on five published reports, to be a reported robust effect of dPDN grafts on corticospinal tract (CST) regeneration and locomotor recovery. Using a standardized rodent spinal cord injury model (200 kD IH contusion) and transplantation procedure (injection of GFP⁺ cells 7 days post-SCI), we demonstrate that dPDN grafts survive within the injured spinal cord and promote the ingrowth of axons to a similar extent as purified Schwann cell (SC) grafts. We also demonstrate for the first time that while both dPDN and SC grafts promote the ingrowth of CGRP axons, neither graft results in mechanical or thermal hyperalgesia. Unlike previous studies, dPDN grafts did not promote long-distance axonal growth of CST axons, brainstem spinal axons, or ascending dorsal column sensory axons. Moreover, using a battery of locomotor tests (Basso Beattie Bresnahan [BBB] score, BBB subscore, inked footprint, Catwalk, and ladderwalk), we failed to detect any beneficial effects of dPDN transplantation on the recovery of locomotor function after SCI. We conclude that dPDN transplants are not sufficient to promote CST regeneration or locomotor recovery after SCI.
Collapse
Affiliation(s)
- Caitlin E Hill
- The Burke Medical Research Institute, 785 Mamaroneck Avenue, White Plains, NY 10605, USA.
| | | | | |
Collapse
|
40
|
Guo X, Zahir T, Mothe A, Shoichet MS, Morshead CM, Katayama Y, Tator CH. The Effect of Growth Factors and Soluble Nogo-66 Receptor Protein on Transplanted Neural Stem/Progenitor Survival and Axonal Regeneration after Complete Transection of Rat Spinal Cord. Cell Transplant 2012; 21:1177-97. [DOI: 10.3727/096368911x612503] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Adult central mammalian axons show minimal regeneration after spinal cord injury due to loss of oligodendrocytes, demyelination of surviving axons, absence of growth-promoting molecules, and inhibitors of axonal outgrowth. In the present study, we attempted to address these impediments to regeneration by using a combinatory strategy to enhance cell survival and regeneration after complete spinal cord transection (SCT) in adult rats. The strategy comprised: 1) adult rat brain-derived neural stem/progenitor cells (NSPCs) preseeded on laminin-coated chitosan channels; 2) extramedullary chitosan channels to promote axonal regrowth and reduce the barrier caused by scarring; 3) local delivery of a novel rat soluble Nogo-66 receptor protein [NgR(310)ecto-Fc, referred to as NgR] to block the inhibitory effect of myelin-based inhibitors; and 4) local delivery of basic fibroblast growth factor, epidermal growth factor, and platelet-derived growth factor to enhance survival and promote differentiation of transplanted cells. Compared with our previous studies where brain-derived NSPCs preseeded in extramedullary chitosan channels were implanted in the same SCT model but without growth factors and NgR, the present channel–growth factor combination produced greater numbers of surviving NSPCs after SCT. Also, the growth factors promoted preferential differentiation of NSPCs toward oligodendrocytes, while NgR significantly decreased astrocytic differentiation of NSPCs. NgR alone or in combination with NSPCs significantly enhanced the total number of myelinated fibers in the bridge and increased the area of the bridging tissue between the cord stumps. The combination of NgR, growth factors, and NSPCs had synergistic effect on bridge formation. However, only a small number of descending corticospinal tract axons grew into the central portions of the bridges as shown by anterograde tracing of the corticospinal tract with BDA. The majority of the regenerated axons in the channels originated from local host neurons adjacent to the tissue bridges. In conclusion, we showed that growth factors increased survival of transplanted NSPCs whereas NgR enhanced axonal regeneration, but the combination did not have additive effects on functional recovery or regeneration.
Collapse
Affiliation(s)
- Xiaodong Guo
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
- Toronto Western Research Institute, Toronto Western Hospital and University of Toronto, Toronto, Ontario, Canada
| | - Tasneem Zahir
- Department of Chemical Engineering & Applied Chemistry, Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Andrea Mothe
- Toronto Western Research Institute, Toronto Western Hospital and University of Toronto, Toronto, Ontario, Canada
| | - Molly S. Shoichet
- Department of Chemical Engineering & Applied Chemistry, Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Cindi M. Morshead
- Department of Surgery and Institute of Medical Sciences, Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Yusuke Katayama
- Department of Chemical Engineering & Applied Chemistry, Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Charles H. Tator
- Toronto Western Research Institute, Toronto Western Hospital and University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
41
|
Ghosh M, Tuesta LM, Puentes R, Patel S, Melendez K, El Maarouf A, Rutishauser U, Pearse DD. Extensive cell migration, axon regeneration, and improved function with polysialic acid-modified Schwann cells after spinal cord injury. Glia 2012; 60:979-92. [PMID: 22460918 DOI: 10.1002/glia.22330] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2011] [Accepted: 02/28/2012] [Indexed: 01/07/2023]
Abstract
Schwann cell (SC) implantation after spinal cord injury (SCI) promotes axonal regeneration, remyelination repair, and functional recovery. Reparative efficacy, however, may be limited because of the inability of SCs to migrate outward from the lesion-implant site. Altering SC cell surface properties by overexpressing polysialic acid (PSA) has been shown to promote SC migration. In this study, a SCI contusion model was used to evaluate the migration, supraspinal axon growth support, and functional recovery associated with polysialyltransferase (PST)-overexpressing SCs [PST-green fluorescent protein (GFP) SCs] or controls (GFP SCs). Compared with GFP SCs, which remained confined to the injection site at the injury center, PST-GFP SCs migrated across the lesion:host cord interface for distances of up to 4.4 mm within adjacent host tissue. In addition, with PST-GFP SCs, there was extensive serotonergic and corticospinal axon in-growth within the implants that was limited in the GFP SC controls. The enhanced migration of PST-GFP SCs was accompanied by significant growth of these axons caudal to lesion. Animals receiving PST-GFP SCs exhibited improved functional outcome, both in the open-field and on the gridwalk test, beyond the modest improvements provided by GFP SC controls. This study for the first time demonstrates that a lack of migration by SCs may hinder their reparative benefits and that cell surface overexpression of PSA enhances the ability of implanted SCs to associate with and support the growth of corticospinal axons. These results provide further promise that PSA-modified SCs will be a potent reparative approach for SCI. © 2012 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Mousumi Ghosh
- Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33101, USA
| | | | | | | | | | | | | | | |
Collapse
|
42
|
|
43
|
|
44
|
Abstract
More than 1 million people in the United States live with a spinal cord injury (SCI). Despite medical advances, many patients with SCIs still experience substantial neurological disability, with loss of motor, sensory, and autonomic function. Cell therapy is ideally suited to address the multifactorial nature of the secondary events following SCI. Remarkable advances in our understanding of the pathophysiology of SCI, structural and functional magnetic resonance imaging, image-guided micro-neurosurgical techniques, and transplantable cell biology have enabled the use of cell-based regenerative techniques in the clinic. It is important to note that there are more than a dozen recently completed, ongoing, or recruiting cell therapy clinical trials for SCI that reflect the views of many key stakeholders. The field of regenerative neuroscience has reached a stage in which the clinical trials are scientifically and ethically justified. Although experimental models and analysis methods and techniques continue to evolve, no model will completely replicate the human condition. It is recognized that more work with cervical models of contusive/compressive SCI are required in parallel with clinical trials. It is also important that the clinical translation of advances made through well-established and validated experimental approaches in animal models move forward to meet the compelling needs of individuals with SCI and to advance the field of regenerative neuroscience. However, it is imperative that such efforts at translation be done in the most rigorous and informed fashion to determine safety and possible efficacy, and to provide key information to clinicians and basic scientists, which will allow improvements in regenerative techniques and the validation and refinement of existing preclinical animal models and research approaches. The field of regenerative neuroscience should not be stalled at the animal model stage, but instead the clinical trials need to be focused, safe, and ethical, backed up by a robust, translationally relevant preclinical research strategy.
Collapse
Affiliation(s)
- Michael G. Fehlings
- University Health Network, Toronto Western Hospital, Toronto, ON M5T 2S8 Canada
| | - Reaz Vawda
- University Health Network, Toronto Western Hospital, Toronto, ON M5T 2S8 Canada
| |
Collapse
|
45
|
Abstract
Understanding restricted functional recovery and designing efficient treatments to alleviate dysfunction after injury of the nervous system remain major challenges in neuroscience and medicine. Numerous molecules of potential significance in neural repair have been identified in vitro, but only few of these have proved to be of major importance in vivo up to now. Among the molecules involved in regeneration are several members of the immunoglobulin superfamily, most notably the neural cell adhesion molecules L1, its close homologue CHL1, and NCAM and, in particular, its polysialic acid glycan moiety. Sufficient evidence is now available to justify the statement that these molecules are major players not only in nervous system development but also in the adult during neural repair and synaptic plasticity. Importantly, insights into the functions of these molecules in promoting or inhibiting functional recovery have allowed the design and assessment of therapeutic approaches in animal models of central nervous system injury that could prove to be applicable in clinical settings.
Collapse
Affiliation(s)
- Andrey Irintchev
- Neuroscience Laboratory, Department of Otorhinolaryngology, University of Jena, Germany
| | - Melitta Schachner
- Zentrum für Molekulare Neurobiologie, Universität Hamburg, Hamburg, Germany
- Keck Center for Collaborative Neuroscience, Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, USA
| |
Collapse
|
46
|
Degeorge ML, Marlowe D, Werner E, Soderstrom KE, Stock M, Mueller A, Bohn MC, Kozlowski DA. Combining glial cell line-derived neurotrophic factor gene delivery (AdGDNF) with L-arginine decreases contusion size but not behavioral deficits after traumatic brain injury. Brain Res 2011; 1403:45-56. [PMID: 21672665 DOI: 10.1016/j.brainres.2011.05.058] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2011] [Revised: 05/20/2011] [Accepted: 05/25/2011] [Indexed: 01/09/2023]
Abstract
Our laboratory has previously demonstrated that viral administration of glial cell line-derived neurotrophic factor (AdGDNF), one week prior to a controlled cortical impact (CCI) over the forelimb sensorimotor cortex of the rat (FL-SMC) is neuroprotective, but does not significantly enhance recovery of sensorimotor function. One possible explanation for this discrepancy is that although protected, neurons may not have been functional due to enduring metabolic deficiencies. Additionally, metabolic events following TBI may interfere with expression of therapeutic proteins administered to the injured brain via gene therapy. The current study focused on enhancing the metabolic function of the brain by increasing cerebral blood flow (CBF) with l-arginine in conjunction with administration of AdGDNF immediately following CCI. An adenoviral vector harboring human GDNF was injected unilaterally into FL-SMC of the rat immediately following a unilateral CCI over the FL-SMC. Within 30min of the CCI and AdGDNF injections, some animals were injected with l-arginine (i.v.). Tests of forelimb function and asymmetry were administered for 4weeks post-injury. Animals were sacrificed and contusion size and GDNF protein expression measured. This study demonstrated that rats treated with AdGDNF and l-arginine post-CCI had a significantly smaller contusion than injured rats who did not receive any treatment, or injured rats treated with either AdGDNF or l-arginine alone. Nevertheless, no amelioration of behavioral deficits was seen. These findings suggest that AdGDNF alone following a CCI was not therapeutic and although combining it with l-arginine decreased contusion size, it did not enhance behavioral recovery.
Collapse
Affiliation(s)
- M L Degeorge
- DePaul University, Department of Biological Sciences, Chicago, IL 60614, USA
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Patel V, Joseph G, Patel A, Patel S, Bustin D, Mawson D, Tuesta LM, Puentes R, Ghosh M, Pearse DD. Suspension matrices for improved Schwann-cell survival after implantation into the injured rat spinal cord. J Neurotrauma 2010; 27:789-801. [PMID: 20144012 DOI: 10.1089/neu.2008.0809] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Trauma to the spinal cord produces endogenously irreversible tissue and functional loss, requiring the application of therapeutic approaches to achieve meaningful restoration. Cellular strategies, in particular Schwann-cell implantation, have shown promise in overcoming many of the obstacles facing successful repair of the injured spinal cord. Here, we show that the implantation of Schwann cells as cell suspensions with in-situ gelling laminin:collagen matrices after spinal-cord contusion significantly enhances long-term cell survival but not proliferation, as well as improves graft vascularization and the degree of axonal in-growth over the standard implantation vehicle, minimal media. The use of a matrix to suspend cells prior to implantation should be an important consideration for achieving improved survival and effectiveness of cellular therapies for future clinical application.
Collapse
Affiliation(s)
- Vivek Patel
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, Florida 33101, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Tetzlaff W, Okon EB, Karimi-Abdolrezaee S, Hill CE, Sparling JS, Plemel JR, Plunet WT, Tsai EC, Baptiste D, Smithson LJ, Kawaja MD, Fehlings MG, Kwon BK. A systematic review of cellular transplantation therapies for spinal cord injury. J Neurotrauma 2010; 28:1611-82. [PMID: 20146557 DOI: 10.1089/neu.2009.1177] [Citation(s) in RCA: 404] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Cell transplantation therapies have become a major focus in pre-clinical research as a promising strategy for the treatment of spinal cord injury (SCI). In this article, we systematically review the available pre-clinical literature on the most commonly used cell types in order to assess the body of evidence that may support their translation to human SCI patients. These cell types include Schwann cells, olfactory ensheathing glial cells, embryonic and adult neural stem/progenitor cells, fate-restricted neural/glial precursor cells, and bone-marrow stromal cells. Studies were included for review only if they described the transplantation of the cell substrate into an in-vivo model of traumatic SCI, induced either bluntly or sharply. Using these inclusion criteria, 162 studies were identified and reviewed in detail, emphasizing their behavioral effects (although not limiting the scope of the discussion to behavioral effects alone). Significant differences between cells of the same "type" exist based on the species and age of donor, as well as culture conditions and mode of delivery. Many of these studies used cell transplantations in combination with other strategies. The systematic review makes it very apparent that cells derived from rodent sources have been the most extensively studied, while only 19 studies reported the transplantation of human cells, nine of which utilized bone-marrow stromal cells. Similarly, the vast majority of studies have been conducted in rodent models of injury, and few studies have investigated cell transplantation in larger mammals or primates. With respect to the timing of intervention, nearly all of the studies reviewed were conducted with transplantations occurring subacutely and acutely, while chronic treatments were rare and often failed to yield functional benefits.
Collapse
Affiliation(s)
- Wolfram Tetzlaff
- University of British Columbia, ICORD, Vancouver, British Columbia, Canada.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Nomura H, Baladie B, Katayama Y, Morshead CM, Shoichet MS, Tator CH. Delayed implantation of intramedullary chitosan channels containing nerve grafts promotes extensive axonal regeneration after spinal cord injury. Neurosurgery 2009; 63:127-41; discussion 141-3. [PMID: 18728578 DOI: 10.1227/01.neu.0000335080.47352.31] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVE We describe a new strategy to promote axonal regeneration after subacute or chronic spinal cord injury consisting of intramedullary implantation of chitosan guidance channels containing peripheral nerve (PN) grafts. METHODS Chitosan channels filled with PN grafts harvested from green fluorescent protein rats were implanted in the cavity 1 week (subacute) or 4 weeks (chronic) after 50-g clip injury at T8 and were compared with similarly injured animals implanted with either unfilled channels or no channels. Functional recovery was measured weekly for 12 weeks by open-field locomotion, after which histological examination was performed. RESULTS The implanted channels with PN grafts contained a thick tissue bridge containing as many as 35,000 myelinated axons in both the subacute and chronic spinal cord injury groups, with the greatest number of axons in the channels containing PN grafts implanted subacutely. There were numerous green fluorescent protein-positive donor Schwann cells in the tissue bridges in all animals with PN grafts. Moreover, these Schwann cells had high functional capacity in terms of myelination of the axons in the channels. In addition, PN-filled chitosan channels showed excellent biocompatibility with the adjacent neural tissue and no obvious signs of degradation and minimal tissue reaction at 14 weeks after implantation. In control animals that had unfilled chitosan channels implanted, there was minimal axonal regeneration in the channels; in control animals without channels, there were large cavities in the spinal cords, and the bridges contained only a small number of axons and Schwann cells. Despite the large numbers of axons in the chitosan channel-PN graft group, there was no significant difference in functional recovery between treatment and control groups. CONCLUSION Intramedullary implantation of chitosan guidance channels containing PN grafts in the cavity after subacute spinal cord injury resulted in a thicker bridge containing a larger number of myelinated axons compared with chitosan channels alone. A chitosan channel containing PN grafts is a promising strategy for spinal cord repair.
Collapse
Affiliation(s)
- Hiroshi Nomura
- Toronto Western Research Institute, Toronto Western Hospital, Toronto, Canada
| | | | | | | | | | | |
Collapse
|
50
|
Combinatorial strategies with Schwann cell transplantation to improve repair of the injured spinal cord. Neurosci Lett 2009; 456:124-32. [PMID: 19429147 DOI: 10.1016/j.neulet.2008.08.092] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2008] [Revised: 07/29/2008] [Accepted: 08/04/2008] [Indexed: 12/11/2022]
|