1
|
Qu W, Wu X, Wu W, Wang Y, Sun Y, Deng L, Walker M, Chen C, Dai H, Han Q, Ding Y, Xia Y, Smith G, Li R, Liu NK, Xu XM. Chondroitinase ABC combined with Schwann cell transplantation enhances restoration of neural connection and functional recovery following acute and chronic spinal cord injury. Neural Regen Res 2025; 20:1467-1482. [PMID: 39075913 DOI: 10.4103/nrr.nrr-d-23-01338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 05/16/2024] [Indexed: 07/31/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202505000-00029/figure1/v/2024-07-28T173839Z/r/image-tiff Schwann cell transplantation is considered one of the most promising cell-based therapy to repair injured spinal cord due to its unique growth-promoting and myelin-forming properties. A the Food and Drug Administration-approved Phase I clinical trial has been conducted to evaluate the safety of transplanted human autologous Schwann cells to treat patients with spinal cord injury. A major challenge for Schwann cell transplantation is that grafted Schwann cells are confined within the lesion cavity, and they do not migrate into the host environment due to the inhibitory barrier formed by injury-induced glial scar, thus limiting axonal reentry into the host spinal cord. Here we introduce a combinatorial strategy by suppressing the inhibitory extracellular environment with injection of lentivirus-mediated transfection of chondroitinase ABC gene at the rostral and caudal borders of the lesion site and simultaneously leveraging the repair capacity of transplanted Schwann cells in adult rats following a mid-thoracic contusive spinal cord injury. We report that when the glial scar was degraded by chondroitinase ABC at the rostral and caudal lesion borders, Schwann cells migrated for considerable distances in both rostral and caudal directions. Such Schwann cell migration led to enhanced axonal regrowth, including the serotonergic and dopaminergic axons originating from supraspinal regions, and promoted recovery of locomotor and urinary bladder functions. Importantly, the Schwann cell survival and axonal regrowth persisted up to 6 months after the injury, even when treatment was delayed for 3 months to mimic chronic spinal cord injury. These findings collectively show promising evidence for a combinatorial strategy with chondroitinase ABC and Schwann cells in promoting remodeling and recovery of function following spinal cord injury.
Collapse
Affiliation(s)
- Wenrui Qu
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Hand Surgery, the Second Hospital of Jilin University, Changchun, Jilin Province, China
| | - Xiangbing Wu
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Wei Wu
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Ying Wang
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Yan Sun
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Lingxiao Deng
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Melissa Walker
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Chen Chen
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Heqiao Dai
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Qi Han
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Ying Ding
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Yongzhi Xia
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - George Smith
- Shriners Hospitals Pediatric Research Center, Temple University School of Medicine, Philadelphia, PA, USA
| | - Rui Li
- Department of Hand Surgery, the Second Hospital of Jilin University, Changchun, Jilin Province, China
| | - Nai-Kui Liu
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Xiao-Ming Xu
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
2
|
Dill-Macky AS, Lee EN, Wertheim JA, Koss KM. Glia in tissue engineering: From biomaterial tools to transplantation. Acta Biomater 2024:S1742-7061(24)00600-7. [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] [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
|
3
|
Guest JD, Santamaria AJ, Solano JP, de Rivero Vaccari JP, Dietrich WD, Pearse DD, Khan A, Levi AD. Challenges in advancing Schwann cell transplantation for spinal cord injury repair. Cytotherapy 2024:S1465-3249(24)00827-2. [PMID: 39387736 DOI: 10.1016/j.jcyt.2024.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 08/05/2024] [Accepted: 08/09/2024] [Indexed: 10/15/2024]
Abstract
BACKGROUND AIMS In this article we aimed to provide an expert synthesis of the current status of Schwann cell (SC)therapeutics and potential steps to increase their clinical utility. METHODS We provide an expert synthesis based on preclinical, clinical and manufacturing experience. RESULTS Schwann cells (SCs) are essential for peripheral nerve regeneration and are of interest in supporting axonal repair after spinal cord injury (SCI). SCs can be isolated and cultivated in tissue culture from adult nerve biopsies or generated from precursors and neural progenitors using specific differentiation protocols leading to expanded quantities. In culture, they undergo dedifferentiation to a state similar to "repair" SCs. The known repertoire of SC functions is increasing beyond axon maintenance, myelination, and axonal regeneration to include immunologic regulation and the release of potentially therapeutic extracellular vesicles. Recently, autologous human SC cultures purified under cGMP conditions have been tested in both nerve repair and subacute and chronic SCI clinical trials. Although the effects of SCs to support nerve regeneration are indisputable, their efficacy for clinical SCI has been limited according to the outcomes examined. CONCLUSIONS This review discusses the current limitations of transplanted SCs within the damaged spinal cord environment. Limitations include limited post-transplant cell survival, the inability of SCs to migrate within astrocytic parenchyma, and restricted axonal regeneration out of SC-rich graft regions. We describe steps to amplify the survival and integration of transplanted SCs and to expand the repertoire of uses of SCs, including SC-derived extracellular vesicles. The relative merits of transplanting autologous versus allogeneic SCs and the role that endogenous SCs play in spinal cord repair are described. Finally, we briefly describe the issues requiring solutions to scale up SC manufacturing for commercial use.
Collapse
Affiliation(s)
- James D Guest
- The Miami Project to Cure Paralysis and Neurological Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA.
| | - Andrea J Santamaria
- The Miami Project to Cure Paralysis, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Juan P Solano
- Pediatric Critical Care, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Juan P de Rivero Vaccari
- The Miami Project to Cure Paralysis and Neurological Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - William D Dietrich
- The Miami Project to Cure Paralysis and Neurological Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Damien D Pearse
- The Miami Project to Cure Paralysis and Neurological Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Aisha Khan
- The Stem Cell Institute, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Allan D Levi
- The Miami Project to Cure Paralysis and Neurological Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| |
Collapse
|
4
|
Du X, Zhang S, Khabbaz A, Cohen KL, Zhang Y, Chakraborty S, Smith GM, Wang H, Yadav AP, Liu N, Deng L. Regeneration of Propriospinal Axons in Rat Transected Spinal Cord Injury through a Growth-Promoting Pathway Constructed by Schwann Cells Overexpressing GDNF. Cells 2024; 13:1160. [PMID: 38995011 PMCID: PMC11240522 DOI: 10.3390/cells13131160] [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: 04/13/2024] [Revised: 06/01/2024] [Accepted: 06/28/2024] [Indexed: 07/13/2024] Open
Abstract
Unsuccessful axonal regeneration in transected spinal cord injury (SCI) is mainly attributed to shortage of growth factors, inhibitory glial scar, and low intrinsic regenerating capacity of severely injured neurons. Previously, we constructed an axonal growth permissive pathway in a thoracic hemisected injury by transplantation of Schwann cells overexpressing glial-cell-derived neurotrophic factor (SCs-GDNF) into the lesion gap as well as the caudal cord and proved that this novel permissive bridge promoted the regeneration of descending propriospinal tract (dPST) axons across and beyond the lesion. In the current study, we subjected rats to complete thoracic (T11) spinal cord transections and examined whether these combinatorial treatments can support dPST axons' regeneration beyond the transected injury. The results indicated that GDNF significantly improved graft-host interface by promoting integration between SCs and astrocytes, especially the migration of reactive astrocyte into SCs-GDNF territory. The glial response in the caudal graft area has been significantly attenuated. The astrocytes inside the grafted area were morphologically characterized by elongated and slim process and bipolar orientation accompanied by dramatically reduced expression of glial fibrillary acidic protein. Tremendous dPST axons have been found to regenerate across the lesion and back to the caudal spinal cord which were otherwise difficult to see in control groups. The caudal synaptic connections were formed, and regenerated axons were remyelinated. The hindlimb locomotor function has been improved.
Collapse
Affiliation(s)
- Xiaolong Du
- Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (X.D.); (S.Z.); (A.K.); (K.L.C.); (Y.Z.); (S.C.)
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Goodman and Campbell Brain and Spine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Vascular Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210005, China
| | - Shengqi Zhang
- Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (X.D.); (S.Z.); (A.K.); (K.L.C.); (Y.Z.); (S.C.)
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Goodman and Campbell Brain and Spine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Rehabilitation Medicine, Zhongda Hospital Southeast University, Nanjing 210009, China;
| | - Aytak Khabbaz
- Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (X.D.); (S.Z.); (A.K.); (K.L.C.); (Y.Z.); (S.C.)
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Goodman and Campbell Brain and Spine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Kristen Lynn Cohen
- Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (X.D.); (S.Z.); (A.K.); (K.L.C.); (Y.Z.); (S.C.)
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Goodman and Campbell Brain and Spine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Yihong Zhang
- Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (X.D.); (S.Z.); (A.K.); (K.L.C.); (Y.Z.); (S.C.)
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Goodman and Campbell Brain and Spine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Samhita Chakraborty
- Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (X.D.); (S.Z.); (A.K.); (K.L.C.); (Y.Z.); (S.C.)
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Goodman and Campbell Brain and Spine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - George M. Smith
- Shriners Hospitals Pediatric Research Center, School of Medicine, Temple University, Philadelphia, PA 19140, USA;
| | - Hongxing Wang
- Department of Rehabilitation Medicine, Zhongda Hospital Southeast University, Nanjing 210009, China;
| | - Amol P. Yadav
- Department of Biomedical Engineering, the University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
| | - Naikui Liu
- Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (X.D.); (S.Z.); (A.K.); (K.L.C.); (Y.Z.); (S.C.)
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Goodman and Campbell Brain and Spine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Lingxiao Deng
- Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (X.D.); (S.Z.); (A.K.); (K.L.C.); (Y.Z.); (S.C.)
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Goodman and Campbell Brain and Spine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
5
|
Ou YC, Huang CC, Kao YL, Ho PC, Tsai KJ. Stem Cell Therapy in Spinal Cord Injury-Induced Neurogenic Lower Urinary Tract Dysfunction. Stem Cell Rev Rep 2023; 19:1691-1708. [PMID: 37115409 DOI: 10.1007/s12015-023-10547-9] [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] [Accepted: 04/14/2023] [Indexed: 04/29/2023]
Abstract
Spinal cord injury (SCI) is a devastating condition that enormously affects an individual's health and quality of life. Neurogenic lower urinary tract dysfunction (NLUTD) is one of the most important sequelae induced by SCI, causing complications including urinary tract infection, renal function deterioration, urinary incontinence, and voiding dysfunction. Current therapeutic methods for SCI-induced NLUTD mainly target on the urinary bladder, but the outcomes are still far from satisfactory. Stem cell therapy has gained increasing attention for years for its ability to rescue the injured spinal cord directly. Stem cell differentiation and their paracrine effects, including exosomes, are the proposed mechanisms to enhance the recovery from SCI. Several animal studies have demonstrated improvement in bladder function using mesenchymal stem cells (MSCs) and neural stem cells (NSCs). Human clinical trials also provide promising results in urodynamic parameters after MSC therapy. However, there is still uncertainty about the ideal treatment window and application protocol for stem cell therapy. Besides, data on the therapeutic effects regarding NSCs and stem cell-derived exosomes in SCI-related NLUTD are scarce. Therefore, there is a pressing need for further well-designed human clinical trials to translate the stem cell therapy into a formal therapeutic option for SCI-induced NLUTD.
Collapse
Affiliation(s)
- Yin-Chien Ou
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, No. 1, University Road, Tainan, 701, Taiwan
- Department of Urology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chi-Chen Huang
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, No. 1, University Road, Tainan, 701, Taiwan
- Section of Neurosurgery, Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yao-Lin Kao
- Department of Urology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Pei-Chuan Ho
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, No. 1, University Road, Tainan, 701, Taiwan
| | - Kuen-Jer Tsai
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, No. 1, University Road, Tainan, 701, Taiwan.
- Research Center of Clinical Medicine, National Cheng Kung University Hospital , College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
6
|
Khaing ZZ, Chen JY, Safarians G, Ezubeik S, Pedroncelli N, Duquette RD, Prasse T, Seidlits SK. Clinical Trials Targeting Secondary Damage after Traumatic Spinal Cord Injury. Int J Mol Sci 2023; 24:3824. [PMID: 36835233 PMCID: PMC9960771 DOI: 10.3390/ijms24043824] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 02/06/2023] [Accepted: 02/09/2023] [Indexed: 02/17/2023] Open
Abstract
Spinal cord injury (SCI) often causes loss of sensory and motor function resulting in a significant reduction in quality of life for patients. Currently, no therapies are available that can repair spinal cord tissue. After the primary SCI, an acute inflammatory response induces further tissue damage in a process known as secondary injury. Targeting secondary injury to prevent additional tissue damage during the acute and subacute phases of SCI represents a promising strategy to improve patient outcomes. Here, we review clinical trials of neuroprotective therapeutics expected to mitigate secondary injury, focusing primarily on those in the last decade. The strategies discussed are broadly categorized as acute-phase procedural/surgical interventions, systemically delivered pharmacological agents, and cell-based therapies. In addition, we summarize the potential for combinatorial therapies and considerations.
Collapse
Affiliation(s)
- Zin Z. Khaing
- Department of Neurological Surgery, University of Washington, Seattle, WA 98195, USA
| | - Jessica Y. Chen
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Gevick Safarians
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Sohib Ezubeik
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Nicolas Pedroncelli
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Rebecca D. Duquette
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095, USA
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX 78712, USA
| | - Tobias Prasse
- Department of Neurological Surgery, University of Washington, Seattle, WA 98195, USA
- Department of Orthopedics and Trauma Surgery, University of Cologne, 50931 Cologne, Germany
| | - Stephanie K. Seidlits
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095, USA
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX 78712, USA
| |
Collapse
|
7
|
Kitagawa T, Nagoshi N, Okano H, Nakamura M. A Narrative Review of Advances in Neural Precursor Cell Transplantation Therapies for Spinal Cord Injury. Neurospine 2022; 19:935-945. [PMID: 36597632 PMCID: PMC9816589 DOI: 10.14245/ns.2244628.314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 09/11/2022] [Indexed: 12/27/2022] Open
Abstract
A spinal cord injury (SCI) is a destructive event that causes a permanent deficit in neurological function because of poor regenerative potential. Transplantation therapies have attracted attention for restoration of the injured spinal cord, and transplantation of neural precursor cells (NPCs) has been studied worldwide. Several groups have demonstrated functional recovery via this therapeutic intervention due to the multiple beneficial effects of NPC transplantation, such as reconstruction of neuronal circuits, remyelination of axons, and neuroprotection by trophic factors. Our group developed a method to induce NPCs from human induced pluripotent stem cells (hiPSCs) and established a transplantation strategy for SCI. Functional improvement in SCI animals treated with hiPSC-NPCs was observed, and the safety of transplanting these cells was evaluated from multiple perspectives. With selection of a safe cell line and pretreatment of the cells to encourage maturation and differentiation, hiPSC-NPC transplantation therapy is now in the clinical phase of testing for subacute SCI. In addition, a research challenge will be to expand the efficacy of transplantation therapy for chronic SCI. More comprehensive strategies involving combination treatments are required to treat this problematic situation.
Collapse
Affiliation(s)
- Takahiro Kitagawa
- Department of Orthopaedic Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Narihito Nagoshi
- Department of Orthopaedic Surgery, Keio University School of Medicine, Tokyo, Japan,Corresponding Author Narihito Nagoshi Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Masaya Nakamura
- Department of Orthopaedic Surgery, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
8
|
Hall A, Fortino T, Spruance V, Niceforo A, Harrop JS, Phelps PE, Priest CA, Zholudeva LV, Lane MA. Cell transplantation to repair the injured spinal cord. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2022; 166:79-158. [PMID: 36424097 PMCID: PMC10008620 DOI: 10.1016/bs.irn.2022.09.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Adam Hall
- Drexel University, Philadelphia, PA, United States; Marion Murray Spinal Cord Research Center, Drexel University, Philadelphia, PA, United States
| | - Tara Fortino
- Drexel University, Philadelphia, PA, United States; Marion Murray Spinal Cord Research Center, Drexel University, Philadelphia, PA, United States
| | - Victoria Spruance
- Drexel University, Philadelphia, PA, United States; Marion Murray Spinal Cord Research Center, Drexel University, Philadelphia, PA, United States; Division of Kidney, Urologic, & Hematologic Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Alessia Niceforo
- Drexel University, Philadelphia, PA, United States; Marion Murray Spinal Cord Research Center, Drexel University, Philadelphia, PA, United States
| | - James S Harrop
- Department of Neurological and Orthopedic Surgery, Thomas Jefferson University, Philadelphia, PA, United States
| | - Patricia E Phelps
- Department of Integrative Biology & Physiology, UCLA, Los Angeles, CA, United States
| | | | - Lyandysha V Zholudeva
- Drexel University, Philadelphia, PA, United States; Marion Murray Spinal Cord Research Center, Drexel University, Philadelphia, PA, United States; Gladstone Institutes, San Francisco, CA, United States
| | - Michael A Lane
- Drexel University, Philadelphia, PA, United States; Marion Murray Spinal Cord Research Center, Drexel University, Philadelphia, PA, United States.
| |
Collapse
|
9
|
Gant KL, Guest JD, Palermo AE, Vedantam A, Jimsheleishvili G, Bunge MB, Brooks AE, Anderson KD, Thomas CK, Santamaria AJ, Perez MA, Curiel R, Nash MS, Saraf-Lavi E, Pearse DD, Widerström-Noga E, Khan A, Dietrich WD, Levi AD. Phase 1 Safety Trial of Autologous Human Schwann Cell Transplantation in Chronic Spinal Cord Injury. J Neurotrauma 2022; 39:285-299. [PMID: 33757304 PMCID: PMC9360180 DOI: 10.1089/neu.2020.7590] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
A phase 1 open-label, non-randomized clinical trial was conducted to determine feasibility and safety of autologous human Schwann cell (ahSC) transplantation accompanied by rehabilitation in participants with chronic spinal cord injury (SCI). Magnetic resonance imaging (MRI) was used to screen eligible participants to estimate an individualized volume of cell suspension to be implanted. The trial incorporated standardized multi-modal rehabilitation before and after cell delivery. Participants underwent sural nerve harvest, and ahSCs were isolated and propagated in culture. The dose of culture-expanded ahSCs injected into the chronic spinal cord lesion of each individual followed a cavity-filling volume approach. Primary outcome measures for safety and trend-toward efficacy were assessed. Two participants with American Spinal Injury Association Impairment Scale (AIS) A and two participants with incomplete chronic SCI (AIS B, C) were each enrolled in cervical and thoracic SCI cohorts (n = 8 total). All participants completed the study per protocol, and no serious adverse events related to sural nerve harvest or ahSC transplantation were reported. Urinary tract infections and skin abrasions were the most common adverse events reported. One participant experienced a 4-point improvement in motor function, a 6-point improvement in sensory function, and a 1-level improvement in neurological level of injury. Follow-up MRI in the cervical (6 months) and thoracic (24 months) cohorts revealed a reduction in cyst volume after transplantation with reduced effect over time. This phase 1 trial demonstrated the feasibility and safety of ahSC transplantation combined with a multi-modal rehabilitation protocol for participants with chronic SCI.
Collapse
Affiliation(s)
- Katie L. Gant
- The Miami Project to Cure Paralysis, University of Miami, Miami, Florida, USA
- Department of Neurological Surgery, University of Miami, Miami, Florida, USA
| | - James D. Guest
- The Miami Project to Cure Paralysis, University of Miami, Miami, Florida, USA
- Department of Neurological Surgery, University of Miami, Miami, Florida, USA
- Department of Neuroscience, University of Miami, Miami, Florida, USA
| | - Anne E. Palermo
- The Miami Project to Cure Paralysis, University of Miami, Miami, Florida, USA
- Department of Neurological Surgery, University of Miami, Miami, Florida, USA
| | - Aditya Vedantam
- The Miami Project to Cure Paralysis, University of Miami, Miami, Florida, USA
- Department of Neurological Surgery, University of Miami, Miami, Florida, USA
| | - George Jimsheleishvili
- The Miami Project to Cure Paralysis, University of Miami, Miami, Florida, USA
- Department of Neurological Surgery, University of Miami, Miami, Florida, USA
| | - Mary Bartlett Bunge
- The Miami Project to Cure Paralysis, University of Miami, Miami, Florida, USA
- Department of Neurological Surgery, University of Miami, Miami, Florida, USA
- Department of Neuroscience, University of Miami, Miami, Florida, USA
- Department of Cell Biology, University of Miami, Miami, Florida, USA
- Department of Neurology, University of Miami, Miami, Florida, USA
- Department of Interdisciplinary Stem Cell Institute, University of Miami, Miami, Florida, USA
| | - Adriana E. Brooks
- The Miami Project to Cure Paralysis, University of Miami, Miami, Florida, USA
- Department of Interdisciplinary Stem Cell Institute, University of Miami, Miami, Florida, USA
| | - Kim D. Anderson
- Department of Physical Medicine and Rehabilitation, Case Western Reserve University, Metrohealth Medical Center, Cleveland, Ohio, USA
| | - Christine K. Thomas
- The Miami Project to Cure Paralysis, University of Miami, Miami, Florida, USA
- Department of Neurological Surgery, University of Miami, Miami, Florida, USA
| | - Andrea J. Santamaria
- The Miami Project to Cure Paralysis, University of Miami, Miami, Florida, USA
- Department of Neurological Surgery, University of Miami, Miami, Florida, USA
| | - Monica A. Perez
- The Miami Project to Cure Paralysis, University of Miami, Miami, Florida, USA
- Department of Neurological Surgery, University of Miami, Miami, Florida, USA
- Bruce W. Carter Department of Veterans Affairs Medical Center, Miami, Florida, USA
- Shirley Ryan AbilityLab, Northwestern University, Edward Hines Jr, VA Hospital, Chicago, Illinois, USA
| | - Rosie Curiel
- Department of Psychiatry, University of Miami, Miami, Florida, USA
| | - Mark S. Nash
- Department of Rehabilitation Medicine, University of Miami, Miami, Florida, USA
| | - Efrat Saraf-Lavi
- Department of Radiology, University of Miami, Miami, Florida, USA
| | - Damien D. Pearse
- Department of Neuroscience, University of Miami, Miami, Florida, USA
- Department of Interdisciplinary Stem Cell Institute, University of Miami, Miami, Florida, USA
- Bruce W. Carter Department of Veterans Affairs Medical Center, Miami, Florida, USA
- Shirley Ryan AbilityLab, Northwestern University, Edward Hines Jr, VA Hospital, Chicago, Illinois, USA
| | - Eva Widerström-Noga
- The Miami Project to Cure Paralysis, University of Miami, Miami, Florida, USA
- Department of Neurological Surgery, University of Miami, Miami, Florida, USA
- Department of Neuroscience, University of Miami, Miami, Florida, USA
- Department of Rehabilitation Medicine, University of Miami, Miami, Florida, USA
- Bruce W. Carter Department of Veterans Affairs Medical Center, Miami, Florida, USA
| | - Aisha Khan
- The Miami Project to Cure Paralysis, University of Miami, Miami, Florida, USA
- Department of Interdisciplinary Stem Cell Institute, University of Miami, Miami, Florida, USA
| | - W. Dalton Dietrich
- The Miami Project to Cure Paralysis, University of Miami, Miami, Florida, USA
- Department of Neurological Surgery, University of Miami, Miami, Florida, USA
- Department of Neuroscience, University of Miami, Miami, Florida, USA
- Department of Cell Biology, University of Miami, Miami, Florida, USA
- Department of Neurology, University of Miami, Miami, Florida, USA
- Department of Interdisciplinary Stem Cell Institute, University of Miami, Miami, Florida, USA
| | - Allan D. Levi
- The Miami Project to Cure Paralysis, University of Miami, Miami, Florida, USA
- Department of Neurological Surgery, University of Miami, Miami, Florida, USA
- Department of Neuroscience, University of Miami, Miami, Florida, USA
| |
Collapse
|
10
|
Khan A, Diaz A, Brooks AE, Burks SS, Athauda G, Wood P, Lee YS, Silvera R, Donaldson M, Pressman Y, Anderson KD, Bunge MB, Pearse DD, Dietrich WD, Guest JD, Levi AD. Scalable culture techniques to generate large numbers of purified human Schwann cells for clinical trials in human spinal cord and peripheral nerve injuries. J Neurosurg Spine 2021; 36:135-144. [PMID: 34479193 DOI: 10.3171/2020.11.spine201433] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Accepted: 11/24/2020] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Schwann cells (SCs) have been shown to play an essential role in axon regeneration in both peripheral nerve injuries (PNIs) and spinal cord injuries (SCIs). The transplantation of SCs as an adjunctive therapy is currently under investigation in human clinical trials due to their regenerative capacity. Therefore, a reliable method for procuring large quantities of SCs from peripheral nerves is necessary. This paper presents a well-developed, validated, and optimized manufacturing protocol for clinical-grade SCs that are compliant with Current Good Manufacturing Practices (CGMPs). METHODS The authors evaluated the SC culture manufacturing data from 18 clinical trial participants who were recruited for autologous SC transplantation due to subacute SCI (n = 7), chronic SCI (n = 8), or PNIs (n = 3). To initiate autologous SC cultures, a mean nerve length of 11.8 ± 3.7 cm was harvested either from the sural nerve alone (n = 17) or with the sciatic nerve (n = 1). The nerves were digested with enzymes and SCs were isolated and further expanded in multiple passages to meet the dose requirements for transplantation. RESULTS An average yield of 87.2 ± 89.2 million cells at P2 and 150.9 ± 129.9 million cells at P3 with high viability and purity was produced. Cell counts and rates of expansion increased with each subsequent passage from P0 to P3, with the largest rate of expansion between P2 and P3. Larger harvest nerve lengths correlated significantly with greater yields at P0 and P1 (p < 0.05). In addition, a viability and purity above 90% was sustained throughout all passages in nearly all cell products. CONCLUSIONS This study presents reliable CGMP-compliant manufacturing methods for autologous SC products that are suitable for regenerative treatment of patients with SCI, PNI, or other conditions.
Collapse
Affiliation(s)
- Aisha Khan
- 1The Miami Project to Cure Paralysis.,3Interdisciplinary Stem Cell Institute, and Departments of
| | - Anthony Diaz
- 1The Miami Project to Cure Paralysis.,2Department of Neurological Surgery
| | - Adriana E Brooks
- 1The Miami Project to Cure Paralysis.,3Interdisciplinary Stem Cell Institute, and Departments of
| | - S Shelby Burks
- 1The Miami Project to Cure Paralysis.,2Department of Neurological Surgery
| | - Gagani Athauda
- 7Department of Cellular Biology and Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida; and
| | - Patrick Wood
- 1The Miami Project to Cure Paralysis.,2Department of Neurological Surgery
| | - Yee-Shuan Lee
- 3Interdisciplinary Stem Cell Institute, and Departments of
| | - Risset Silvera
- 1The Miami Project to Cure Paralysis.,3Interdisciplinary Stem Cell Institute, and Departments of
| | - Maxwell Donaldson
- 1The Miami Project to Cure Paralysis.,3Interdisciplinary Stem Cell Institute, and Departments of
| | | | - Kim D Anderson
- 8Department of Physical Medicine and Rehabilitation, MetroHealth Medical Center, Institute for Functional Restoration, Case Western Reserve University School, Cleveland, Ohio
| | - Mary Bartlett Bunge
- 1The Miami Project to Cure Paralysis.,2Department of Neurological Surgery.,4Cell Biology and
| | - Damien D Pearse
- 1The Miami Project to Cure Paralysis.,2Department of Neurological Surgery.,3Interdisciplinary Stem Cell Institute, and Departments of.,6Bruce W. Carter Department of Veterans Affairs, Veterans Affairs Medical Center, Miami
| | - W Dalton Dietrich
- 1The Miami Project to Cure Paralysis.,2Department of Neurological Surgery.,4Cell Biology and.,5Neurology, University of Miami Miller School of Medicine, Miami
| | - James D Guest
- 1The Miami Project to Cure Paralysis.,2Department of Neurological Surgery
| | - Allan D Levi
- 1The Miami Project to Cure Paralysis.,2Department of Neurological Surgery
| |
Collapse
|
11
|
Dpp and Hedgehog promote the glial response to neuronal apoptosis in the developing Drosophila visual system. PLoS Biol 2021; 19:e3001367. [PMID: 34379617 PMCID: PMC8396793 DOI: 10.1371/journal.pbio.3001367] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 08/27/2021] [Accepted: 07/16/2021] [Indexed: 11/19/2022] Open
Abstract
Damage in the nervous system induces a stereotypical response that is mediated by glial cells. Here, we use the eye disc of Drosophila melanogaster as a model to explore the mechanisms involved in promoting glial cell response after neuronal cell death induction. We demonstrate that these cells rapidly respond to neuronal apoptosis by increasing in number and undergoing morphological changes, which will ultimately grant them phagocytic abilities. We found that this glial response is controlled by the activity of Decapentaplegic (Dpp) and Hedgehog (Hh) signalling pathways. These pathways are activated after cell death induction, and their functions are necessary to induce glial cell proliferation and migration to the eye discs. The latter of these 2 processes depend on the function of the c-Jun N-terminal kinase (JNK) pathway, which is activated by Dpp signalling. We also present evidence that a similar mechanism controls glial response upon apoptosis induction in the leg discs, suggesting that our results uncover a mechanism that might be involved in controlling glial cells response to neuronal cell death in different regions of the peripheral nervous system (PNS). In reaction to neuronal damage, glial cells proliferate, change their morphology and alter their behaviour; this response is associated with glial cells’ regenerative function and is necessary to preserve the structural integrity and function of the nervous system. This study identifies a role for the Decapentaplegic and Hedgehog pathways in controlling the glial response.
Collapse
|
12
|
Monje PV, Deng L, Xu XM. Human Schwann Cell Transplantation for Spinal Cord Injury: Prospects and Challenges in Translational Medicine. Front Cell Neurosci 2021; 15:690894. [PMID: 34220455 PMCID: PMC8249939 DOI: 10.3389/fncel.2021.690894] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Accepted: 05/21/2021] [Indexed: 01/18/2023] Open
Abstract
The benefits of transplanting cultured Schwann cells (SCs) for the treatment of spinal cord injury (SCI) have been systematically investigated in experimental animals since the early 1990s. Importantly, human SC (hSC) transplantation for SCI has advanced to clinical testing and safety has been established via clinical trials conducted in the USA and abroad. However, multiple barriers must be overcome to enable accessible and effective treatments for SCI patients. This review presents available information on hSC transplantation for SCI with the intention to uncover gaps in our knowledge and discuss areas for future development. To this end, we introduce the historical progression of the work that supports existing and prospective clinical initiatives and explain the reasons for the choice of hSCs while also addressing their limitations as cell therapy products. A search of the relevant literature revealed that rat SCs have served as a preclinical model of reference since the onset of investigations, and that hSC transplants are relatively understudied, possibly due to the sophisticated resources and expertise needed for the traditional processing of hSC cultures from human nerves. In turn, we reason that additional experimentation and a reexamination of the available data are needed to understand the therapeutic value of hSC transplants taking into consideration that the manufacturing of the hSCs themselves may require further development for extended uses in basic research and clinical settings.
Collapse
Affiliation(s)
- Paula V. Monje
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Lingxiao Deng
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Xiao-Ming Xu
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
13
|
Siddiqui AM, Brunner R, Harris GM, Miller AL, Waletzki BE, Schmeichel AM, Schwarzbauer JE, Schwartz J, Yaszemski MJ, Windebank AJ, Madigan NN. Promoting Neuronal Outgrowth Using Ridged Scaffolds Coated with Extracellular Matrix Proteins. Biomedicines 2021; 9:biomedicines9050479. [PMID: 33925613 PMCID: PMC8146557 DOI: 10.3390/biomedicines9050479] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 04/22/2021] [Accepted: 04/23/2021] [Indexed: 12/25/2022] Open
Abstract
Spinal cord injury (SCI) results in cell death, demyelination, and axonal loss. The spinal cord has a limited ability to regenerate, and current clinical therapies for SCI are not effective in helping promote neurologic recovery. We have developed a novel scaffold biomaterial that is fabricated from the biodegradable hydrogel oligo(poly(ethylene glycol)fumarate) (OPF). We have previously shown that positively charged OPF scaffolds (OPF+) in an open spaced, multichannel design can be loaded with Schwann cells to support axonal generation and functional recovery following SCI. We have now developed a hybrid OPF+ biomaterial that increases the surface area available for cell attachment and that contains an aligned microarchitecture and extracellular matrix (ECM) proteins to better support axonal regeneration. OPF+ was fabricated as 0.08 mm thick sheets containing 100 μm high polymer ridges that self-assemble into a spiral shape when hydrated. Laminin, fibronectin, or collagen I coating promoted neuron attachment and axonal outgrowth on the scaffold surface. In addition, the ridges aligned axons in a longitudinal bipolar orientation. Decreasing the space between the ridges increased the number of cells and neurites aligned in the direction of the ridge. Schwann cells seeded on laminin coated OPF+ sheets aligned along the ridges over a 6-day period and could myelinate dorsal root ganglion neurons over 4 weeks. This novel scaffold design, with closer spaced ridges and Schwann cells, is a novel biomaterial construct to promote regeneration after SCI.
Collapse
Affiliation(s)
- Ahad M. Siddiqui
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA; (A.M.S.); (A.M.S.); (A.J.W.)
| | - Rosa Brunner
- Program in Human Medicine, Paracelsus Medical University Salzburg, 5020 Salzburg, Austria;
| | - Gregory M. Harris
- Department of Molecular Biology, Princeton University, Princeton, NJ 08540, USA; (G.M.H.); (J.E.S.)
| | - Alan Lee Miller
- Department of Orthopaedic Surgery, Mayo Clinic, Rochester, MN 55905, USA; (A.L.M.II); (B.E.W.)
| | - Brian E. Waletzki
- Department of Orthopaedic Surgery, Mayo Clinic, Rochester, MN 55905, USA; (A.L.M.II); (B.E.W.)
| | - Ann M. Schmeichel
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA; (A.M.S.); (A.M.S.); (A.J.W.)
| | - Jean E. Schwarzbauer
- Department of Molecular Biology, Princeton University, Princeton, NJ 08540, USA; (G.M.H.); (J.E.S.)
| | - Jeffrey Schwartz
- Department of Chemistry, Princeton University, Princeton, NJ 08544, USA; (J.S.); (M.J.Y.)
| | - Michael J. Yaszemski
- Department of Chemistry, Princeton University, Princeton, NJ 08544, USA; (J.S.); (M.J.Y.)
| | - Anthony J. Windebank
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA; (A.M.S.); (A.M.S.); (A.J.W.)
| | - Nicolas N. Madigan
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA; (A.M.S.); (A.M.S.); (A.J.W.)
- Correspondence:
| |
Collapse
|
14
|
Combined Use of Chitosan and Olfactory Mucosa Mesenchymal Stem/Stromal Cells to Promote Peripheral Nerve Regeneration In Vivo. Stem Cells Int 2021; 2021:6613029. [PMID: 33488738 PMCID: PMC7801080 DOI: 10.1155/2021/6613029] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 11/27/2020] [Accepted: 12/10/2020] [Indexed: 12/20/2022] Open
Abstract
Peripheral nerve injury remains a clinical challenge with severe physiological and functional consequences. Despite the existence of multiple possible therapeutic approaches, until now, there is no consensus regarding the advantages of each option or the best methodology in promoting nerve regeneration. Regenerative medicine is a promise to overcome this medical limitation, and in this work, chitosan nerve guide conduits and olfactory mucosa mesenchymal stem/stromal cells were applied in different therapeutic combinations to promote regeneration in sciatic nerves after neurotmesis injury. Over 20 weeks, the intervened animals were subjected to a regular functional assessment (determination of motor performance, nociception, and sciatic indexes), and after this period, they were evaluated kinematically and the sciatic nerves and cranial tibial muscles were evaluated stereologically and histomorphometrically, respectively. The results obtained allowed confirming the beneficial effects of using these therapeutic approaches. The use of chitosan NGCs and cells resulted in better motor performance, better sciatic indexes, and lower gait dysfunction after 20 weeks. The use of only NGGs demonstrated better nociceptive recoveries. The stereological evaluation of the sciatic nerve revealed identical values in the different parameters for all therapeutic groups. In the muscle histomorphometric evaluation, the groups treated with NGCs and cells showed results close to those of the group that received traditional sutures, the one with the best final values. The therapeutic combinations studied show promising outcomes and should be the target of new future works to overcome some irregularities found in the results and establish the combination of nerve guidance conduits and olfactory mucosa mesenchymal stem/stromal cells as viable options in the treatment of peripheral nerves after injury.
Collapse
|
15
|
Deng LX, Liu NK, Wen RN, Yang SN, Wen X, Xu XM. Laminin-coated multifilament entubulation, combined with Schwann cells and glial cell line-derived neurotrophic factor, promotes unidirectional axonal regeneration in a rat model of thoracic spinal cord hemisection. Neural Regen Res 2021; 16:186-191. [PMID: 32788475 PMCID: PMC7818857 DOI: 10.4103/1673-5374.289436] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Biomaterial bridging provides physical substrates to guide axonal growth across the lesion. To achieve efficient directional guidance, combinatory strategies using permissive matrix, cells and trophic factors are necessary. In the present study, we evaluated permissive effect of poly (acrylonitrile-co-vinyl chloride) guidance channels filled by different densities of laminin-precoated unidirectional polypropylene filaments combined with Schwann cells, and glial cell line-derived neurotrophic factor for axonal regeneration through a T10 hemisected spinal cord gap in adult rats. We found that channels with filaments significantly reduced the lesion cavity, astrocytic gliosis, and inflammatory responses at the graft-host boundaries. The laminin coated low density filament provided the most favorable directional guidance for axonal regeneration which was enhanced by co-grafting of Schwann cells and glial cell line-derived neurotrophic factor. These results demonstrate that the combinatorial strategy of filament-filled guiding scaffold, adhesive molecular laminin, Schwann cells, and glial cell line-derived neurotrophic factor, provides optimal topographical cues in stimulating directional axonal regeneration following spinal cord injury. This study was approved by Indiana University Institutional Animal Care and Use Committees (IACUC #:11011) on October 29, 2015.
Collapse
Affiliation(s)
- Ling-Xiao Deng
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute; Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Nai-Kui Liu
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute; Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Ryan Ning Wen
- Maggie L. Walker Governor's School, Richmond, VA, USA
| | - Shuang-Ni Yang
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute; Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Xuejun Wen
- Institute for Engineering and Medicine, Department of Chemical and Life Science Engineering, Virginia Commonwealth University, Richmond, VA, USA
| | - Xiao-Ming Xu
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute; Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
16
|
Sahib S, Sharma A, Menon PK, Muresanu DF, Castellani RJ, Nozari A, Lafuente JV, Bryukhovetskiy I, Tian ZR, Patnaik R, Buzoianu AD, Wiklund L, Sharma HS. Cerebrolysin enhances spinal cord conduction and reduces blood-spinal cord barrier breakdown, edema formation, immediate early gene expression and cord pathology after injury. PROGRESS IN BRAIN RESEARCH 2020; 258:397-438. [PMID: 33223040 DOI: 10.1016/bs.pbr.2020.09.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Spinal cord evoked potentials (SCEP) are good indicators of spinal cord function in health and disease. Disturbances in SCEP amplitudes and latencies during spinal cord monitoring predict spinal cord pathology following trauma. Treatment with neuroprotective agents preserves SCEP and reduces cord pathology after injury. The possibility that cerebrolysin, a balanced composition of neurotrophic factors improves spinal cord conduction, attenuates blood-spinal cord barrier (BSCB) disruption, edema formation, and cord pathology was examined in spinal cord injury (SCI). SCEP is recorded from epidural space over rat spinal cord T9 and T12 segments after peripheral nerves stimulation. SCEP consists of a small positive peak (MPP), followed by a prominent negative peak (MNP) that is stable before SCI. A longitudinal incision (2mm deep and 5mm long) into the right dorsal horn (T10 and T11 segments) resulted in an immediate long-lasting depression of the rostral MNP with an increase in the latencies. Pretreatment with either cerebrolysin (CBL 5mL/kg, i.v. 30min before) alone or TiO2 nanowired delivery of cerebrolysin (NWCBL 2.5mL/kg, i.v.) prevented the loss of MNP amplitude and even enhanced further from the pre-injury level after SCI without affecting latencies. At 5h, SCI induced edema, BSCB breakdown, and cell injuries were significantly reduced by CBL and NWCBL pretreatment. Interestingly this effect on SCEP and cord pathology was still prominent when the NWCBL was delivered 2min after SCI. Moreover, expressions of c-fos and c-jun genes that are prominent at 5h in untreated SCI are also considerably reduced by CBL and NWCBL treatment. These results are the first to show that CBL and NWCBL enhanced SCEP activity and thwarted the development of cord pathology after SCI. Furthermore, NWCBL in low doses has superior neuroprotective effects on SCEP and cord pathology, not reported earlier. The functional significance and future clinical potential of CBL and NWCBL in SCI are discussed.
Collapse
Affiliation(s)
- Seaab Sahib
- Department of Chemistry & Biochemistry, University of Arkansas, Fayetteville, AR, United States
| | - Aruna Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| | - Preeti K Menon
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden; Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Dafin F Muresanu
- Department of Clinical Neurosciences, University of Medicine & Pharmacy, Cluj-Napoca, Romania; "RoNeuro" Institute for Neurological Research and Diagnostic, Cluj-Napoca, Romania
| | - Rudy J Castellani
- Department of Pathology, University of Maryland, Baltimore, MD, United States
| | - Ala Nozari
- Anesthesiology & Intensive Care, Massachusetts General Hospital, Boston, MA, United States
| | - José Vicente Lafuente
- LaNCE, Department of Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
| | - Igor Bryukhovetskiy
- Department of Fundamental Medicine, School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia; Laboratory of Pharmacology, National Scientific Center of Marine Biology, Far East Branch of the Russian Academy of Sciences, Vladivostok, Russia
| | - Z Ryan Tian
- Department of Chemistry & Biochemistry, University of Arkansas, Fayetteville, AR, United States
| | - Ranjana Patnaik
- Department of Biomaterials, School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University, Varanasi, India
| | - Anca D Buzoianu
- Department of Clinical Pharmacology and Toxicology, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Lars Wiklund
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden
| | - Hari Shanker Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
17
|
Diabetes Mellitus-Related Dysfunction of the Motor System. Int J Mol Sci 2020; 21:ijms21207485. [PMID: 33050583 PMCID: PMC7589125 DOI: 10.3390/ijms21207485] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 10/08/2020] [Accepted: 10/08/2020] [Indexed: 12/20/2022] Open
Abstract
Although motor deficits in humans with diabetic neuropathy have been extensively researched, its effect on the motor system is thought to be lesser than that on the sensory system. Therefore, motor deficits are considered to be only due to sensory and muscle impairment. However, recent clinical and experimental studies have revealed that the brain and spinal cord, which are involved in the motor control of voluntary movement, are also affected by diabetes. This review focuses on the most important systems for voluntary motor control, mainly the cortico-muscular pathways, such as corticospinal tract and spinal motor neuron abnormalities. Specifically, axonal damage characterized by the proximodistal phenotype occurs in the corticospinal tract and motor neurons with long axons, and the transmission of motor commands from the brain to the muscles is impaired. These findings provide a new perspective to explain motor deficits in humans with diabetes. Finally, pharmacological and non-pharmacological treatment strategies for these disorders are presented.
Collapse
|
18
|
Schwann Cell Cultures: Biology, Technology and Therapeutics. Cells 2020; 9:cells9081848. [PMID: 32781699 PMCID: PMC7465416 DOI: 10.3390/cells9081848] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 08/01/2020] [Accepted: 08/05/2020] [Indexed: 12/14/2022] Open
Abstract
Schwann cell (SC) cultures from experimental animals and human donors can be prepared using nearly any type of nerve at any stage of maturation to render stage- and patient-specific populations. Methods to isolate, purify, expand in number, and differentiate SCs from adult, postnatal and embryonic sources are efficient and reproducible as these have resulted from accumulated refinements introduced over many decades of work. Albeit some exceptions, SCs can be passaged extensively while maintaining their normal proliferation and differentiation controls. Due to their lineage commitment and strong resistance to tumorigenic transformation, SCs are safe for use in therapeutic approaches in the peripheral and central nervous systems. This review summarizes the evolution of work that led to the robust technologies used today in SC culturing along with the main features of the primary and expanded SCs that make them irreplaceable models to understand SC biology in health and disease. Traditional and emerging approaches in SC culture are discussed in light of their prospective applications. Lastly, some basic assumptions in vitro SC models are identified in an attempt to uncover the combined value of old and new trends in culture protocols and the cellular products that are derived.
Collapse
|
19
|
Sadik ME, Ozturk AK, Albayar A, Branche M, Sullivan PZ, Schlosser LO, Browne KD, Jaye AH, Smith DH. A Strategy Toward Bridging a Complete Spinal Cord Lesion Using Stretch-Grown Axons. Tissue Eng Part A 2020; 26:623-635. [DOI: 10.1089/ten.tea.2019.0230] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Affiliation(s)
- Mindy Ezra Sadik
- Center for Brain Injury and Repair, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ali K. Ozturk
- Department of Neurosurgery, Pennsylvania Hospital, University of Pennsylvania Health System, Philadelphia, Pennsylvania
| | - Ahmed Albayar
- Center for Brain Injury and Repair, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Marc Branche
- Center for Brain Injury and Repair, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Patricia Zadnik Sullivan
- Department of Neurosurgery, Pennsylvania Hospital, University of Pennsylvania Health System, Philadelphia, Pennsylvania
| | - Laura O. Schlosser
- Center for Brain Injury and Repair, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Kevin D. Browne
- Center for Brain Injury and Repair, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Andrew H. Jaye
- Center for Brain Injury and Repair, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Douglas H. Smith
- Center for Brain Injury and Repair, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
20
|
Han Q, Xie Y, Ordaz JD, Huh AJ, Huang N, Wu W, Liu N, Chamberlain KA, Sheng ZH, Xu XM. Restoring Cellular Energetics Promotes Axonal Regeneration and Functional Recovery after Spinal Cord Injury. Cell Metab 2020; 31:623-641.e8. [PMID: 32130884 PMCID: PMC7188478 DOI: 10.1016/j.cmet.2020.02.002] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 11/24/2019] [Accepted: 01/31/2020] [Indexed: 01/30/2023]
Abstract
Axonal regeneration in the central nervous system (CNS) is a highly energy-demanding process. Extrinsic insults and intrinsic restrictions lead to an energy crisis in injured axons, raising the question of whether recovering energy deficits facilitates regeneration. Here, we reveal that enhancing axonal mitochondrial transport by deleting syntaphilin (Snph) recovers injury-induced mitochondrial depolarization. Using three CNS injury mouse models, we demonstrate that Snph-/- mice display enhanced corticospinal tract (CST) regeneration passing through a spinal cord lesion, accelerated regrowth of monoaminergic axons across a transection gap, and increased compensatory sprouting of uninjured CST. Notably, regenerated CST axons form functional synapses and promote motor functional recovery. Administration of the bioenergetic compound creatine boosts CST regenerative capacity in Snph-/- mice. Our study provides mechanistic insights into intrinsic regeneration failure in CNS and suggests that enhancing mitochondrial transport and cellular energetics are promising strategies to promote regeneration and functional restoration after CNS injuries.
Collapse
Affiliation(s)
- Qi Han
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Yuxiang Xie
- Synaptic Function Section, The Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Josue D Ordaz
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Andrew J Huh
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Ning Huang
- Synaptic Function Section, The Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Wei Wu
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Naikui Liu
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Kelly A Chamberlain
- Synaptic Function Section, The Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Zu-Hang Sheng
- Synaptic Function Section, The Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Xiao-Ming Xu
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| |
Collapse
|
21
|
Griffin JM, Bradke F. Therapeutic repair for spinal cord injury: combinatory approaches to address a multifaceted problem. EMBO Mol Med 2020; 12:e11505. [PMID: 32090481 PMCID: PMC7059014 DOI: 10.15252/emmm.201911505] [Citation(s) in RCA: 110] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 01/07/2020] [Accepted: 01/31/2020] [Indexed: 12/21/2022] Open
Abstract
The recent years saw the advent of promising preclinical strategies that combat the devastating effects of a spinal cord injury (SCI) that are progressing towards clinical trials. However, individually, these treatments produce only modest levels of recovery in animal models of SCI that could hamper their implementation into therapeutic strategies in spinal cord injured humans. Combinational strategies have demonstrated greater beneficial outcomes than their individual components alone by addressing multiple aspects of SCI pathology. Clinical trial designs in the future will eventually also need to align with this notion. The scenario will become increasingly complex as this happens and conversations between basic researchers and clinicians are required to ensure accurate study designs and functional readouts.
Collapse
Affiliation(s)
- Jarred M Griffin
- Laboratory for Axonal Growth and Regeneration, German Centre for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Frank Bradke
- Laboratory for Axonal Growth and Regeneration, German Centre for Neurodegenerative Diseases (DZNE), Bonn, Germany
| |
Collapse
|
22
|
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: 9.5] [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
|
23
|
Li X, Zhou D, Jin Z, Chen H, Wang X, Zhang X, Xu T. A coaxially extruded heterogeneous core- shell fiber with Schwann cells and neural stem cells. Regen Biomater 2019; 7:131-139. [PMID: 32296532 PMCID: PMC7147360 DOI: 10.1093/rb/rbz037] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 09/05/2019] [Accepted: 10/03/2019] [Indexed: 12/29/2022] Open
Abstract
Cellular therapies play a critical role in the treatment of spinal cord injury (SCI). Compared with cell-seeded conduits, fully cellular grafts have more similarities with autografts, and thus might result in better regeneration effects. In this study, we fabricated Schwann cell (SC)-neural stem cell (NSC) core–shell alginate hydrogel fibers in a coaxial extrusion manner. The rat SC line RSC96 and mouse NSC line NE-4C were used in this experiment. Fully cellular components were achieved in the core portion and the relative spatial positions of these two cells partially mimic the construction of nerve fibers in vivo. SCs were demonstrated to express more genes of neurotrophic factors in alginate shell. Enhanced proliferation and differentiation tendency of NSCs was observed when they were co-cultured with SCs. This model has strong potential for application in SCI repair.
Collapse
Affiliation(s)
- Xinda Li
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Department of Mechanical Engineering, Tsinghua University, Beijing 100084, People's Republic of China.,Key Laboratory for Advanced Materials Processing Technology, Ministry of Education, Department of Mechanical Engineering, Tsinghua University, Beijing 100084, People's Republic of China
| | - Dezhi Zhou
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Department of Mechanical Engineering, Tsinghua University, Beijing 100084, People's Republic of China.,Key Laboratory for Advanced Materials Processing Technology, Ministry of Education, Department of Mechanical Engineering, Tsinghua University, Beijing 100084, People's Republic of China
| | - Zhizhong Jin
- Department of Neurosurgery, the First Hospital of China Medical University, Shenyang 110122, People's Republic of China
| | - Hongqing Chen
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, People's Republic of China
| | - Xuanzhi Wang
- Department of Neurosurgery, the First Affiliated Hospital of Wannan Medical College, Wuhu 241001, People's Republic of China
| | - Xinzhi Zhang
- East China Institute of Digital Medical Engineering, Shangrao 334000, People's Republic of China.,Medprin Regenerative Medical Technologies Co., Ltd, Shenzhen 518102, People's Republic of China
| | - Tao Xu
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Department of Mechanical Engineering, Tsinghua University, Beijing 100084, People's Republic of China.,Key Laboratory for Advanced Materials Processing Technology, Ministry of Education, Department of Mechanical Engineering, Tsinghua University, Beijing 100084, People's Republic of China.,Department of Precision Medicine and Healthcare, Tsinghua Berkeley Shenzhen Institute, Shenzhen 518055, People's Republic of China
| |
Collapse
|
24
|
Abstract
Biomaterials can be utilized to assist in the transplantation of Schwann cells to the central and peripheral nervous system. The biomaterials can be natural or man-made, and can have preformed shapes or injectable formats. Biomaterials can play multiple roles in cellular transplantation; for example, they can assist with cellular integration and protect Schwann cells from cell death initiated by the lack of a substrate, an occurrence known as "anoikis." In addition, biomaterials can be engineered to increase cell proliferation and differentiation by the addition of ligands bound to the substrate. Here, we describe the incorporation of Schwann cells to both man-made and natural matrices for in vitro and in vivo measures relevant to Schwann cell transplantation strategies.
Collapse
|
25
|
Cerqueira SR, Lee YS, Cornelison RC, Mertz MW, Wachs RA, Schmidt CE, Bunge MB. Decellularized peripheral nerve supports Schwann cell transplants and axon growth following spinal cord injury. Biomaterials 2018; 177:176-185. [PMID: 29929081 PMCID: PMC6034707 DOI: 10.1016/j.biomaterials.2018.05.049] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 05/25/2018] [Accepted: 05/28/2018] [Indexed: 01/10/2023]
Abstract
Schwann cell (SC) transplantation has been comprehensively studied as a strategy for spinal cord injury (SCI) repair. SCs are neuroprotective and promote axon regeneration and myelination. Nonetheless, substantial SC death occurs post-implantation, which limits therapeutic efficacy. The use of extracellular matrix (ECM)-derived matrices, such as Matrigel, supports transplanted SC survival and axon growth, resulting in improved motor function. Because appropriate matrices are needed for clinical translation, we test here the use of an acellular injectable peripheral nerve (iPN) matrix. Implantation of SCs in iPN into a contusion lesion did not alter immune cell infiltration compared to injury only controls. iPN implants were larger and contained twice as many SC-myelinated axons as Matrigel grafts. SC/iPN animals performed as well as the SC/Matrigel group in the BBB locomotor test, and made fewer errors on the grid walk at 4 weeks, equalizing at 8 weeks. The fact that this clinically relevant iPN matrix is immunologically tolerated and supports SC survival and axon growth within the graft offers a highly translational possibility for improving efficacy of SC treatment after SCI. To our knowledge, it is the first time that an injectable PN matrix is being evaluated to improve the efficacy of SC transplantation in SCI repair.
Collapse
Affiliation(s)
- Susana R Cerqueira
- The Miami Project to Cure Paralysis, University of Miami, Miller School of Medicine, Miami, FL, USA.
| | - Yee-Shuan Lee
- The Miami Project to Cure Paralysis, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Robert C Cornelison
- Department of Chemical Engineering, University of Texas at Austin, Austin, TX, USA
| | - Michaela W Mertz
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Rebecca A Wachs
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Christine E Schmidt
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Mary Bartlett Bunge
- The Miami Project to Cure Paralysis, University of Miami, Miller School of Medicine, Miami, FL, USA; Department of Cell Biology, University of Miami, Miller School of Medicine, Miami, FL, USA; Department of Neurological Surgery, University of Miami, Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|
26
|
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: 7.2] [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
|
27
|
Jin Y, Shumsky JS, Fischer I. Axonal regeneration of different tracts following transplants of human glial restricted progenitors into the injured spinal cord in rats. Brain Res 2018; 1686:101-112. [PMID: 29408659 DOI: 10.1016/j.brainres.2018.01.030] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 01/18/2018] [Accepted: 01/21/2018] [Indexed: 12/15/2022]
Abstract
The goal of this study was to compare the efficacy of human glial restricted progenitors (hGRPs) in promoting axonal growth of different tracts. We examined the potential of hGRPs grafted into a cervical (C4) dorsal column lesion to test sensory axons, and into a C4 hemisection to test motor tracts. The hGRPs, thawed from frozen stocks, were suspended in a PureCol matrix and grafted acutely into a C4 dorsal column or hemisection lesion. Control rats received PureCol only. Five weeks after transplantation, all transplanted cells survived in rats with the dorsal column lesion but only about half of the grafts in the hemisection. In the dorsal column lesion group, few sensory axons grew short distances into the lesion site of control animals. The presence of hGRPs transplants enhanced axonal growth significantly farther into the transplants. In the hemisection group, coerulospinal axons extended similarly into both control and transplant groups with no enhancement by the presence of hGRPs. Rubrospinal axons did not grow into the lesion even in the presence of hGRPs. However, reticulospinal and raphespinal axons grew for a significantly longer distance into the transplants. These results demonstrate the differential capacity of axonal growth/regeneration of the motor and sensory tracts based on their intrinsic abilities as well as their response to the modified environment induced by the hGRPs transplants. We conclude that hGRP transplants can modify the injury site for axon growth of sensory and some motor tracts, and suggest they could be combined with other interventions to restore connectivity.
Collapse
Affiliation(s)
- Ying Jin
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, USA.
| | - Jed S Shumsky
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, USA
| | - Itzhak Fischer
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, USA
| |
Collapse
|
28
|
Novikova LN, Kolar MK, Kingham PJ, Ullrich A, Oberhoffner S, Renardy M, Doser M, Müller E, Wiberg M, Novikov LN. Trimethylene carbonate-caprolactone conduit with poly-p-dioxanone microfilaments to promote regeneration after spinal cord injury. Acta Biomater 2018; 66:177-191. [PMID: 29174588 DOI: 10.1016/j.actbio.2017.11.028] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 11/09/2017] [Accepted: 11/17/2017] [Indexed: 11/25/2022]
Abstract
Spinal cord injury (SCI) is often associated with scarring and cavity formation and therefore bridging strategies are essential to provide a physical substrate for axonal regeneration. In this study we investigated the effects of a biodegradable conduit made from trimethylene carbonate and ε-caprolactone (TC) containing poly-p-dioxanone microfilaments (PDO) with longitudinal grooves on regeneration after SCI in adult rats. In vitro studies demonstrated that different cell types including astrocytes, meningeal fibroblasts, Schwann cells and adult sensory dorsal root ganglia neurons can grow on the TC and PDO material. For in vivo experiments, the TC/PDO conduit was implanted into a small 2-3 mm long cavity in the C3-C4 cervical segments immediately after injury (acute SCI) or at 2-5 months after initial surgery (chronic SCI). At 8 weeks after implantation into acute SCI, numerous 5HT-positive descending raphaespinal axons and sensory CGRP-positive axons regenerated across the conduit and were often associated with PDO microfilaments and migrated host cells. Implantation into chronically injured SCI induced regeneration mainly of the sensory CGRP-positive axons. Although the conduit had no effect on the density of OX42-positive microglial cells when compared with SCI control, the activity of GFAP-positive astrocytes was reduced. The results suggest that a TC/PDO conduit can support axonal regeneration after acute and chronic SCI even without addition of exogenous glial or stem cells. STATEMENT OF SIGNIFICANCE Biosynthetic conduits can support regeneration after spinal cord injury but often require addition of cell therapy and neurotrophic factors. This study demonstrates that biodegradable conduits made from trimethylene carbonate and ε-caprolactone with poly-p-dioxanone microfilaments alone can promote migration of different host cells and stimulate axonal regeneration after implantation into acute and chronic spinal cord injury. These results can be used to develop biosynthetic conduits for future clinical applications.
Collapse
|
29
|
Liu S, Schackel T, Weidner N, Puttagunta R. Biomaterial-Supported Cell Transplantation Treatments for Spinal Cord Injury: Challenges and Perspectives. Front Cell Neurosci 2018; 11:430. [PMID: 29375316 PMCID: PMC5768640 DOI: 10.3389/fncel.2017.00430] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 12/20/2017] [Indexed: 12/17/2022] Open
Abstract
Spinal cord injury (SCI), resulting in para- and tetraplegia caused by the partial or complete disruption of descending motor and ascending sensory neurons, represents a complex neurological condition that remains incurable. Following SCI, numerous obstacles comprising of the loss of neural tissue (neurons, astrocytes, and oligodendrocytes), formation of a cavity, inflammation, loss of neuronal circuitry and function must be overcome. Given the multifaceted primary and secondary injury events that occur with SCI treatment options are likely to require combinatorial therapies. While several methods have been explored, only the intersection of two, cell transplantation and biomaterial implantation, will be addressed in detail here. Owing to the constant advance of cell culture technologies, cell-based transplantation has come to the forefront of SCI treatment in order to replace/protect damaged tissue and provide physical as well as trophic support for axonal regrowth. Biomaterial scaffolds provide cells with a protected environment from the surrounding lesion, in addition to bridging extensive damage and providing physical and directional support for axonal regrowth. Moreover, in this combinatorial approach cell transplantation improves scaffold integration and therefore regenerative growth potential. Here, we review the advances in combinatorial therapies of Schwann cells (SCs), astrocytes, olfactory ensheathing cells (OECs), mesenchymal stem cells, as well as neural stem and progenitor cells (NSPCs) with various biomaterial scaffolds.
Collapse
Affiliation(s)
- Shengwen Liu
- Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Thomas Schackel
- Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| | - Norbert Weidner
- Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| | - Radhika Puttagunta
- Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
30
|
Abstract
Adult Schwann cells (SCs) can provide both a permissive substrate for axonal growth and a source of cells to ensheath and myelinate axons when transplanted into the injured spinal cord. Multiple studies have demonstrated that SC transplants can be used as part of a combinatorial approach to repairing the injured spinal cord. Here, we describe the protocols for collection and transplantation of adult rat primary SCs into the injured spinal cord. Protocols are included for the tissue culture procedures necessary for collection, quantification, and suspension of the cells for transplantation and for the surgical procedures for spinal cord injury at thoracic level nine (T9), reexposure of the injury site for delayed transplantation, and injection of the cells into the spinal cord.
Collapse
Affiliation(s)
- Ying Dai
- Burke Medical Research Institute, White Plains, NY, USA.,Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Caitlin E Hill
- Burke Medical Research Institute, White Plains, NY, USA. .,Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
31
|
|
32
|
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: 1.0] [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
|
33
|
Belin S, Zuloaga KL, Poitelon Y. Influence of Mechanical Stimuli on Schwann Cell Biology. Front Cell Neurosci 2017; 11:347. [PMID: 29209171 PMCID: PMC5701625 DOI: 10.3389/fncel.2017.00347] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 10/19/2017] [Indexed: 12/05/2022] Open
Abstract
Schwann cells are the glial cells of the peripheral nervous system (PNS). They insulate axons by forming a specialized extension of plasma membrane called the myelin sheath. The formation of myelin is essential for the rapid saltatory propagation of action potentials and to maintain the integrity of axons. Although both axonal and extracellular matrix (ECM) signals are necessary for myelination to occur, the cellular and molecular mechanisms regulating myelination continue to be elucidated. Schwann cells in peripheral nerves are physiologically exposed to mechanical stresses (i.e., tensile, compressive and shear strains), occurring during development, adulthood and injuries. In addition, there is a growing body of evidences that Schwann cells are sensitive to the stiffness of their environment. In this review, we detail the mechanical constraints of Schwann cells and peripheral nerves. We explore the regulation of Schwann cell signaling pathways in response to mechanical stimulation. Finally, we provide a comprehensive overview of the experimental studies addressing the mechanobiology of Schwann cells. Understanding which mechanical properties can interfere with the cellular and molecular biology of Schwann cell during development, myelination and following injuries opens new insights in the regulation of PNS development and treatment approaches in peripheral neuropathies.
Collapse
Affiliation(s)
- Sophie Belin
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY, United States
| | - Kristen L. Zuloaga
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY, United States
| | - Yannick Poitelon
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY, United States
| |
Collapse
|
34
|
AAV-KLF7 Promotes Descending Propriospinal Neuron Axonal Plasticity after Spinal Cord Injury. Neural Plast 2017; 2017:1621629. [PMID: 28884027 PMCID: PMC5572611 DOI: 10.1155/2017/1621629] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 05/27/2017] [Accepted: 06/12/2017] [Indexed: 01/16/2023] Open
Abstract
DPSN axons mediate and maintain a variety of normal spinal functions. Unsurprisingly, DPSN tracts have been shown to mediate functional recovery following SCI. KLF7 could contribute to CST axon plasticity after spinal cord injury. In the present study, we assessed whether KLF7 could effectively promote DPSN axon regeneration and synapse formation following SCI. An AAV-KLF7 construct was used to overexpress KLF7. In vitro, KLF7 and target proteins were successfully elevated and axonal outgrowth was enhanced. In vivo, young adult C57BL/6 mice received a T10 contusion followed by an AAV-KLF7 injection at the T7–9 levels above the lesion. Five weeks later, overexpression of KLF7 was expressed in DPSN. KLF7 and KLF7 target genes (NGF, TrkA, GAP43, and P0) were detectably increased in the injured spinal cord. Myelin sparring at the lesion site, DPSN axonal regeneration and synapse formation, muscle weight, motor endplate morphology, and functional parameters were all additionally improved by KLF7 treatment. Our findings suggest that KLF7 promotes DPSN axonal plasticity and the formation of synapses with motor neurons at the caudal spinal cord, leading to improved functional recovery and further supporting the potential of AAV-KLF7 as a therapeutic agent for spinal cord injury.
Collapse
|
35
|
Tuszynski MH, Weidner N, McCormack M, Miller I, Powell H, Conner J. Grafts of Genetically Modified Schwann Cells to the Spinal Cord: Survival, Axon Growth, and Myelination. Cell Transplant 2017; 7:187-96. [PMID: 9588600 DOI: 10.1177/096368979800700213] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Schwann cells naturally support axonal regeneration after injury in the peripheral nervous system, and have also shown a significant, albeit limited, ability to support axonal growth and remyelination after grafting to the central nervous system (CNS). It is possible that Schwann cell-induced axonal growth in the CNS could be substantially increased by genetic manipulation to secrete augmented amounts of neurotrophic factors. To test this hypothesis, cultured primary adult rat Schwann cells were genetically modified using retroviral vectors to produce and secrete high levels of human nerve growth factor (NGF). These cells were then grafted to the midthoracic spinal cords of adult rats. Findings were compared to animals that received grafts of nontransduced Schwann cells. Spinal cord lesions were not placed prior to grafting because the primary aim of this study was to examine features of grafted Schwann cell survival, growth, and effects on host axons. In vitro prior to grafting, Schwann cells secreted 1.5 + 0.1 ng human NGF/ml/106 cells/day. Schwann cell transplants readily survived for 2 wk to 1 yr after in vivo placement. Some NGF-transduced grafts slowly increased in size over time compared to nontransduced grafts; the latter remained stable in size. NGF-transduced transplants were densely penetrated by primary sensory nociceptive axons originating from the dorsolateral fasciculus of the spinal cord, whereas control grafts showed significantly fewer penetrating sensory axons. Over time, Schwann cell grafts also became penetrated by TH- and DBH-labeled axons of putative coerulospinal origin, unlike control cell grafts. Ultrastructurally, axons in both graft types were extensively myelinated by Schwann cells. Grafted animals showed no changes in gross locomotor function. In vivo expression of the human NGF transgene was demonstrated for periods of at least 6 m. These findings demonstrate that primary adult Schwann cells 1) can be transduced to secrete augmented levels of neurotrophic factors, 2) survive grafting to the CNS for prolonged time periods, 3) elicit robust growth of host neurotrophin-responsive axons, 4) myelinate CNS axons, and 5) express the transgene for prolonged time periods in vivo. Some grafts slowly enlarge over time, a feature that may be attributable to the propensity of Schwann cells to immortalize after multiple passages. Transduced Schwann cells merit further study as tools for promoting CNS regeneration.
Collapse
Affiliation(s)
- M H Tuszynski
- Department of Neurosciences, University of California-San Diego, La Jolla 92093-0608, USA
| | | | | | | | | | | |
Collapse
|
36
|
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: 9.1] [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
|
37
|
Blits B, Boer GJ, Verhaagen J. Pharmacological, Cell, and Gene Therapy Strategies to Promote Spinal Cord Regeneration. Cell Transplant 2017. [DOI: 10.3727/000000002783985521] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
In this review, recent studies using pharmacological treatment, cell transplantation, and gene therapy to promote regeneration of the injured spinal cord in animal models will be summarized. Pharmacological and cell transplantation treatments generally revealed some degree of effect on the regeneration of the injured ascending and descending tracts, but further improvements to achieve a more significant functional recovery are necessary. The use of gene therapy to promote repair of the injured nervous system is a relatively new concept. It is based on the development of methods for delivering therapeutic genes to neurons, glia cells, or nonneural cells. Direct in vivo gene transfer or gene transfer in combination with (neuro)transplantation (ex vivo gene transfer) appeared powerful strategies to promote neuronal survival and axonal regrowth following traumatic injury to the central nervous system. Recent advances in understanding the cellular and molecular mechanisms that govern neuronal survival and neurite outgrowth have enabled the design of experiments aimed at viral vector-mediated transfer of genes encoding neurotrophic factors, growth-associated proteins, cell adhesion molecules, and antiapoptotic genes. Central to the success of these approaches was the development of efficient, nontoxic vectors for gene delivery and the acquirement of the appropriate (genetically modified) cells for neurotransplantation. Direct gene transfer in the nervous system was first achieved with herpes viral and E1-deleted adenoviral vectors. Both vector systems are problematic in that these vectors elicit immunogenic and cytotoxic responses. Adeno-associated viral vectors and lentiviral vectors constitute improved gene delivery systems and are beginning to be applied in neuroregeneration research of the spinal cord. Ex vivo approaches were initially based on the implantation of genetically modified fibroblasts. More recently, transduced Schwann cells, genetically modified pieces of peripheral nerve, and olfactory ensheathing glia have been used as implants into the injured spinal cord.
Collapse
Affiliation(s)
- Bas Blits
- Graduate School Neurosciences Amsterdam, Netherlands Institute for Brain Research, Meibergdreef 33, 1105 AZ Amsterdam-ZO, The Netherlands
| | - Gerard J. Boer
- Graduate School Neurosciences Amsterdam, Netherlands Institute for Brain Research, Meibergdreef 33, 1105 AZ Amsterdam-ZO, The Netherlands
| | - Joost Verhaagen
- Graduate School Neurosciences Amsterdam, Netherlands Institute for Brain Research, Meibergdreef 33, 1105 AZ Amsterdam-ZO, The Netherlands
| |
Collapse
|
38
|
Bastidas J, Athauda G, De La Cruz G, Chan WM, Golshani R, Berrocal Y, Henao M, Lalwani A, Mannoji C, Assi M, Otero PA, Khan A, Marcillo AE, Norenberg M, Levi AD, Wood PM, Guest JD, Dietrich WD, Bartlett Bunge M, Pearse DD. Human Schwann cells exhibit long-term cell survival, are not tumorigenic and promote repair when transplanted into the contused spinal cord. Glia 2017; 65:1278-1301. [PMID: 28543541 DOI: 10.1002/glia.23161] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2016] [Revised: 04/07/2017] [Accepted: 04/10/2017] [Indexed: 12/26/2022]
Abstract
The transplantation of rodent Schwann cells (SCs) provides anatomical and functional restitution in a variety of spinal cord injury (SCI) models, supporting the recent translation of SCs to phase 1 clinical trials for human SCI. Whereas human (Hu)SCs have been examined experimentally in a complete SCI transection paradigm, to date the reported behavior of SCs when transplanted after a clinically relevant contusive SCI has been restricted to the use of rodent SCs. Here, in a xenotransplant, contusive SCI paradigm, the survival, biodistribution, proliferation and tumorgenicity as well as host responses to HuSCs, cultured according to a protocol analogous to that developed for clinical application, were investigated. HuSCs persisted within the contused nude rat spinal cord through 6 months after transplantation (longest time examined), exhibited low cell proliferation, displayed no evidence of tumorigenicity and showed a restricted biodistribution to the lesion. Neuropathological examination of the CNS revealed no adverse effects of HuSCs. Animals exhibiting higher numbers of surviving HuSCs within the lesion showed greater volumes of preserved white matter and host rat SC and astrocyte ingress as well as axon ingrowth and myelination. These results demonstrate the safety of HuSCs when employed in a clinically relevant experimental SCI paradigm. Further, signs of a potentially positive influence of HuSC transplants on host tissue pathology were observed. These findings show that HuSCs exhibit a favorable toxicity profile for up to 6 months after transplantation into the contused rat spinal cord, an important outcome for FDA consideration of their use in human clinical trials.
Collapse
Affiliation(s)
- Johana Bastidas
- The Miami Project to Cure Paralysis, The Department of Neurological Surgery, The University of Miami Miller School of Medicine, Miami, Florida, 33136
| | - Gagani Athauda
- The Department of Cellular Biology, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida, 33199.,The Department of Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida, 33199
| | - Gabriela De La Cruz
- Translational Pathology Laboratory, Lineberger Comprehensive Cancer Center, Department of Pathology, University of North Carolina School of Medicine, Chapel Hill, North Carolina, 27599
| | - Wai-Man Chan
- The Miami Project to Cure Paralysis, The Department of Neurological Surgery, The University of Miami Miller School of Medicine, Miami, Florida, 33136
| | - Roozbeh Golshani
- The Miami Project to Cure Paralysis, The Department of Neurological Surgery, The University of Miami Miller School of Medicine, Miami, Florida, 33136
| | - Yerko Berrocal
- The Department of Cellular Biology, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida, 33199.,The Department of Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida, 33199
| | - Martha Henao
- The Miami Project to Cure Paralysis, The Department of Neurological Surgery, The University of Miami Miller School of Medicine, Miami, Florida, 33136
| | - Anil Lalwani
- The Miami Project to Cure Paralysis, The Department of Neurological Surgery, The University of Miami Miller School of Medicine, Miami, Florida, 33136
| | - Chikato Mannoji
- The Department of Orthopedic Surgery, Chiba University School of Medicine, Chiba, Japan
| | - Mazen Assi
- The Miami Project to Cure Paralysis, The Department of Neurological Surgery, The University of Miami Miller School of Medicine, Miami, Florida, 33136
| | - P Anthony Otero
- The Miami Project to Cure Paralysis, The Department of Neurological Surgery, The University of Miami Miller School of Medicine, Miami, Florida, 33136
| | - Aisha Khan
- The Miami Project to Cure Paralysis, The Department of Neurological Surgery, The University of Miami Miller School of Medicine, Miami, Florida, 33136
| | - Alexander E Marcillo
- The Miami Project to Cure Paralysis, The Department of Neurological Surgery, The University of Miami Miller School of Medicine, Miami, Florida, 33136
| | - Michael Norenberg
- The Department of Pathology, The University of Miami Miller School of Medicine, Miami, Florida, 33136
| | - Allan D Levi
- The Miami Project to Cure Paralysis, The Department of Neurological Surgery, The University of Miami Miller School of Medicine, Miami, Florida, 33136.,The Department of Neurological Surgery, The University of Miami Miller School of Medicine, Miami, Florida, 33136
| | - Patrick M Wood
- The Miami Project to Cure Paralysis, The Department of Neurological Surgery, The University of Miami Miller School of Medicine, Miami, Florida, 33136
| | - James D Guest
- The Miami Project to Cure Paralysis, The Department of Neurological Surgery, The University of Miami Miller School of Medicine, Miami, Florida, 33136.,The Department of Neurological Surgery, The University of Miami Miller School of Medicine, Miami, Florida, 33136
| | - W Dalton Dietrich
- The Miami Project to Cure Paralysis, The Department of Neurological Surgery, The University of Miami Miller School of Medicine, Miami, Florida, 33136.,The Department of Neurological Surgery, The University of Miami Miller School of Medicine, Miami, Florida, 33136.,The Department of Neurology, The University of Miami Miller School of Medicine, Miami, Florida, 33136.,The Neuroscience Program, The University of Miami Miller School of Medicine, Miami, Florida, 33136.,The Interdisciplinary Stem Cell Institute, The University of Miami Miller School of Medicine, Miami, Florida, 33136.,The Department of Cell Biology, The University of Miami Miller School of Medicine, Miami, Florida, 33136
| | - Mary Bartlett Bunge
- The Miami Project to Cure Paralysis, The Department of Neurological Surgery, The University of Miami Miller School of Medicine, Miami, Florida, 33136.,The Department of Neurological Surgery, The University of Miami Miller School of Medicine, Miami, Florida, 33136.,The Neuroscience Program, The University of Miami Miller School of Medicine, Miami, Florida, 33136.,The Interdisciplinary Stem Cell Institute, The University of Miami Miller School of Medicine, Miami, Florida, 33136.,The Department of Cell Biology, The University of Miami Miller School of Medicine, Miami, Florida, 33136
| | - Damien D Pearse
- The Miami Project to Cure Paralysis, The Department of Neurological Surgery, The University of Miami Miller School of Medicine, Miami, Florida, 33136.,The Department of Neurological Surgery, The University of Miami Miller School of Medicine, Miami, Florida, 33136.,The Neuroscience Program, The University of Miami Miller School of Medicine, Miami, Florida, 33136.,The Interdisciplinary Stem Cell Institute, The University of Miami Miller School of Medicine, Miami, Florida, 33136.,Bruce W. Carter Department of Veterans Affairs Medical Center, Miami, Florida, 33136
| |
Collapse
|
39
|
Führmann T, Anandakumaran PN, Shoichet MS. Combinatorial Therapies After Spinal Cord Injury: How Can Biomaterials Help? Adv Healthc Mater 2017; 6. [PMID: 28247563 DOI: 10.1002/adhm.201601130] [Citation(s) in RCA: 115] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Revised: 12/05/2016] [Indexed: 12/31/2022]
Abstract
Traumatic spinal cord injury (SCI) results in an immediate loss of motor and sensory function below the injury site and is associated with a poor prognosis. The inhibitory environment that develops in response to the injury is mainly due to local expression of inhibitory factors, scarring and the formation of cystic cavitations, all of which limit the regenerative capacity of endogenous or transplanted cells. Strategies that demonstrate promising results induce a change in the microenvironment at- and around the lesion site to promote endogenous cell repair, including axonal regeneration or the integration of transplanted cells. To date, many of these strategies target only a single aspect of SCI; however, the multifaceted nature of SCI suggests that combinatorial strategies will likely be more effective. Biomaterials are a key component of combinatorial strategies, as they have the potential to deliver drugs locally over a prolonged period of time and aid in cell survival, integration and differentiation. Here we summarize the advantages and limitations of widely used strategies to promote recovery after injury and highlight recent research where biomaterials aided combinatorial strategies to overcome some of the barriers of spinal cord regeneration.
Collapse
Affiliation(s)
- Tobias Führmann
- The Donnelly Centre for Cellular and Biomolecular Research; 160 College Street, Room 514 Toronto ON M5S 3E1 Canada
- Department of Chemical Engineering and Applied Chemistry; 200 College Street Toronto ON M5S 3E5 Canada
| | - Priya N. Anandakumaran
- The Donnelly Centre for Cellular and Biomolecular Research; 160 College Street, Room 514 Toronto ON M5S 3E1 Canada
- Institute of Biomaterials and Biomedical Engineering; 164 College Street Toronto ON M5S 3G9 Canada
| | - Molly S. Shoichet
- The Donnelly Centre for Cellular and Biomolecular Research; 160 College Street, Room 514 Toronto ON M5S 3E1 Canada
- Department of Chemical Engineering and Applied Chemistry; 200 College Street Toronto ON M5S 3E5 Canada
- Institute of Biomaterials and Biomedical Engineering; 164 College Street Toronto ON M5S 3G9 Canada
- Department of Chemistry; University of Toronto; 80 St George St Toronto ON M5S 3H6 Canada
| |
Collapse
|
40
|
Multichannel polymer scaffold seeded with activated Schwann cells and bone mesenchymal stem cells improves axonal regeneration and functional recovery after rat spinal cord injury. Acta Pharmacol Sin 2017; 38:623-637. [PMID: 28392569 DOI: 10.1038/aps.2017.11] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 01/23/2017] [Indexed: 12/17/2022] Open
Abstract
The adult mammalian CNS has a limited capacity to regenerate after traumatic injury. In this study, a combinatorial strategy to promote axonal regeneration and functional recovery after spinal cord injury (SCI) was evaluated in adult rats. The rats were subjected to a complete transection in the thoracic spinal cord, and multichannel scaffolds seeded with activated Schwann cells (ASCs) and/or rat bone marrow-derived mesenchymal stem cells (MSCs) were acutely grafted into the 3-mm-wide transection gap. At 4 weeks post-transplantation and thereafter, the rats receiving scaffolds seeded with ASCs and MSCs exhibited significant recovery of nerve function as shown by the Basso, Beattie and Bresnahan (BBB) score and electrophysiological test results. Immunohistochemical analyses at 4 and 8 weeks after transplantation revealed that the implanted MSCs at the lesion/graft site survived and differentiated into neuron-like cells and co-localized with host neurons. Robust bundles of regenerated fibers were identified in the lesion/graft site in the ASC and MSC co-transplantation rats, and neurofilament 200 (NF) staining confirmed that these fibers were axons. Furthermore, myelin basic protein (MBP)-positive myelin sheaths were also identified at the lesion/graft site and confirmed via electron microscopy. In addition to expressing mature neuronal markers, sparse MSC-derived neuron-like cells expressed choline acetyltransferase (ChAT) at the injury site of the ASC and MSC co-transplantation rats. These findings suggest that co-transplantation of ASCs and MSCs in a multichannel polymer scaffold may represent a novel combinatorial strategy for the treatment of spinal cord injury.
Collapse
|
41
|
Cell transplantation therapy for spinal cord injury. Nat Neurosci 2017; 20:637-647. [DOI: 10.1038/nn.4541] [Citation(s) in RCA: 435] [Impact Index Per Article: 62.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 02/22/2017] [Indexed: 02/07/2023]
|
42
|
Lu Y, Gao H, Zhang M, Chen B, Yang H. Glial Cell Line-Derived Neurotrophic Factor-Transfected Placenta-Derived Versus Bone Marrow-Derived Mesenchymal Cells for Treating Spinal Cord Injury. Med Sci Monit 2017; 23:1800-1811. [PMID: 28408732 PMCID: PMC5400030 DOI: 10.12659/msm.902754] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Background Placenta-derived mesenchymal stem cells (PMSCs) were isolated from placenta and had differentiation and self-renewal potential. We transfected PMSCs with glial cell line-derived neurotrophic factor (GDNF) and compared their effect for repairing spinal cord injury (SCI) with that of GDNF-transfected bone marrow-derived mesenchymal stem cell (BMSC). Material/Methods The PMSCs were isolated from Sprague-Dawley rat placenta; BMSCs were isolated from Sprague-Dawley rat thigh bone marrow. Primary cultured BMSCs and PMSCs were uniformly spindle-shaped. Flow cytometry indicated that both cell types were CD29- and CD90-positive and CD34- and CD45-negative, confirming that they were MSCs. The PMSCs and BMSCs were transfected with recombinant lentivirus containing the GDNF gene in vitro. PMSC and BMSC viability was increased after transfection, and GDNF expression was increased until 10 d after transfection. SCI was created in the rats (n=64) and was repaired using transfected PMSCs and BMSCs or untransfected PMSCs and BMSCs. Results The transfected PMSCs and BMSCs repaired the SCI. Flow cytometry, histology, immunohistochemical, kinesiology properties, and Basso-Beattie-Bresnahan locomotion score measurements determined no significant difference between transfected PMSCs and BMSCs at 7, 14, and 21 d post-transplantation (P>0.05); the injury healed better in transfected PMSCs and BMSCs than in untransfected PMSCs and BMSCs (P<0.05). Conclusions MSCs have similar biology characteristics and capacity for SCI repair to BMSCs and can be used as a new resource for treating SCI.
Collapse
Affiliation(s)
- Yao Lu
- Department of Orthopedics, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China (mainland)
| | - Hui Gao
- Department of Dermatology, College of Clinical of Yangzhou University, Yangzhou, Jiangsu, China (mainland)
| | - Man Zhang
- Department of Dermatology, College of Clinical of Yangzhou University, Yangzhou, Jiangsu, China (mainland)
| | - Bing Chen
- Comparative Medicine Center, Yangzhou University, Yangzhou, Jiangsu, China (mainland)
| | - Huilin Yang
- Department of Orthopedics, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China (mainland)
| |
Collapse
|
43
|
Nawrotek K, Marqueste T, Modrzejewska Z, Zarzycki R, Rusak A, Decherchi P. Thermogelling chitosan lactate hydrogel improves functional recovery after a C2 spinal cord hemisection in rat. J Biomed Mater Res A 2017; 105:2004-2019. [PMID: 28324618 DOI: 10.1002/jbm.a.36067] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 01/23/2017] [Accepted: 03/15/2017] [Indexed: 11/06/2022]
Abstract
The present study was designed to provide an appropriate micro-environment for regenerating axotomized neurons and proliferating/migrating cells. Because of its intrinsic permissive properties, biocompatibility and biodegradability, we chose to evaluate the therapeutic effectiveness of a chitosan-based biopolymer. The biomaterial toxicity was measured through in vitro test based on fibroblast cell survival on thermogelling chitosan lactate hydrogel substrate and then polymer was implanted into a C2 hemisection of the rat spinal cord. Animals were randomized into three experimental groups (Control, Lesion and Lesion + Hydrogel) and functional tests (ladder walking and forelimb grip strength tests, respiratory assessment by whole-body plethysmography measurements) were used, once a week during 10 weeks, to evaluate post-traumatic recoveries. Then, electrophysiological examinations (reflexivity of the sub-lesional region, ventilatory adjustments to muscle fatigue known to elicit the muscle metaboreflex and phrenic nerve recordings during normoxia and temporary hypoxia) were performed. In vitro results indicated that the chitosan matrix is a non-toxic biomaterial that allowed fibroblast survival. Furthermore, implanted animals showed improvements of their ladder walking scores from the 4th week post-implantation. Finally, electrophysiological recordings indicated that animals receiving the chitosan matrix exhibited recovery of the H-reflex rate sensitive depression, the ventilatory response to repetitive muscle stimulation and an increase of the phrenic nerve activity to asphyxia compared to lesioned and nonimplanted animals. This study indicates that hydrogel based on chitosan constitute a promising therapeutic approach to repair damaged spinal cord or may be used as an adjuvant with other treatments to enhance functional recovery after a central nervous system damage. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 105A: 2004-2019, 2017.
Collapse
Affiliation(s)
- Katarzyna Nawrotek
- Faculty of Process and Environmental Engineering, Department of Chemical Engineering, Lodz University of Technology, Wolczanska 175 Street, Lodz, 90-924, Poland
| | - Tanguy Marqueste
- Aix-Marseille Université (AMU) and Centre National de la Recherche Scientifique (CNRS), Institut des Sciences du Mouvement (UMR 7287), Equipe « Plasticité des Systèmes Nerveux et Musculaire », Parc Scientifique et Technologique de Luminy, CC910-163, Avenue de Luminy, F-13288, Marseille Cedex 09, France
| | - Zofia Modrzejewska
- Faculty of Process and Environmental Engineering, Department of Chemical Engineering, Lodz University of Technology, Wolczanska 175 Street, Lodz, 90-924, Poland
| | - Roman Zarzycki
- Faculty of Process and Environmental Engineering, Department of Chemical Engineering, Lodz University of Technology, Wolczanska 175 Street, Lodz, 90-924, Poland
| | - Agnieszka Rusak
- Department of Experimental Surgery and Biomaterials Research, Wroclaw Medical University, Medico-Dental Faculty, Krakowska 26 Street, Wroclaw, Poland, 50-425
| | - Patrick Decherchi
- Aix-Marseille Université (AMU) and Centre National de la Recherche Scientifique (CNRS), Institut des Sciences du Mouvement (UMR 7287), Equipe « Plasticité des Systèmes Nerveux et Musculaire », Parc Scientifique et Technologique de Luminy, CC910-163, Avenue de Luminy, F-13288, Marseille Cedex 09, France
| |
Collapse
|
44
|
Shahriari D, Koffler JY, Tuszynski MH, Campana WM, Sakamoto JS. Hierarchically Ordered Porous and High-Volume Polycaprolactone Microchannel Scaffolds Enhanced Axon Growth in Transected Spinal Cords. Tissue Eng Part A 2017; 23:415-425. [PMID: 28107810 DOI: 10.1089/ten.tea.2016.0378] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
The goal of this work was to design nerve guidance scaffolds with a unique architecture to maximize the open volume available for nerve growth. Polycaprolactone (PCL) was selected as the scaffold material based on its biocompatibility and month-long degradation. Yet, dense PCL does not exhibit suitable properties such as porosity, stiffness, strength, and cell adhesion to function as an effective nerve guidance scaffold. To address these shortcomings, PCL was processed using a modified salt-leaching technique to create uniquely controlled interconnected porosity. By controlling porosity, we demonstrated that the elastic modulus could be controlled between 2.09 and 182.1 MPa. In addition, introducing porosity and/or coating with fibronectin enhanced the PCL cell attachment properties. To produce PCL scaffolds with maximized open volume, porous PCL microtubes were fabricated and translated into scaffolds with 60 volume percent open volume. The scaffolds were tested in transected rat spinal cords. Linear axon growth within both the microtubes as well as the interstitial space between the tubes was observed, demonstrating that the entire open volume of the scaffold was available for nerve growth. Overall, a novel scaffold architecture and fabrication technique are presented. The scaffolds exhibit significantly higher volume than state-of-the-art scaffolds for promising spinal cord nerve repair.
Collapse
Affiliation(s)
- Dena Shahriari
- 1 Department of Macromolecular Science and Engineering, University of Michigan , Ann Arbor, Michigan
| | - Jacob Y Koffler
- 2 Department of Neuroscience, University of California San Diego , La Jolla, California
| | - Mark H Tuszynski
- 2 Department of Neuroscience, University of California San Diego , La Jolla, California.,3 Veterans Administration Medical Center , La Jolla, California
| | - Wendy M Campana
- 4 Department of Anesthesiology, University of California San Diego , La Jolla, California.,5 Program in Neuroscience, University of California San Diego , La Jolla, California
| | - Jeff S Sakamoto
- 1 Department of Macromolecular Science and Engineering, University of Michigan , Ann Arbor, Michigan.,6 Department of Mechanical Engineering, University of Michigan , Ann Arbor, Michigan
| |
Collapse
|
45
|
Bunge MB, Monje PV, Khan A, Wood PM. From transplanting Schwann cells in experimental rat spinal cord injury to their transplantation into human injured spinal cord in clinical trials. PROGRESS IN BRAIN RESEARCH 2017; 231:107-133. [PMID: 28554394 DOI: 10.1016/bs.pbr.2016.12.012] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Among the potential therapies designed to repair the injured spinal cord is cell transplantation, notably the use of autologous adult human Schwann cells (SCs). Here, we detail some of the critical research accomplished over the last four decades to establish a foundation that enables these cells to be tested in clinical trials. New culture systems allowed novel information to be gained about SCs, including discovering ways to stimulate their proliferation to acquire adequately large numbers for transplantation into the injured human spinal cord. Transplantation of rat SCs into rat models of spinal cord injury has demonstrated that SCs promote repair of injured spinal cord. Additional work required to gain approval from the Food and Drug Administration for the first SC trial in the Miami Project is disclosed. This trial and a second one now underway are described.
Collapse
Affiliation(s)
- Mary B Bunge
- The Miami Project to Cure Paralysis, University of Miami Leonard M. Miller School of Medicine, Miami, FL, United States; Department of Cell Biology, University of Miami Leonard M. Miller School of Medicine, Miami, FL, United States; Department of Neurological Surgery, University of Miami Leonard M. Miller School of Medicine, Miami, FL, United States.
| | - Paula V Monje
- The Miami Project to Cure Paralysis, University of Miami Leonard M. Miller School of Medicine, Miami, FL, United States; Department of Neurological Surgery, University of Miami Leonard M. Miller School of Medicine, Miami, FL, United States
| | - Aisha Khan
- The Interdisciplinary Stem Cell Institute, University of Miami Leonard M. Miller School of Medicine, Miami, FL, United States
| | - Patrick M Wood
- The Miami Project to Cure Paralysis, University of Miami Leonard M. Miller School of Medicine, Miami, FL, United States; Department of Neurological Surgery, University of Miami Leonard M. Miller School of Medicine, Miami, FL, United States
| |
Collapse
|
46
|
Anderson KD, Guest JD, Dietrich WD, Bartlett Bunge M, Curiel R, Dididze M, Green BA, Khan A, Pearse DD, Saraf-Lavi E, Widerström-Noga E, Wood P, Levi AD. Safety of Autologous Human Schwann Cell Transplantation in Subacute Thoracic Spinal Cord Injury. J Neurotrauma 2017; 34:2950-2963. [PMID: 28225648 DOI: 10.1089/neu.2016.4895] [Citation(s) in RCA: 170] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The rationale for implantation of autologous human Schwann cells (SCs) in persons with subacute spinal cord injury (SCI) is based on evidence that transplanted SCs are neuroprotective, support local axonal plasticity, and are capable of myelinating axons. A Phase I clinical trial was conducted to evaluate the safety of autologous human SC transplantation into the injury epicenter of six subjects with subacute SCI. The trial was an open-label, unblinded, non-randomized, non-placebo controlled study with a dose escalation design and standard medical rehabilitation. Participants were paraplegics with neurologically complete, trauma-induced spinal lesions. Autologous SCs were cultured in vitro from a sural nerve harvested from each participant and injected into the epicenter of the spinal lesion. Outcome measures for safety were protocol compliance, feasibility, adverse events, stability of neurological level, absence of detectable mass lesion, and the emergence of clinically significant neuropathic pain or muscle spasticity no greater than expected for a natural course cohort. One year post-transplantation, there were no surgical, medical, or neurological complications to indicate that the timing or procedure for the cell transplantation was unsafe. There were no adverse events or serious adverse events related to the cell therapy. There was no evidence of additional spinal cord damage, mass lesion, or syrinx formation. We conclude that it is feasible to identify eligible candidates, appropriately obtain informed consent, perform a peripheral nerve harvest to obtain SCs within 5-30 days of injury, and perform an intra-spinal transplantation of highly purified autologous SCs within 4-7 weeks of injury.
Collapse
Affiliation(s)
- Kim D Anderson
- 1 The Miami Project to Cure Paralysis, The University of Miami Miller School of Medicine , Miami, Florida.,2 Department of Neurological Surgery, The University of Miami Miller School of Medicine , Miami, Florida
| | - James D Guest
- 1 The Miami Project to Cure Paralysis, The University of Miami Miller School of Medicine , Miami, Florida.,2 Department of Neurological Surgery, The University of Miami Miller School of Medicine , Miami, Florida.,3 The Neuroscience Program, The University of Miami Miller School of Medicine , Miami, Florida
| | - W Dalton Dietrich
- 1 The Miami Project to Cure Paralysis, The University of Miami Miller School of Medicine , Miami, Florida.,2 Department of Neurological Surgery, The University of Miami Miller School of Medicine , Miami, Florida.,3 The Neuroscience Program, The University of Miami Miller School of Medicine , Miami, Florida.,4 Department of Cell Biology, The University of Miami Miller School of Medicine , Miami, Florida.,5 Department of Neurology, The University of Miami Miller School of Medicine , Miami, Florida.,6 Department of The Interdisciplinary Stem Cell Institute, The University of Miami Miller School of Medicine , Miami, Florida
| | - Mary Bartlett Bunge
- 1 The Miami Project to Cure Paralysis, The University of Miami Miller School of Medicine , Miami, Florida.,2 Department of Neurological Surgery, The University of Miami Miller School of Medicine , Miami, Florida.,3 The Neuroscience Program, The University of Miami Miller School of Medicine , Miami, Florida.,4 Department of Cell Biology, The University of Miami Miller School of Medicine , Miami, Florida.,5 Department of Neurology, The University of Miami Miller School of Medicine , Miami, Florida.,6 Department of The Interdisciplinary Stem Cell Institute, The University of Miami Miller School of Medicine , Miami, Florida
| | - Rosie Curiel
- 7 Department of Psychiatry, The University of Miami Miller School of Medicine , Miami, Florida
| | - Marine Dididze
- 1 The Miami Project to Cure Paralysis, The University of Miami Miller School of Medicine , Miami, Florida.,2 Department of Neurological Surgery, The University of Miami Miller School of Medicine , Miami, Florida
| | - Barth A Green
- 1 The Miami Project to Cure Paralysis, The University of Miami Miller School of Medicine , Miami, Florida.,2 Department of Neurological Surgery, The University of Miami Miller School of Medicine , Miami, Florida.,5 Department of Neurology, The University of Miami Miller School of Medicine , Miami, Florida.,8 Department of Orthopaedics, The University of Miami Miller School of Medicine , Miami, Florida.,9 Department of Rehabilitation Medicine, The University of Miami Miller School of Medicine , Miami, Florida
| | - Aisha Khan
- 1 The Miami Project to Cure Paralysis, The University of Miami Miller School of Medicine , Miami, Florida.,6 Department of The Interdisciplinary Stem Cell Institute, The University of Miami Miller School of Medicine , Miami, Florida
| | - Damien D Pearse
- 1 The Miami Project to Cure Paralysis, The University of Miami Miller School of Medicine , Miami, Florida.,2 Department of Neurological Surgery, The University of Miami Miller School of Medicine , Miami, Florida.,3 The Neuroscience Program, The University of Miami Miller School of Medicine , Miami, Florida.,6 Department of The Interdisciplinary Stem Cell Institute, The University of Miami Miller School of Medicine , Miami, Florida.,11 Bruce W. Carter Department of Veterans Affairs Medical Center , Miami, Florida
| | - Efrat Saraf-Lavi
- 10 Department of Radiology, The University of Miami Miller School of Medicine , Miami, Florida
| | - Eva Widerström-Noga
- 1 The Miami Project to Cure Paralysis, The University of Miami Miller School of Medicine , Miami, Florida.,2 Department of Neurological Surgery, The University of Miami Miller School of Medicine , Miami, Florida.,3 The Neuroscience Program, The University of Miami Miller School of Medicine , Miami, Florida.,9 Department of Rehabilitation Medicine, The University of Miami Miller School of Medicine , Miami, Florida.,11 Bruce W. Carter Department of Veterans Affairs Medical Center , Miami, Florida
| | - Patrick Wood
- 1 The Miami Project to Cure Paralysis, The University of Miami Miller School of Medicine , Miami, Florida.,2 Department of Neurological Surgery, The University of Miami Miller School of Medicine , Miami, Florida
| | - Allan D Levi
- 1 The Miami Project to Cure Paralysis, The University of Miami Miller School of Medicine , Miami, Florida.,2 Department of Neurological Surgery, The University of Miami Miller School of Medicine , Miami, Florida.,8 Department of Orthopaedics, The University of Miami Miller School of Medicine , Miami, Florida
| |
Collapse
|
47
|
Dell'Anno MT, Strittmatter SM. Rewiring the spinal cord: Direct and indirect strategies. Neurosci Lett 2016; 652:25-34. [PMID: 28007647 DOI: 10.1016/j.neulet.2016.12.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Revised: 11/15/2016] [Accepted: 12/02/2016] [Indexed: 12/23/2022]
Abstract
Spinal cord injury is currently incurable. Treatment is limited to minimizing secondary complications and maximizing residual function by rehabilitation. Neurologic recovery is prevented by the poor intrinsic regenerative capacity of neurons in the adult central nervous system and by the presence of growth inhibitors in the adult brain and spinal cord. Here we identify three approaches to rewire the spinal cord after injury: axonal regeneration (direct endogenous reconnection), axonal sprouting (indirect endogenous reconnection) and neural stem cell transplantation (indirect exogenous reconnection). Regeneration and sprouting of axonal fibers can be both enhanced through the neutralization of myelin- and extracellular matrix-associated inhibitors described in the first part of this review. Alternatively, in the second part we focus on the formation of a novel circuit through the grafting of neural stem cells in the lesion site. Transplanted neural stem cells differentiate in vivo into neurons and glial cells which form an intermediate station between the rostral and caudal segment of the recipient spinal cord. In particular, here we describe how neural stem cells-derived neurons are endowed with the ability to extend long-distance axons to regain the transmission of motor and sensory information.
Collapse
Affiliation(s)
- Maria Teresa Dell'Anno
- Program in Cellular Neuroscience, Neurodegeneration & Repair, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Stephen M Strittmatter
- Program in Cellular Neuroscience, Neurodegeneration & Repair, Yale University School of Medicine, New Haven, CT 06536, USA.
| |
Collapse
|
48
|
Lin X, Zhao T, Walker M, Ding A, Lin S, Cao Y, Zheng J, Liu X, Geng M, Xu XM, Liu S. Transplantation of Pro-Oligodendroblasts, Preconditioned by LPS-Stimulated Microglia, Promotes Recovery After Acute Contusive Spinal Cord Injury. Cell Transplant 2016; 25:2111-2128. [PMID: 27513556 DOI: 10.3727/096368916x692636] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Spinal cord injury (SCI) is a significant clinical challenge, and to date no effective treatment is available. Oligodendrocyte progenitor cell (OPC) transplantation has been a promising strategy for SCI repair. However, the poor posttransplantation survival and deficiency in differentiation into myelinating oligodendrocytes (OLs) are two major challenges that limit the use of OPCs as donor cells. Here we report the generation of an OL lineage population [i.e., pro-oligodendroblasts (proOLs)] that is relatively more mature than OPCs for transplantation after SCI. We found that proOLs responded to lipopolysaccharide (LPS)-stimulated microglia conditioned medium (L+M) by preserving toll-like receptor 4 (TLR4) expression, improving cell viability, and enhancing the expression of a myelinating OL marker myelin basic protein (MBP), compared to other OL lineage cells exposed to either LPS-stimulated (L+M) or nonstimulated microglia conditioned medium (LM). When L+M-stimulated proOLs were intrathecally delivered through a lumbar puncture after a T10 thoracic contusive SCI, they promoted behavioral recovery, as assessed by the BassoBeattieBresnahan (BBB) locomotor rating scale, stride length, and slips on the grid tests. Histologically, transplantation of L+M proOLs caused a considerable increase in intralesional axon numbers and myelination, and less accumulation of invading macrophages when compared with the vehicle control or OPC transplantation. Thus, transplantation of proOLs, preconditioned by L+M, may offer a better therapeutic potential for SCI than OPCs since the former may have initiated the differentiation process toward OLs prior to transplantation.
Collapse
|
49
|
Yue Y, Yang X, Zhang L, Xiao X, Nabar NR, Lin Y, Hao L, Zhang D, Huo J, Li J, Cai X, Wang M. Low-intensity pulsed ultrasound upregulates pro-myelination indicators of Schwann cells enhanced by co-culture with adipose-derived stem cells. Cell Prolif 2016; 49:720-728. [PMID: 27625295 PMCID: PMC6496622 DOI: 10.1111/cpr.12298] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 08/24/2016] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVES Peripheral nerve injuries are a common occurrence, resulting in considerable patient suffering; it also represents a major economic burden on society. To improve treatment options following peripheral nerve injuries, scientists aim to find a way to promote Schwann cell (SC) myelination to help nerves to carry out their functions effectively. In this study, we investigated myelination ability of SCs, regulated by co-culture with adipose-derived stem cells (ASCs) or low-intensity pulsed ultrasound (LIPUS), and synergistic effects of combined treatments. MATERIALS AND METHODS Schwann cells were co-cultured with or without ASCs, and either left untreated or treated with LIPUS for 10 min/d for 1, 4 or 7 days. Effects of LIPUS and ASC co-culture on pro-myelination indicators of SCs were analysed by real-time PCR (RT-PCR), Western blotting and immunofluorescence staining (IF). RESULTS Our results indicate that ASC-SC co-culture and LIPUS, together or individually, promoted mRNA levels of epidermal growth factor receptor 3 (EGFR3/ErbB3), neuregulin1 (NRG1), early growth response protein 2 (Egr2/Krox20) and myelin basic protein (MBP), with corresponding increases in protein levels of ErbB3, NRG1 and Krox20. Interestingly, combination of ASC-SC co-culture and LIPUS displayed the most remarkable effects. CONCLUSION We demonstrated that ASCs upregulated pro-myelination indicators of SCs by indirect contact (through co-culture) and that effects could be potentiated by LIPUS. We conclude that LIPUS, as a mechanical stress, may have potential in nerve regeneration with potential clinical relevance.
Collapse
Affiliation(s)
- Yuan Yue
- State Key Laboratory of Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, China
- Department of Prosthodontics, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Xingmei Yang
- State Key Laboratory of Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, China.
- Oral Implant Center, West China School of Stomatology, Sichuan University, Chengdu, China.
| | - Liang Zhang
- State Key Laboratory of Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, China
- Oral Implant Center, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Xun Xiao
- State Key Laboratory of Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, China
- Department of Prosthodontics, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Neel R Nabar
- Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA, USA
| | - Yunfeng Lin
- State Key Laboratory of Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Liang Hao
- State Key Laboratory of Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Dongjiao Zhang
- State Key Laboratory of Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, China
- Department of Prosthodontics, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Jingyi Huo
- State Key Laboratory of Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, China
- Department of Prosthodontics, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Jingle Li
- State Key Laboratory of Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, China
- Department of Prosthodontics, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Xiaoxiao Cai
- State Key Laboratory of Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, China
- Oral Implant Center, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Min Wang
- State Key Laboratory of Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, China.
- Department of Prosthodontics, West China School of Stomatology, Sichuan University, Chengdu, China.
| |
Collapse
|
50
|
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: 5.4] [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
|