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 PMCID: PMC11624882 DOI: 10.4103/nrr.nrr-d-23-01338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 12/20/2023] [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
|
Hajimirzaei P, Tabatabaei FSA, Nasibi-Sis H, Razavian RS, Nasirinezhad F. Schwann cell transplantation for remyelination, regeneration, tissue sparing, and functional recovery in spinal cord injury: A systematic review and meta-analysis of animal studies. Exp Neurol 2025; 384:115062. [PMID: 39579959 DOI: 10.1016/j.expneurol.2024.115062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 11/02/2024] [Accepted: 11/12/2024] [Indexed: 11/25/2024]
Abstract
INTRODUCTION Spinal cord injury (SCI) is a significant global health challenge that results in profound physical and neurological impairments. Despite progress in medical care, the treatment options for SCI are still restricted and often focus on symptom management rather than promoting neural repair and functional recovery. This study focused on clarifying the impact of Schwann cell (SC) transplantation on the molecular, cellular, and functional basis of recovery in animal models of SCI. MATERIAL AND METHODS Relevant studies were identified by conducting searches across multiple databases, which included PubMed, Web of Science, Scopus, and ProQuest. The data were analyzed via comprehensive meta-analysis software. We assessed the risk of bias via the SYRCLE method. RESULTS The analysis included 59 studies, 48 of which provided quantitative data. The results revealed significant improvements in various outcome variables, including protein zero structures (SMD = 1.66, 95 %CI: 0.96-2.36; p < 0.001; I2 = 49.8 %), peripherally myelinated axons (SMD = 1.81, 95 %CI: 0.99-2.63; p < 0.001; I2 = 39.3 %), biotinylated dextran amine-labeled CST only rostral (SMD = 1.31, 95 % CI: 0.50-2.12, p < 0.01, I2 = 49.7 %), fast blue-labeled reticular formation (SMD = 0.96, 95 %CI: 0.43-1.49, p < 0.001, I2 = 0.0 %), 5-hydroxytryptamine caudally (SMD = 0.83, 95 %CI: 0.36-1.29, p < 0.001, I2 = 17.2 %) and epicenter (SMD = 0.85, 95 %CI: 0.17-1.53, p < 0.05, I2 = 62.7 %), tyrosine hydroxylase caudally (SMD = 1.86, 95 %CI: 1.14-2.59, p < 0.001, I2 = 0.0 %) and epicenter (SMD = 1.82, 95 %CI: 1.18-2.47, p < 0.001, I2 = 0.0 %), cavity volume (SMD = -2.07, 95 %CI: -2.90 - -1.24, p < 0.001, I2 = 67.2 %), and Basso, Beattie, and Bresnahan (SMD = 1.26, 95 %CI: 0.93-1.58; p < 0.001; I2 = 79.4 %). CONCLUSIONS This study demonstrates the promising potential of SC transplantation as a therapeutic approach for SCI, clarifying its impact on various biological processes critical for recovery.
Collapse
Affiliation(s)
- Pooya Hajimirzaei
- Department of Radiation Sciences, Allied Medicine Faculty, Iran University of Medical Sciences, Tehran, Iran; Radiation Biology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | | | - Hamed Nasibi-Sis
- Department of Medical Library and Information Sciences, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Farinaz Nasirinezhad
- Department of Physiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Physiology Research Center, Iran University of Medical sciences, Tehran, Iran; Center of Experimental and Comparative Study, Iran University of Medical sciences, Tehran, Iran.
| |
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 2025; 27:36-50. [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] [MESH Headings] [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
|
Dill-Macky AS, Lee EN, Wertheim JA, Koss KM. Glia in tissue engineering: From biomaterial tools to transplantation. Acta Biomater 2024; 190:24-49. [PMID: 39396630 DOI: 10.1016/j.actbio.2024.10.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 10/01/2024] [Accepted: 10/10/2024] [Indexed: 10/15/2024]
Abstract
Glia are imperative in nearly every function of the nervous system, including neurotransmission, neuronal repair, development, immunity, and myelination. Recently, the reparative roles of glia in the central and peripheral nervous systems have been elucidated, suggesting a tremendous potential for these cells as novel treatments to central nervous system disorders. Glial cells often behave as 'double-edged swords' in neuroinflammation, ultimately deciding the life or death of resident cells. Compared to glia, neuronal cells have limited mobility, lack the ability to divide and self-renew, and are generally more delicate. Glia have been candidates for therapeutic use in many successful grafting studies, which have been largely focused on restoring myelin with Schwann cells, olfactory ensheathing glia, and oligodendrocytes with support from astrocytes. However, few therapeutics of this class have succeeded past clinical trials. Several tools and materials are being developed to understand and re-engineer these grafting concepts for greater success, such as extra cellular matrix-based scaffolds, bioactive peptides, biomolecular delivery systems, biomolecular discovery for neuroinflammatory mediation, composite microstructures such as artificial channels for cell trafficking, and graft enhanced electrical stimulation. Furthermore, advances in stem cell-derived cortical/cerebral organoid differentiation protocols have allowed for the generation of patient-derived glia comparable to those acquired from tissues requiring highly invasive procedures or are otherwise inaccessible. However, research on bioengineered tools that manipulate glial cells is nowhere near as comprehensive as that for systems of neurons and neural stem cells. This article explores the therapeutic potential of glia in transplantation with an emphasis on novel bioengineered tools for enhancement of their reparative properties. STATEMENT OF SIGNIFICANCE: Neural glia are responsible for a host of developmental, homeostatic, and reparative roles in the central nervous system but are often a major cause of tissue damage and cellular loss in insults and degenerative pathologies. Most glial grafts have employed Schwann cells for remyelination, but other glial with novel biomaterials have been employed, emphasizing their diverse functionality. Promising strategies have emerged, including neuroimmune mediation of glial scar tissues and facilitated migration and differentiation of stem cells for neural replacement. Herein, a comprehensive review of biomaterial tools for glia in transplantation is presented, highlighting Schwann cells, astrocytes, olfactory ensheating glia, oligodendrocytes, microglia, and ependymal cells.
Collapse
Affiliation(s)
- A S Dill-Macky
- Department of Surgery, University of Arizona, 1501 N Campbell Ave, Tucson, AZ 85724, United States
| | - E N Lee
- Department of Surgery, University of Arizona, 1501 N Campbell Ave, Tucson, AZ 85724, United States
| | - J A Wertheim
- Department of Surgery, University of Arizona, 1501 N Campbell Ave, Tucson, AZ 85724, United States
| | - K M Koss
- Department of Neurobiology, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555-0625, United States; Sealy Institute for Drug Discovery, University of Texas Medical Branch, 105 11th Street Galveston, TX 77555-1110, United States.
| |
Collapse
|
5
|
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
|
6
|
Hosseini SM, Borys B, Karimi-Abdolrezaee S. Neural stem cell therapies for spinal cord injury repair: an update on recent preclinical and clinical advances. Brain 2024; 147:766-793. [PMID: 37975820 DOI: 10.1093/brain/awad392] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 10/22/2023] [Accepted: 11/02/2023] [Indexed: 11/19/2023] Open
Abstract
Traumatic spinal cord injury (SCI) is a leading cause of lifelong disabilities. Permanent sensory, motor and autonomic impairments after SCI are substantially attributed to degeneration of spinal cord neurons and axons, and disintegration of neural network. To date, minimal regenerative treatments are available for SCI with an unmet need for new therapies to reconstruct the damaged spinal cord neuron-glia network and restore connectivity with the supraspinal pathways. Multipotent neural precursor cells (NPCs) have a unique capacity to generate neurons, oligodendrocytes and astrocytes. Due to this capacity, NPCs have been an attractive cell source for cellular therapies for SCI. Transplantation of NPCs has been extensively tested in preclinical models of SCI in the past two decades. These studies have identified opportunities and challenges associated with NPC therapies. While NPCs have the potential to promote neuroregeneration through various mechanisms, their low long-term survival and integration within the host injured spinal cord limit the functional benefits of NPC-based therapies for SCI. To address this challenge, combinatorial strategies have been developed to optimize the outcomes of NPC therapies by enriching SCI microenvironment through biomaterials, genetic and pharmacological therapies. In this review, we will provide an in-depth discussion on recent advances in preclinical NPC-based therapies for SCI. We will discuss modes of actions and mechanism by which engrafted NPCs contribute to the repair process and functional recovery. We will also provide an update on current clinical trials and new technologies that have facilitated preparation of medical-grade human NPCs suitable for transplantation in clinical studies.
Collapse
Affiliation(s)
- Seyed Mojtaba Hosseini
- Department of Physiology and Pathophysiology, Spinal Cord Research Centre, Rady Faculty of Health Sciences, University of Manitoba Winnipeg, Manitoba R3E 0J9, Canada
- Manitoba Multiple Sclerosis Research Center, Winnipeg, Manitoba R3E 0J9, Canada
| | - Ben Borys
- Department of Physiology and Pathophysiology, Spinal Cord Research Centre, Rady Faculty of Health Sciences, University of Manitoba Winnipeg, Manitoba R3E 0J9, Canada
| | - Soheila Karimi-Abdolrezaee
- Department of Physiology and Pathophysiology, Spinal Cord Research Centre, Rady Faculty of Health Sciences, University of Manitoba Winnipeg, Manitoba R3E 0J9, Canada
- Manitoba Multiple Sclerosis Research Center, Winnipeg, Manitoba R3E 0J9, Canada
- Children's Hospital Research Institute of Manitoba, Winnipeg, Manitoba R3E 3P4, Canada
| |
Collapse
|
7
|
Mehranpour M, Sani M, Beirami A, Hasanzadeh M, Taghizadeh M, Banihashemi M, Moghaddam MH, Fathi M, Vakili K, Yaghoobpoor S, Eskandari N, Abdollahifar MA, Bayat AH, Aliaghaei A, Heidari MH. Grafted Sertoli cells prevent neuronal cell death and memory loss induced by seizures. Metab Brain Dis 2023; 38:2735-2750. [PMID: 37851137 DOI: 10.1007/s11011-023-01309-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 10/04/2023] [Indexed: 10/19/2023]
Abstract
Epilepsy significantly reduces the patient's quality of life, and we still need to develop new therapeutic approaches to control it. Transplantation of cells such as Sertoli cells (SCs), having a potent ability to release a variety of growth and immunoprotective substances, have made them a potential candidate to deal with neurological diseases like epilepsy. Hence, this study aims to evaluate whether SCs transplant effectively protects the hippocampus astrocytes and neurons to oppose seizure damage. For this purpose, the effects of bilateral intrahippocampal transplantation of SCs were investigated on the rats with the pentylenetetrazol (PTZ) induced seizure. After one-month, post-graft analysis was performed regarding behavior, immunohistopathology, and the distribution of the hippocampal cells. Our findings showed SCs transplantation reduced astrogliosis, astrocytes process length, the number of branches, and intersections distal to the soma of the hippocampus in the seizure group. In rats with grafted SCs, there was a drop in the hippocampal caspase-3 expression. Moreover, the SCs showed another protective impact, as shown by an improvement in pyramidal neurons' number and spatial distribution. The findings suggested that SCs transplantation can potently modify astrocytes' reactivation and inflammatory responses.
Collapse
Affiliation(s)
- Maryam Mehranpour
- Hearing Disorders Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Genetics, Faculty of Biological Sciences, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Mojtaba Sani
- Department of Educational Neuroscience, Aras International Campus, University of Tabriz, Tabriz, Iran
| | - Amirreza Beirami
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maral Hasanzadeh
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Taghizadeh
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mobina Banihashemi
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Meysam Hassani Moghaddam
- Department of Anatomical Sciences, Faculty of Medicine, AJA University of Medical Sciences, Tehran, Iran.
| | - Mobina Fathi
- Student Research Committee, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kimia Vakili
- Student Research Committee, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shirin Yaghoobpoor
- Student Research Committee, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Neda Eskandari
- Department of Anatomical Sciences, Faculty of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Mohammad-Amin Abdollahifar
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amir-Hossein Bayat
- Department of Neuroscience, School of Sciences and Advanced Technology in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Abbas Aliaghaei
- Hearing Disorders Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Mohammad Hossain Heidari
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
8
|
Liu JP, Wang JL, Hu BE, Zou FL, Wu CL, Shen J, Zhang WJ. Olfactory ensheathing cells and neuropathic pain. Front Cell Dev Biol 2023; 11:1147242. [PMID: 37223000 PMCID: PMC10201020 DOI: 10.3389/fcell.2023.1147242] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 03/27/2023] [Indexed: 08/29/2023] Open
Abstract
Damage to the nervous system can lead to functional impairment, including sensory and motor functions. Importantly, neuropathic pain (NPP) can be induced after nerve injury, which seriously affects the quality of life of patients. Therefore, the repair of nerve damage and the treatment of pain are particularly important. However, the current treatment of NPP is very weak, which promotes researchers to find new methods and directions for treatment. Recently, cell transplantation technology has received great attention and has become a hot spot for the treatment of nerve injury and pain. Olfactory ensheathing cells (OECs) are a kind of glial cells with the characteristics of lifelong survival in the nervous system and continuous division and renewal. They also secrete a variety of neurotrophic factors, bridge the fibers at both ends of the injured nerve, change the local injury microenvironment, and promote axon regeneration and other biological functions. Different studies have revealed that the transplantation of OECs can repair damaged nerves and exert analgesic effect. Some progress has been made in the effect of OECs transplantation in inhibiting NPP. Therefore, in this paper, we provided a comprehensive overview of the biology of OECs, described the possible pathogenesis of NPP. Moreover, we discussed on the therapeutic effect of OECs transplantation on central nervous system injury and NPP, and prospected some possible problems of OECs transplantation as pain treatment. To provide some valuable information for the treatment of pain by OECs transplantation in the future.
Collapse
Affiliation(s)
- Ji-peng Liu
- Department of Rehabilitation Medicine, The Second Affiliated Hospital, Nanchang University, Nanchang, Jiangxi, China
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Nanchang University, Nanchang, Jiangxi, China
| | - Jia-ling Wang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Nanchang University, Nanchang, Jiangxi, China
| | - Bai-er Hu
- Department of Physical Examination, The Second Affiliated Hospital, Nanchang University, Nanchang, Jiangxi, China
| | - Fei-long Zou
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Nanchang University, Nanchang, Jiangxi, China
| | - Chang-lei Wu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Nanchang University, Nanchang, Jiangxi, China
| | - Jie Shen
- Department of Rehabilitation Medicine, The Second Affiliated Hospital, Nanchang University, Nanchang, Jiangxi, China
| | - Wen-jun Zhang
- Department of Rehabilitation Medicine, The Second Affiliated Hospital, Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
9
|
Conklin B, Conley BM, Hou Y, Chen M, Lee KB. Advanced theragnostics for the central nervous system (CNS) and neurological disorders using functional inorganic nanomaterials. Adv Drug Deliv Rev 2023; 192:114636. [PMID: 36481291 PMCID: PMC11829738 DOI: 10.1016/j.addr.2022.114636] [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: 08/30/2022] [Revised: 10/13/2022] [Accepted: 11/23/2022] [Indexed: 12/12/2022]
Abstract
Various types of inorganic nanomaterials are capable of diagnostic biomarker detection and the therapeutic delivery of a disease or inflammatory modulating agent. Those multi-functional nanomaterials have been utilized to treat neurodegenerative diseases and central nervous system (CNS) injuries in an effective and personalized manner. Even though many nanomaterials can deliver a payload and detect a biomarker of interest, only a few studies have yet to fully utilize this combined strategy to its full potential. Combining a nanomaterial's ability to facilitate targeted delivery, promote cellular proliferation and differentiation, and carry a large amount of material with various sensing approaches makes it possible to diagnose a patient selectively and sensitively while offering preventative measures or early disease-modifying strategies. By tuning the properties of an inorganic nanomaterial, the dimensionality, hydrophilicity, size, charge, shape, surface chemistry, and many other chemical and physical parameters, different types of cells in the central nervous system can be monitored, modulated, or further studies to elucidate underlying disease mechanisms. Scientists and clinicians have better understood the underlying processes of pathologies for many neurologically related diseases and injuries by implementing multi-dimensional 0D, 1D, and 2D theragnostic nanomaterials. The incorporation of nanomaterials has allowed scientists to better understand how to detect and treat these conditions at an early stage. To this end, having the multi-modal ability to both sense and treat ailments of the central nervous system can lead to favorable outcomes for patients suffering from such injuries and diseases.
Collapse
Affiliation(s)
- Brandon Conklin
- Department of Chemistry and Chemical Biology, Rutgers-the State University of New Jersey, 123, Bevier Road, Piscataway, NJ 08854, USA
| | - Brian M Conley
- Department of Chemistry and Chemical Biology, Rutgers-the State University of New Jersey, 123, Bevier Road, Piscataway, NJ 08854, USA
| | - Yannan Hou
- Department of Chemistry and Chemical Biology, Rutgers-the State University of New Jersey, 123, Bevier Road, Piscataway, NJ 08854, USA
| | - Meizi Chen
- Department of Chemistry and Chemical Biology, Rutgers-the State University of New Jersey, 123, Bevier Road, Piscataway, NJ 08854, USA
| | - Ki-Bum Lee
- Department of Chemistry and Chemical Biology, Rutgers-the State University of New Jersey, 123, Bevier Road, Piscataway, NJ 08854, USA.
| |
Collapse
|
10
|
Almeida F, Marques S, Santos A, Prins C, Cardoso F, Heringer L, Mendonça H, Martinez A. Molecular approaches for spinal cord injury treatment. Neural Regen Res 2023; 18:23-30. [PMID: 35799504 PMCID: PMC9241396 DOI: 10.4103/1673-5374.344830] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Injuries to the spinal cord result in permanent disabilities that limit daily life activities. The main reasons for these poor outcomes are the limited regenerative capacity of central neurons and the inhibitory milieu that is established upon traumatic injuries. Despite decades of research, there is still no efficient treatment for spinal cord injury. Many strategies are tested in preclinical studies that focus on ameliorating the functional outcomes after spinal cord injury. Among these, molecular compounds are currently being used for neurological recovery, with promising results. These molecules target the axon collapsed growth cone, the inhibitory microenvironment, the survival of neurons and glial cells, and the re-establishment of lost connections. In this review we focused on molecules that are being used, either in preclinical or clinical studies, to treat spinal cord injuries, such as drugs, growth and neurotrophic factors, enzymes, and purines. The mechanisms of action of these molecules are discussed, considering traumatic spinal cord injury in rodents and humans.
Collapse
|
11
|
Molecular Mechanisms in the Vascular and Nervous Systems following Traumatic Spinal Cord Injury. LIFE (BASEL, SWITZERLAND) 2022; 13:life13010009. [PMID: 36675958 PMCID: PMC9866624 DOI: 10.3390/life13010009] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/26/2022] [Accepted: 12/13/2022] [Indexed: 12/24/2022]
Abstract
Traumatic spinal cord injury (SCI) induces various complex pathological processes that cause physical impairment and psychological devastation. The two phases of SCI are primary mechanical damage (the immediate result of trauma) and secondary injury (which occurs over a period of minutes to weeks). After the mechanical impact, vascular disruption, inflammation, demyelination, neuronal cell death, and glial scar formation occur during the acute phase. This sequence of events impedes nerve regeneration. In the nervous system, various extracellular secretory factors such as neurotrophic factors, growth factors, and cytokines are involved in these events. In the vascular system, the blood-spinal cord barrier (BSCB) is damaged, allowing immune cells to infiltrate the parenchyma. Later, endogenous angiogenesis is promoted during the subacute phase. In this review, we describe the roles of secretory factors in the nervous and vascular systems following traumatic SCI, and discuss the outcomes of their therapeutic application in traumatic SCI.
Collapse
|
12
|
Cell-based and stem-cell-based treatments for spinal cord injury: evidence from clinical trials. Lancet Neurol 2022; 21:659-670. [DOI: 10.1016/s1474-4422(21)00464-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 11/01/2021] [Accepted: 12/17/2021] [Indexed: 12/22/2022]
|
13
|
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: 15] [Impact Index Per Article: 3.8] [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
|
14
|
Tan C, Yang C, Liu H, Tang C, Huang S. Effect of Schwann cell transplantation combined with electroacupuncture on axonal regeneration and remyelination in rats with spinal cord injury. Anat Rec (Hoboken) 2021; 304:2506-2520. [PMID: 34319000 DOI: 10.1002/ar.24721] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 05/26/2021] [Accepted: 06/16/2021] [Indexed: 11/08/2022]
Abstract
Axonal impairment and demyelination after compressed spinal cord injury lead to serious neurological dysfunction. Increasing studies have suggested that Schwann cells (SCs) transplantation is a reliable, effective, and promising method for treating spinal cord injury. However, single SCs transplantation is insufficient to promote the full recovery of neurological function. Additional approaches are required to support SCs transplantation as a treatment for spinal cord injury. In the study, we investigated whether the combination of electroacupuncture (EA) and SCs transplantation was a reliable intervention for spinal cord injury. We found that rats in the combination group had significantly higher functional locomotor scores than those received single treatment. By immunostaining, we found EA can not only improve survival and proliferation of transplanted SCs but also inhibit SC apoptosis and block the formation of an astrocytic scar. Additionally, EA promoted regenerated axons extending "bullet-shaped" growth cones into the lesion. Remarkably, EA can modify astrogliosis to promote axonal regeneration following SCs transplantation through inducing extension of astrocytic processes in the SCs graft interface. More importantly, the combination of SCs engraftment and EA can enhance corticospinal-tract axonal regeneration and remyelination after spinal cord injury through up-regulating neuregulin 1 type III in SCs and its downstream signaling mediators. Thus, it is concluded that SCs effectively promote axonal recovery after spinal cord injury when combined with EA stimulation. The experimental results have reinforced the theoretical basis of EA for its clinical efficacy in patients with spinal cord injury and merited further investigation for potential clinical application.
Collapse
Affiliation(s)
- Chengfang Tan
- Traditional Chinese Medicine College, Chongqing Medical University, Chongqing, China
| | - Cheng Yang
- Traditional Chinese Medicine College, Chongqing Medical University, Chongqing, China
| | - Hui Liu
- Institute of Neuroscience, Chongqing Medical University, Chongqing, China
| | - Chenglin Tang
- Traditional Chinese Medicine College, Chongqing Medical University, Chongqing, China
| | - Siqin Huang
- Traditional Chinese Medicine College, Chongqing Medical University, Chongqing, China
| |
Collapse
|
15
|
Lee HL, Yeum CE, Lee H, Oh J, Kim JT, Lee WJ, Ha Y, Yang YI, Kim KN. Peripheral Nerve-Derived Stem Cell Spheroids Induce Functional Recovery and Repair after Spinal Cord Injury in Rodents. Int J Mol Sci 2021; 22:ijms22084141. [PMID: 33923671 PMCID: PMC8072978 DOI: 10.3390/ijms22084141] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 04/05/2021] [Accepted: 04/12/2021] [Indexed: 01/09/2023] Open
Abstract
Stem cell therapy is one of the most promising candidate treatments for spinal cord injury. Research has shown optimistic results for this therapy, but clinical limitations remain, including poor viability, engraftment, and differentiation. Here, we isolated novel peripheral nerve-derived stem cells (PNSCs) from adult peripheral nerves with similar characteristics to neural-crest stem cells. These PNSCs expressed neural-crest specific markers and showed multilineage differentiation potential into Schwann cells, neuroglia, neurons, and mesodermal cells. In addition, PNSCs showed therapeutic potential by releasing the neurotrophic factors, including glial cell-line-derived neurotrophic factor, insulin-like growth factor, nerve growth factor, and neurotrophin-3. PNSC abilities were also enhanced by their development into spheroids which secreted neurotrophic factors several times more than non-spheroid PNSCs and expressed several types of extra cellular matrix. These features suggest that the potential for these PNSC spheroids can overcome their limitations. In an animal spinal cord injury (SCI) model, these PNSC spheroids induced functional recovery and neuronal regeneration. These PNSC spheroids also reduced the neuropathic pain which accompanies SCI after remyelination. These PNSC spheroids may represent a new therapeutic approach for patients suffering from SCI.
Collapse
Affiliation(s)
- Hye-Lan Lee
- Spine & Spinal Cord Institute, Department of Neurosurgery, College of Medicine, Yonsei University, Seoul 03722, Korea; (H.-L.L.); (H.L.); (J.O.); (Y.H.)
| | - Chung-Eun Yeum
- Paik Inje Memorial Institute for Clinical Research, Inje University College of Medicine, Busan 47392, Korea; (C.-E.Y.); (J.-T.K.); (W.-J.L.)
| | - HyeYeong Lee
- Spine & Spinal Cord Institute, Department of Neurosurgery, College of Medicine, Yonsei University, Seoul 03722, Korea; (H.-L.L.); (H.L.); (J.O.); (Y.H.)
| | - Jinsoo Oh
- Spine & Spinal Cord Institute, Department of Neurosurgery, College of Medicine, Yonsei University, Seoul 03722, Korea; (H.-L.L.); (H.L.); (J.O.); (Y.H.)
| | - Jong-Tae Kim
- Paik Inje Memorial Institute for Clinical Research, Inje University College of Medicine, Busan 47392, Korea; (C.-E.Y.); (J.-T.K.); (W.-J.L.)
| | - Won-Jin Lee
- Paik Inje Memorial Institute for Clinical Research, Inje University College of Medicine, Busan 47392, Korea; (C.-E.Y.); (J.-T.K.); (W.-J.L.)
| | - Yoon Ha
- Spine & Spinal Cord Institute, Department of Neurosurgery, College of Medicine, Yonsei University, Seoul 03722, Korea; (H.-L.L.); (H.L.); (J.O.); (Y.H.)
- POSTECH Biotech Center, Pohang University of Science and Technology (POSTECH), Pohang, Gyeongbuk 37673, Korea
| | - Young-Il Yang
- Paik Inje Memorial Institute for Clinical Research, Inje University College of Medicine, Busan 47392, Korea; (C.-E.Y.); (J.-T.K.); (W.-J.L.)
- Correspondence: (Y.-I.Y.); (K.-N.K.)
| | - Keung-Nyun Kim
- Spine & Spinal Cord Institute, Department of Neurosurgery, College of Medicine, Yonsei University, Seoul 03722, Korea; (H.-L.L.); (H.L.); (J.O.); (Y.H.)
- Correspondence: (Y.-I.Y.); (K.-N.K.)
| |
Collapse
|
16
|
Khazaei M, Ahuja CS, Nakashima H, Nagoshi N, Li L, Wang J, Chio J, Badner A, Seligman D, Ichise A, Shibata S, Fehlings MG. GDNF rescues the fate of neural progenitor grafts by attenuating Notch signals in the injured spinal cord in rodents. Sci Transl Med 2021; 12:12/525/eaau3538. [PMID: 31915299 DOI: 10.1126/scitranslmed.aau3538] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 04/08/2019] [Accepted: 11/13/2019] [Indexed: 12/13/2022]
Abstract
Neural progenitor cell (NPC) transplantation is a promising strategy for the treatment of spinal cord injury (SCI). In this study, we show that injury-induced Notch activation in the spinal cord microenvironment biases the fate of transplanted NPCs toward astrocytes in rodents. In a screen for potential clinically relevant factors to modulate Notch signaling, we identified glial cell-derived neurotrophic factor (GDNF). GDNF attenuates Notch signaling by mediating delta-like 1 homolog (DLK1) expression, which is independent of GDNF's effect on cell survival. When transplanted into a rodent model of cervical SCI, GDNF-expressing human-induced pluripotent stem cell-derived NPCs (hiPSC-NPCs) demonstrated higher differentiation toward a neuronal fate compared to control cells. In addition, expression of GDNF promoted endogenous tissue sparing and enhanced electrical integration of transplanted cells, which collectively resulted in improved neurobehavioral recovery. CRISPR-induced knockouts of the DLK1 gene in GDNF-expressing hiPSC-NPCs attenuated the effect on functional recovery, demonstrating that this effect is partially mediated through DLK1 expression. These results represent a mechanistically driven optimization of hiPSC-NPC therapy to redirect transplanted cells toward a neuronal fate and enhance their integration.
Collapse
Affiliation(s)
- Mohamad Khazaei
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, ON M5T 0S8, Canada
| | - Christopher S Ahuja
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, ON M5T 0S8, Canada.,Institute of Medical Sciences, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Hiroaki Nakashima
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, ON M5T 0S8, Canada
| | - Narihito Nagoshi
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, ON M5T 0S8, Canada
| | - Lijun Li
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, ON M5T 0S8, Canada
| | - Jian Wang
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, ON M5T 0S8, Canada
| | - Jonathon Chio
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, ON M5T 0S8, Canada.,Institute of Medical Sciences, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Anna Badner
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, ON M5T 0S8, Canada.,Institute of Medical Sciences, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - David Seligman
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, ON M5T 0S8, Canada
| | - Ayaka Ichise
- Electron Microscope Laboratory, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Shinsuke Shibata
- Electron Microscope Laboratory, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Michael G Fehlings
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, ON M5T 0S8, Canada. .,Institute of Medical Sciences, University of Toronto, Toronto, ON M5S 1A8, Canada.,Department of Surgery, University of Toronto, Toronto, ON M5T 1P5, Canada.,Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| |
Collapse
|
17
|
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: 2.8] [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
|
18
|
Muheremu A, Shu L, Liang J, Aili A, Jiang K. Sustained delivery of neurotrophic factors to treat spinal cord injury. Transl Neurosci 2021; 12:494-511. [PMID: 34900347 PMCID: PMC8633588 DOI: 10.1515/tnsci-2020-0200] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 11/04/2021] [Accepted: 11/05/2021] [Indexed: 12/16/2022] Open
Abstract
Acute spinal cord injury (SCI) is a devastating condition that results in tremendous physical and psychological harm and a series of socioeconomic problems. Although neurons in the spinal cord need neurotrophic factors for their survival and development to reestablish their connections with their original targets, endogenous neurotrophic factors are scarce and the sustainable delivery of exogeneous neurotrophic factors is challenging. The widely studied neurotrophic factors such as brain-derived neurotrophic factor, neurotrophin-3, nerve growth factor, ciliary neurotrophic factor, basic fibroblast growth factor, and glial cell-derived neurotrophic factor have a relatively short cycle that is not sufficient enough for functionally significant neural regeneration after SCI. In the past decades, scholars have tried a variety of cellular and viral vehicles as well as tissue engineering scaffolds to safely and sustainably deliver those necessary neurotrophic factors to the injury site, and achieved satisfactory neural repair and functional recovery on many occasions. Here, we review the neurotrophic factors that have been used in trials to treat SCI, and vehicles that were commonly used for their sustained delivery.
Collapse
Affiliation(s)
- Aikeremujiang Muheremu
- Department of Spine Surgery, Sixth Affiliated Hospital of Xinjiang Medical University, 39 Wuxing Nan Rd, Tianshan District, Urumqi, Xinjiang, 86830001, People’s Republic of China
| | - Li Shu
- Department of Orthopedics, Sixth Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 86830001, People’s Republic of China
| | - Jing Liang
- Department of Laboratory Medicine, Sixth Affiliated Hospital of Xinjiang Medical University, 39, Wuxing Nan Rd, Tianshan District, Urumqi, Xinjiang, 86830001, People’s Republic of China
| | - Abudunaibi Aili
- Department of Spine Surgery, Sixth Affiliated Hospital of Xinjiang Medical University, 39 Wuxing Nan Rd, Tianshan District, Urumqi, Xinjiang, 86830001, People’s Republic of China
| | - Kan Jiang
- Department of Orthopedics, Sixth Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 86830001, People’s Republic of China
| |
Collapse
|
19
|
Bellák T, Fekécs Z, Török D, Táncos Z, Nemes C, Tézsla Z, Gál L, Polgári S, Kobolák J, Dinnyés A, Nógrádi A, Pajer K. Grafted human induced pluripotent stem cells improve the outcome of spinal cord injury: modulation of the lesion microenvironment. Sci Rep 2020; 10:22414. [PMID: 33376249 PMCID: PMC7772333 DOI: 10.1038/s41598-020-79846-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 12/10/2020] [Indexed: 02/06/2023] Open
Abstract
Spinal cord injury results in irreversible tissue damage followed by a very limited recovery of function. In this study we investigated whether transplantation of undifferentiated human induced pluripotent stem cells (hiPSCs) into the injured rat spinal cord is able to induce morphological and functional improvement. hiPSCs were grafted intraspinally or intravenously one week after a thoracic (T11) spinal cord contusion injury performed in Fischer 344 rats. Grafted animals showed significantly better functional recovery than the control rats which received only contusion injury. Morphologically, the contusion cavity was significantly smaller, and the amount of spared tissue was significantly greater in grafted animals than in controls. Retrograde tracing studies showed a statistically significant increase in the number of FB-labeled neurons in different segments of the spinal cord, the brainstem and the sensorimotor cortex. The extent of functional improvement was inversely related to the amount of chondroitin-sulphate around the cavity and the astrocytic and microglial reactions in the injured segment. The grafts produced GDNF, IL-10 and MIP1-alpha for at least one week. These data suggest that grafted undifferentiated hiPSCs are able to induce morphological and functional recovery after spinal cord contusion injury.
Collapse
Affiliation(s)
- Tamás Bellák
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Szeged, Kossuth Lajos sgt. 40., 6724, Szeged, Hungary.,BioTalentum Ltd., Gödöllő, Hungary
| | - Zoltán Fekécs
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Szeged, Kossuth Lajos sgt. 40., 6724, Szeged, Hungary
| | - Dénes Török
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Szeged, Kossuth Lajos sgt. 40., 6724, Szeged, Hungary
| | | | - Csilla Nemes
- BioTalentum Ltd., Gödöllő, Hungary.,Department of Diagnostic Laboratory, State Health Centre, Military Hospital, Budapest, Hungary
| | - Zsófia Tézsla
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Szeged, Kossuth Lajos sgt. 40., 6724, Szeged, Hungary
| | - László Gál
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Szeged, Kossuth Lajos sgt. 40., 6724, Szeged, Hungary
| | | | | | - András Dinnyés
- BioTalentum Ltd., Gödöllő, Hungary.,HCEMM-USZ StemCell Research Group, Szeged, Hungary.,Department of Dermatology and Allergology, Research Institute of Translational Biomedicine, University of Szeged, Szeged, Hungary
| | - Antal Nógrádi
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Szeged, Kossuth Lajos sgt. 40., 6724, Szeged, Hungary.
| | - Krisztián Pajer
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Szeged, Kossuth Lajos sgt. 40., 6724, Szeged, Hungary
| |
Collapse
|
20
|
Meftahi GH, Moafi M, Mirbehbahani SH, Fotouhi F, Toreyhi H, Ezi S, Aghajanpour F, Forouzannia A, Boroujeni ME, Peirouvi T, Abbaszadeh HA, Aliaghaei A. Chronic administration of methylphenidate did not affect memory and GDNF levels but increase astrogliosis in adult male rat’s hippocampus. J Chem Neuroanat 2020; 108:101818. [DOI: 10.1016/j.jchemneu.2020.101818] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 05/02/2020] [Accepted: 05/27/2020] [Indexed: 10/24/2022]
|
21
|
Li X, Li M, Tian L, Chen J, Liu R, Ning B. Reactive Astrogliosis: Implications in Spinal Cord Injury Progression and Therapy. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:9494352. [PMID: 32884625 PMCID: PMC7455824 DOI: 10.1155/2020/9494352] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 05/06/2020] [Accepted: 07/31/2020] [Indexed: 12/11/2022]
Abstract
Astrocytes are the most populous glial cells in the central nervous system (CNS). They are essential to CNS physiology and play important roles in the maintenance of homeostasis, development of synaptic plasticity, and neuroprotection. Nevertheless, under the influence of certain factors, astrocytes may also exert detrimental effects through a process of reactive astrogliosis. Previous studies have shown that astrocytes have more than one type of polarization. Two types have been extensively researched. One is a damaging change that occurs under inflammation and has been termed A1 astrocyte, while the other is a restorative change that occurs under ischemic induction and was termed A2 astrocyte. Researchers are now increasingly paying attention to the role of astrocytes in spinal cord injury (SCI), degenerative diseases, chronic pain, neurological tumors, and other CNS disorders. In this review, we discuss (a) the characteristics of polarized astrocytes, (b) the relationship between astrocyte polarization and SCI, and (c) new implications of reactive astrogliosis for future SCI therapies.
Collapse
Affiliation(s)
- Xinyu Li
- Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, No. 105, Jiefang Road, Jinan, Shandong 250013, China
| | - Meng Li
- Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, No. 105, Jiefang Road, Jinan, Shandong 250013, China
| | - Lige Tian
- Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, No. 105, Jiefang Road, Jinan, Shandong 250013, China
| | - Jianan Chen
- Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, No. 105, Jiefang Road, Jinan, Shandong 250013, China
| | - Ronghan Liu
- Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, No. 105, Jiefang Road, Jinan, Shandong 250013, China
| | - Bin Ning
- Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, No. 105, Jiefang Road, Jinan, Shandong 250013, China
| |
Collapse
|
22
|
Itoyama T, Yoshida S, Tomokiyo A, Hasegawa D, Hamano S, Sugii H, Ono T, Fujino S, Maeda H. Possible function of GDNF and Schwann cells in wound healing of periodontal tissue. J Periodontal Res 2020; 55:830-839. [PMID: 32562261 DOI: 10.1111/jre.12774] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 05/07/2020] [Accepted: 05/16/2020] [Indexed: 12/22/2022]
Abstract
OBJECTIVE The purpose of this study was to evaluate the function of Schwann cells in wound healing of periodontal tissue. BACKGROUND In our previous study, glial cell line-derived neurotrophic factor (GDNF) promoted the migration of human periodontal ligament (PDL) cells and that GDNF expression increased in wounded periodontal tissue. GDNF reportedly induces the migration of Schwann cell precursors. Schwann cells play a crucial role in the regeneration of peripheral tissues, including bone tissue. However, the role of Schwann cells on periodontal tissue regeneration remains unclear. METHODS A transwell assay and a WST-1 (water-soluble tetrazolium compound-1) proliferation assay were used to determine whether GDNF promotes the migration and proliferation of Schwann cells, respectively. Quantitative RT-PCR and Alizarin Red S staining were performed to examine the effect of these cells on the differentiation of human preosteoblast (Saos2 cells) using conditioned medium from YST-1 (YST-1-CM). Western blotting analysis was performed to determine whether YST-1-CM activates ERK signaling pathway in Saos2 cells. The expression of Schwann cell markers, S100 calcium-binding protein B (S100-B) and growth associated protein 43 (GAP-43), was determined in normal and wounded periodontal tissue by immunofluorescent staining. RESULTS Glial cell line-derived neurotrophic factor promoted the migration of YST-1 cells but did not affect the proliferation of YST-1 cells. Saos2 cells cultured with YST-1-CM increased the expression of osteoblastic markers and mineralization. YST-1-CM also induced phosphorylation of ERK1/2 in Saos2 cells. The number of S100-B-immunoreactive cells which also expressed GAP-43 was increased in rat wounded periodontal tissue during healing process. CONCLUSION The accumulation of Schwann cells in wounded periodontal tissue suggests that they play a significant role in wound healing of this tissue, especially alveolar bone tissue.
Collapse
Affiliation(s)
- Tomohiro Itoyama
- Department of Endodontology and Operative Dentistry, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | | | - Atsushi Tomokiyo
- Department of Endodontology, Kyushu University Hospital, Fukuoka, Japan
| | - Daigaku Hasegawa
- Department of Endodontology, Kyushu University Hospital, Fukuoka, Japan
| | - Sayuri Hamano
- Department of Endodontology and Operative Dentistry, Faculty of Dental Science, Kyushu University, Fukuoka, Japan.,OBT Research Center, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Hideki Sugii
- Department of Endodontology and Operative Dentistry, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Taiga Ono
- Department of Endodontology and Operative Dentistry, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Shoko Fujino
- Department of Endodontology and Operative Dentistry, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Hidefumi Maeda
- Department of Endodontology and Operative Dentistry, Faculty of Dental Science, Kyushu University, Fukuoka, Japan.,Department of Endodontology, Kyushu University Hospital, Fukuoka, Japan
| |
Collapse
|
23
|
Wang G, He X, Zhu G, Li D, Shi J, Zhang F. Ellagic acid supports neuron by regulating astroglia Nrf2. Biotechnol Appl Biochem 2019; 66:738-743. [DOI: 10.1002/bab.1791] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 06/08/2019] [Indexed: 12/23/2022]
Affiliation(s)
- Guo‐Qing Wang
- Key Laboratory of Basic Pharmacology of Guizhou Zunyi Medical University Zunyi Guizhou People's Republic of China
- Joint International Research Laboratory of Ethnomedicine of Ministry of Education Zunyi Medical University Zunyi Guizhou People's Republic of China
- Key Laboratory of Basic Pharmacology of Ministry of Education Zunyi Medical University Zunyi Guizhou People's Republic of China
| | - Xue‐Mei He
- Key Laboratory of Basic Pharmacology of Guizhou Zunyi Medical University Zunyi Guizhou People's Republic of China
- Joint International Research Laboratory of Ethnomedicine of Ministry of Education Zunyi Medical University Zunyi Guizhou People's Republic of China
- Key Laboratory of Basic Pharmacology of Ministry of Education Zunyi Medical University Zunyi Guizhou People's Republic of China
| | - Guo‐Fu Zhu
- Key Laboratory of Basic Pharmacology of Guizhou Zunyi Medical University Zunyi Guizhou People's Republic of China
- Joint International Research Laboratory of Ethnomedicine of Ministry of Education Zunyi Medical University Zunyi Guizhou People's Republic of China
- Key Laboratory of Basic Pharmacology of Ministry of Education Zunyi Medical University Zunyi Guizhou People's Republic of China
| | - Dai‐Di Li
- Key Laboratory of Basic Pharmacology of Guizhou Zunyi Medical University Zunyi Guizhou People's Republic of China
- Joint International Research Laboratory of Ethnomedicine of Ministry of Education Zunyi Medical University Zunyi Guizhou People's Republic of China
- Key Laboratory of Basic Pharmacology of Ministry of Education Zunyi Medical University Zunyi Guizhou People's Republic of China
| | - Jing‐Shan Shi
- Key Laboratory of Basic Pharmacology of Guizhou Zunyi Medical University Zunyi Guizhou People's Republic of China
- Joint International Research Laboratory of Ethnomedicine of Ministry of Education Zunyi Medical University Zunyi Guizhou People's Republic of China
- Key Laboratory of Basic Pharmacology of Ministry of Education Zunyi Medical University Zunyi Guizhou People's Republic of China
| | - Feng Zhang
- Key Laboratory of Basic Pharmacology of Guizhou Zunyi Medical University Zunyi Guizhou People's Republic of China
- Joint International Research Laboratory of Ethnomedicine of Ministry of Education Zunyi Medical University Zunyi Guizhou People's Republic of China
- Key Laboratory of Basic Pharmacology of Ministry of Education Zunyi Medical University Zunyi Guizhou People's Republic of China
| |
Collapse
|
24
|
Baez-Jurado E, Hidalgo-Lanussa O, Barrera-Bailón B, Sahebkar A, Ashraf GM, Echeverria V, Barreto GE. Secretome of Mesenchymal Stem Cells and Its Potential Protective Effects on Brain Pathologies. Mol Neurobiol 2019; 56:6902-6927. [PMID: 30941733 DOI: 10.1007/s12035-019-1570-x] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 03/18/2019] [Indexed: 02/06/2023]
Abstract
Previous studies have indicated that mesenchymal stem cells (MSCs) have a fundamental role in the repair and regeneration of damaged tissues. There is strong evidence showing that much of the beneficial effects of these cells are due to the secretion of bioactive molecules-besides microRNAs, hormones, and neurotrophins-with anti-inflammatory, immunoregulatory, angiogenic, and trophic effects. These factors have been reported by many studies to possess protective effects on the nervous tissue. Although the beneficial effects of the secretory factors of MSCs have been suggested for various neurological diseases, their actions on astrocytic cells are not well understood. Hence, it is important to recognize the specific effects of MSCs derived from adipose tissue, in addition to the differences presented by the secretome, depending on the source and methods of analysis. In this paper, the different sources of MSCs and their main characteristics are described, as well as the most significant advances in regeneration and protection provided by the secretome of MSCs. Also, we discuss the possible neuroprotective mechanisms of action of the MSC-derived biomolecules, with special emphasis on the effect of MSCs derived from adipose tissue and their impact on glial cells and brain pathologies.
Collapse
Affiliation(s)
- Eliana Baez-Jurado
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, DC, Colombia
| | - Oscar Hidalgo-Lanussa
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, DC, Colombia
| | - Biviana Barrera-Bailón
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, DC, Colombia
| | - Amirhossein Sahebkar
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ghulam Md Ashraf
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Valentina Echeverria
- Facultad de Ciencias de la Salud, Universidad San Sebastian, Lientur 1457, 4080871, Concepción, Chile.,Research & Development Service, Bay Pines VA Healthcare System, Bay Pines, FL, 33744, USA
| | - George E Barreto
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, DC, Colombia.
| |
Collapse
|
25
|
Walker CL, Wu X, Liu NK, Xu XM. Bisperoxovanadium Mediates Neuronal Protection through Inhibition of PTEN and Activation of PI3K/AKT-mTOR Signaling after Traumatic Spinal Injuries. J Neurotrauma 2019; 36:2676-2687. [PMID: 30672370 DOI: 10.1089/neu.2018.6294] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Although mechanisms involved in progression of cell death in spinal cord injury (SCI) have been studied extensively, few are clear targets for translation to clinical application. One of the best-understood mechanisms of cell survival in SCI is phosphatidylinositol-3-kinase (PI3K)/Akt and associated downstream signaling. Clear therapeutic efficacy of a phosphatase and tensin homologue (PTEN) inhibitor called bisperoxovanadium (bpV) has been shown in SCI, traumatic brain injury, stroke, and other neurological disease models in both neuroprotection and functional recovery. The present study aimed to elucidate mechanistic influences of bpV activity in neuronal survival in in vitro and in vivo models of SCI. Treatment with 100 nM bpV(pic) reduced cell death in a primary spinal neuron injury model (p < 0.05) in vitro, and upregulated both Akt and ribosomal protein S6 (pS6) activity (p < 0.05) compared with non-treated injured neurons. Pre-treatment of spinal neurons with a PI3K inhibitor, LY294002 or mammalian target of rapamycin (mTOR) inhibitor, rapamycin blocked bpV activation of Akt and ribosomal protein S6 activity, respectively. Treatment with bpV increased extracellular signal-related kinase (Erk) activity after scratch injury in vitro, and rapamycin reduced influence by bpV on Erk phosphorylation. After a cervical hemicontusive SCI, Akt phosphorylation decreased in total tissue via Western blot analysis (p < 0.01) as well as in penumbral ventral horn motor neurons throughout the first week post-injury (p < 0.05). Conversely, PTEN activity appeared to increase over this period. As observed in vitro, bpV also increased Erk activity post-SCI (p < 0.05). Our results suggest that PI3K/Akt signaling is the likely primary mechanism of bpV action in mediating neuroprotection in injured spinal neurons.
Collapse
Affiliation(s)
- Chandler L Walker
- Department of Biomedical and Applied Sciences, Indiana University School of Dentistry, Indianapolis, Indiana.,Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery and Goodman Campbell Brain and Spine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Xiangbing Wu
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery and Goodman Campbell Brain and Spine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Nai-Kui Liu
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery and Goodman Campbell Brain and Spine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Xiao-Ming Xu
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery and Goodman Campbell Brain and Spine, Indiana University School of Medicine, Indianapolis, Indiana
| |
Collapse
|
26
|
Oliveira-Junior MS, Pereira EP, de Amorim VCM, Reis LTC, do Nascimento RP, da Silva VDA, Costa SL. Lupeol inhibits LPS-induced neuroinflammation in cerebellar cultures and induces neuroprotection associated to the modulation of astrocyte response and expression of neurotrophic and inflammatory factors. Int Immunopharmacol 2019; 70:302-312. [PMID: 30852286 DOI: 10.1016/j.intimp.2019.02.055] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 02/25/2019] [Accepted: 02/28/2019] [Indexed: 12/25/2022]
Abstract
In the central nervous system (CNS), neuroinflammation, especially that modulated by the cell response of astrocytes and microglia, is associated with damage to neurons in neurodegenerative disorders such as Parkinson's disease, Alzheimer's disease and, Multiple Sclerosis. Lupeol is a dietary triterpene that has demonstrated biological activities as antioxidant. This study investigated the anti-inflammatory and neuroprotective effects of lupeol in an in vitro model of neuroinflammation in primary cerebellar cultures. Cultures were obtained from 6-day-old Wistar rats, subjected to inflammatory damage with lipopolysaccharide (LPS, 1 μg/mL) and treated with lupeol (0.1 μM). We observed, after a 48-hour treatment, through Fluorjade-B staining and immunocytochemistry (ICQ) for βIII-tubulin, that lupeol induced neuroprotection in cultures submitted to inflammatory damage. On the other hand, through ICQ for GFAP, it was possible to observe that lupeol modulated the astrocyte morphology for Bergmann glia-like phenotype and, especially for velate astrocyte-like phenotype, both phenotypes associated with the neuroprotective profile. Moreover, RT-qPCR analysis showed that lupeol induced the down-regulation of the mRNA expression for proinflammatory markers TNF, iNOS and NLRP3, as well as the production of nitric oxide (method of Greiss), which were up-regulated by LPS, and also induced up-regulation of the mRNA expression for arginase and IL-6 mRNA. In addition, lupeol induced up-regulation of mRNA expression for neurotrophins GDNF and NGF and also for the sonic hedgehog-Gli pathway. Together, these results lead to the conclusion that lupeol inhibits neuroinflammation in cerebellar cultures and induces neuroprotection associated with the modulation of astrocyte response and expression of neurotrophic and inflammatory factors.
Collapse
Affiliation(s)
| | - Erica Patricia Pereira
- Department of Biochemistry and Biophysics, Institute of Health Sciences, Federal University of Bahia, Brazil
| | | | - Luã Tainã Costa Reis
- Department of Biochemistry and Biophysics, Institute of Health Sciences, Federal University of Bahia, Brazil
| | | | | | - Silvia Lima Costa
- Department of Biochemistry and Biophysics, Institute of Health Sciences, Federal University of Bahia, Brazil.
| |
Collapse
|
27
|
Naderain H, Khanlarkhani N, Ragerdi Kashani I, Atlasi A, Atlasi MA. Comparison of the effects of progesterone and 17 β-estradiol on Schwann cell markers expression in rat adipose-derived stem cells. VETERINARY RESEARCH FORUM : AN INTERNATIONAL QUARTERLY JOURNAL 2019; 9:307-313. [PMID: 30713608 PMCID: PMC6346486 DOI: 10.30466/vrf.2018.33103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 02/21/2018] [Indexed: 11/22/2022]
Abstract
Steroids promote the myelination and regeneration in the peripheral nervous system. Whereas, little is known about the inducing effects by which the hormones exert their effects on Schwann cells differentiation. This could be revealed by the expression of Schwann cell markers in adipose-derived stem cells (ADSCs). The purpose of this study was to present the effects of progesterone and 17 β-estradiol on the Schwann cell markers in rat ADSCs. The mesenchymal stem cell markers (CD73, and CD90) were assayed by flow cytometry. Rat ADSCs were sequentially treated with β-mercaptoethanol, and all-trans-retinoic acid, followed by a mixture of basic fibrobroblast growth factor, platelet-derived growth factor, forskolin and heregulin. In experimental groups, forskolin and heregulin were substituted by progesterone and 17 β-estradiol. After induction, the expression of Schwann cell markers P0, and S-100 and the cellular immunocytochemical staining positive rate of anti-S100 and anti-glial fibrillary acidic protein (GFAP) antibodies were compared in the experimental and control groups. Progesterone and 17 β-estradiol triggered P0 and S-100 genes expression and induced a cellular immunocytochemical staining positive rate of S-100 and GFAP in rats ADSCs. Progesterone induced these changes stronger than 17 β-estradiol. Thus, progesterone may induce rat ADSCs toward Schwann-like cells by expression of Schwann cell markers and is more potent than 17 β-estradiol in the expression of these markers.
Collapse
Affiliation(s)
- Homayoun Naderain
- Anatomical Sciences Research Center, Kashan University of Medical Sciences, Kashan, Iran
| | - Neda Khanlarkhani
- Department of Anatomy, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Iraj Ragerdi Kashani
- Department of Anatomy, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Amirabbas Atlasi
- Student Research Committee, Faculty of Dentistry, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohammad Ali Atlasi
- Anatomical Sciences Research Center, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
28
|
Mitochondrial Neuroglobin Is Necessary for Protection Induced by Conditioned Medium from Human Adipose-Derived Mesenchymal Stem Cells in Astrocytic Cells Subjected to Scratch and Metabolic Injury. Mol Neurobiol 2018; 56:5167-5187. [PMID: 30536184 DOI: 10.1007/s12035-018-1442-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 11/29/2018] [Indexed: 12/27/2022]
Abstract
Astrocytes are specialized cells capable of regulating inflammatory responses in neurodegenerative diseases or traumatic brain injury. In addition to playing an important role in neuroinflammation, these cells regulate essential functions for the preservation of brain tissue. Therefore, the search for therapeutic alternatives to preserve these cells and maintain their functions contributes in some way to counteract the progress of the injury and maintain neuronal survival in various brain pathologies. Among these strategies, the conditioned medium from human adipose-derived mesenchymal stem cells (CM-hMSCA) has been reported with a potential beneficial effect against several neuropathologies. In this study, we evaluated the potential effect of CM-hMSCA in a model of human astrocytes (T98G cells) subjected to scratch injury. Our findings demonstrated that CM-hMSCA regulates the cytokines IL-2, IL-6, IL-8, IL-10, GM-CSF, and TNF-α, downregulates calcium at the cytoplasmic level, and regulates mitochondrial dynamics and the respiratory chain. These actions are accompanied by modulation of the expression of different proteins involved in signaling pathways such as AKT/pAKT and ERK1/2/pERK, and may mediate the localization of neuroglobin (Ngb) at the cellular level. We also confirmed that Ngb mediated the protective effects of CM-hMSCA through regulation of proteins involved in survival pathways and oxidative stress. In conclusion, regulation of brain inflammation combined with the recovery of fundamental cellular aspects in the face of injury makes CM-hMSCA a promising candidate for the protection of astrocytes in brain pathologies.
Collapse
|
29
|
History of Glial Cell Line-Derived Neurotrophic Factor (GDNF) and Its Use for Spinal Cord Injury Repair. Brain Sci 2018; 8:brainsci8060109. [PMID: 29899247 PMCID: PMC6025482 DOI: 10.3390/brainsci8060109] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 06/10/2018] [Accepted: 06/11/2018] [Indexed: 01/01/2023] Open
Abstract
Following an initial mechanical insult, traumatic spinal cord injury (SCI) induces a secondary wave of injury, resulting in a toxic lesion environment inhibitory to axonal regeneration. This review focuses on the glial cell line-derived neurotrophic factor (GDNF) and its application, in combination with other factors and cell transplantations, for repairing the injured spinal cord. As studies of recent decades strongly suggest that combinational treatment approaches hold the greatest therapeutic potential for the central nervous system (CNS) trauma, future directions of combinational therapies will also be discussed.
Collapse
|
30
|
Zhang LQ, Zhang WM, Deng L, Xu ZX, Lan WB, Lin JH. Transplantation of a Peripheral Nerve with Neural Stem Cells Plus Lithium Chloride Injection Promote the Recovery of Rat Spinal Cord Injury. Cell Transplant 2018; 27:471-484. [PMID: 29756516 PMCID: PMC6038036 DOI: 10.1177/0963689717752945] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Transplantation of neural stem cells (NSCs) holds great potential for the treatment of spinal cord injury (SCI). However, transplanted NSCs poorly survive in the SCI environment. We injected NSCs into tibial nerve and transplanted tibial nerve into a hemisected spinal cord and investigated the effects of lithium chloride (LiCl) on the survival of spinal neurons, axonal regeneration, and functional recovery. Our results show that most of the transplanted NSCs expressed glial fibrillary acidic protein, while there was no obvious expression of nestin, neuronal nuclei, or acetyltransferase found in NSCs. LiCl treatment produced less macrosialin (ED1) expression and axonal degeneration in tibial nerve after NSC injection. Our results also show that a regimen of LiCl treatment promoted NSC differentiation into NF200-positive neurons with neurite extension into the host spinal cord. The combination of tibial nerve transplantation with NSCs and LiCl injection resulted in more host motoneurons surviving in the spinal cord, more regenerated axons in tibial nerve, less glial scar area, and decreased ED1 expression. We conclude that lithium may have therapeutic potential in cell replacement strategies for central nervous system injury due to its ability to promote survival and neuronal generation of grafted NSCs and reduced host immune reaction.
Collapse
Affiliation(s)
- Li-Qun Zhang
- 1 Department of Orthopedics, the First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Wen-Ming Zhang
- 1 Department of Orthopedics, the First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Lingxiao Deng
- 2 Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA.,3 Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Zi-Xing Xu
- 1 Department of Orthopedics, the First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Wen-Bin Lan
- 1 Department of Orthopedics, the First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Jian-Hua Lin
- 1 Department of Orthopedics, the First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| |
Collapse
|
31
|
Liu S, Schackel T, Weidner N, Puttagunta R. Biomaterial-Supported Cell Transplantation Treatments for Spinal Cord Injury: Challenges and Perspectives. Front Cell Neurosci 2018; 11:430. [PMID: 29375316 PMCID: PMC5768640 DOI: 10.3389/fncel.2017.00430] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 12/20/2017] [Indexed: 12/17/2022] Open
Abstract
Spinal cord injury (SCI), resulting in para- and tetraplegia caused by the partial or complete disruption of descending motor and ascending sensory neurons, represents a complex neurological condition that remains incurable. Following SCI, numerous obstacles comprising of the loss of neural tissue (neurons, astrocytes, and oligodendrocytes), formation of a cavity, inflammation, loss of neuronal circuitry and function must be overcome. Given the multifaceted primary and secondary injury events that occur with SCI treatment options are likely to require combinatorial therapies. While several methods have been explored, only the intersection of two, cell transplantation and biomaterial implantation, will be addressed in detail here. Owing to the constant advance of cell culture technologies, cell-based transplantation has come to the forefront of SCI treatment in order to replace/protect damaged tissue and provide physical as well as trophic support for axonal regrowth. Biomaterial scaffolds provide cells with a protected environment from the surrounding lesion, in addition to bridging extensive damage and providing physical and directional support for axonal regrowth. Moreover, in this combinatorial approach cell transplantation improves scaffold integration and therefore regenerative growth potential. Here, we review the advances in combinatorial therapies of Schwann cells (SCs), astrocytes, olfactory ensheathing cells (OECs), mesenchymal stem cells, as well as neural stem and progenitor cells (NSPCs) with various biomaterial scaffolds.
Collapse
Affiliation(s)
- Shengwen Liu
- Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Thomas Schackel
- Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| | - Norbert Weidner
- Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| | - Radhika Puttagunta
- Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
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: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Among various models for spinal cord injury in rats, the contusion model is the most often used because it is the most common type of human spinal cord injury. The complete transection model, although not as clinically relevant as the contusion model, is the most rigorous method to evaluate axon regeneration. In the contusion model, it is difficult to distinguish regenerated from sprouted or spared axons due to the presence of remaining tissue post injury. In the complete transection model, a bridging method is necessary to fill the gap and create continuity from the rostral to the caudal stumps in order to evaluate the effectiveness of the treatments. A reliable bridging surgery is essential to test outcome measures by reducing the variability due to the surgical method. The protocols described here are used to prepare Schwann cells (SCs) and conduits prior to transplantation, complete transection of the spinal cord at thoracic level 8 (T8), insert the conduit, and transplant SCs into the conduit. This approach also uses in situ gelling of an injectable basement membrane matrix with SC transplantation that allows improved axon growth across the rostral and caudal interfaces with the host tissue.
Collapse
Affiliation(s)
- Yee-Shuan Lee
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine
| | - Siliang Wu
- Department of Materials Science and Engineering, New Jersey Institute of Technology
| | | | - Mary Bartlett Bunge
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine; Department of Cell Biology, University of Miami Miller School of Medicine; Department of Neurological Surgery, University of Miami Miller School of Medicine;
| |
Collapse
|
33
|
Postinjury Induction of Activated ErbB2 Selectively Hyperactivates Denervated Schwann Cells and Promotes Robust Dorsal Root Axon Regeneration. J Neurosci 2017; 37:10955-10970. [PMID: 28982707 DOI: 10.1523/jneurosci.0903-17.2017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 09/20/2017] [Accepted: 09/27/2017] [Indexed: 11/21/2022] Open
Abstract
Following nerve injury, denervated Schwann cells (SCs) convert to repair SCs, which enable regeneration of peripheral axons. However, the repair capacity of SCs and the regenerative capacity of peripheral axons are limited. In the present studies we examined a potential therapeutic strategy to enhance the repair capacity of SCs, and tested its efficacy in enhancing regeneration of dorsal root (DR) axons, whose regenerative capacity is particularly weak. We used male and female mice of a doxycycline-inducible transgenic line to induce expression of constitutively active ErbB2 (caErbB2) selectively in SCs after DR crush or transection. Two weeks after injury, injured DRs of induced animals contained far more SCs and SC processes. These SCs had not redifferentiated and continued to proliferate. Injured DRs of induced animals also contained far more axons that regrew along SC processes past the transection or crush site. Remarkably, SCs and axons in uninjured DRs remained quiescent, indicating that caErbB2 enhanced regeneration of injured DRs, without aberrantly activating SCs and axons in intact nerves. We also found that intraspinally expressed glial cell line-derived neurotrophic factor (GDNF), but not the removal of chondroitin sulfate proteoglycans, greatly enhanced the intraspinal migration of caErbB2-expressing SCs, enabling robust penetration of DR axons into the spinal cord. These findings indicate that SC-selective, post-injury activation of ErbB2 provides a novel strategy to powerfully enhance the repair capacity of SCs and axon regeneration, without substantial off-target damage. They also highlight that promoting directed migration of caErbB2-expressing SCs by GDNF might be useful to enable axon regrowth in a non-permissive environment.SIGNIFICANCE STATEMENT Repair of injured peripheral nerves remains a critical clinical problem. We currently lack a therapy that potently enhances axon regeneration in patients with traumatic nerve injury. It is extremely challenging to substantially increase the regenerative capacity of damaged nerves without deleterious off-target effects. It was therefore of great interest to discover that caErbB2 markedly enhances regeneration of damaged dorsal roots, while evoking little change in intact roots. To our knowledge, these findings are the first demonstration that repair capacity of denervated SCs can be efficaciously enhanced without altering innervated SCs. Our study also demonstrates that oncogenic ErbB2 signaling can be activated in SCs but not impede transdifferentiation of denervated SCs to regeneration-promoting repair SCs.
Collapse
|
34
|
Zhao YZ, Jiang X, Lin Q, Xu HL, Huang YD, Lu CT, Cai J. Thermosensitive heparin-poloxamer hydrogels enhance the effects of GDNF on neuronal circuit remodeling and neuroprotection after spinal cord injury. J Biomed Mater Res A 2017; 105:2816-2829. [PMID: 28593744 DOI: 10.1002/jbm.a.36134] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 05/14/2017] [Accepted: 06/01/2017] [Indexed: 12/22/2022]
Abstract
Traumatic spinal cord injury (SCI) results in paraplegia or quadriplegia, and currently, therapeutic interventions for axonal regeneration after SCI are not clinically available. Animal studies have revealed that glial cell-derived neurotrophic factor (GDNF) plays multiple beneficial roles in neuroprotection, glial scarring remodeling, axon regeneration and remyelination in SCI. However, the poor physicochemical stability of GDNF, as well as its limited ability to cross the blood-spinal cord barrier, hampers the development of GDNF as an effective therapeutic intervention in clinical practice. In this study, a novel temperature-sensitive heparin-poloxamer (HP) hydrogel with high GDNF-binding affinity was developed. HP hydrogels showed a supporting scaffold for GDNF when it was injected into the lesion epicenter after SCI. GDNF-HP by orthotopic injection on lesioned spinal cord promoted the beneficial effects of GDNF on neural stem cell proliferation, reactive astrogliosis inhibition, axonal regeneration or plasticity, neuroprotection against cell apoptosis, and body functional recovery. Most interestingly, GDNF demonstrated a bidirectional regulation of autophagy, which inhibited cell apoptosis at different stages of SCI. Furthermore, the HP hydrogel promoted the inhibition of autophagy-induced apoptosis by GDNF in SCI. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 105A: 2816-2829, 2017.
Collapse
Affiliation(s)
- Ying-Zheng Zhao
- The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, People's Republic of China.,College of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, People's Republic of China.,Hainan Medical College, Haikou, Hainan, 570102, People's Republic of China
| | - Xi Jiang
- The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, People's Republic of China.,Zhejiang University Mingzhou Hospital, Zhejiang, 315104, People's Republic of China
| | - Qian Lin
- College of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, People's Republic of China.,Kosair Children's Hospital Research Institute at the Department of Pediatrics, University of Louisville School of Medicine, Louisville, Kentucky, 40202
| | - He-Lin Xu
- College of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, People's Republic of China
| | - Ya-Dong Huang
- Biopharmaceutical R&D Center of Jinan University, Guangzhou, Guangdong, 510000, People's Republic of China
| | - Cui-Tao Lu
- The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, People's Republic of China.,College of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, People's Republic of China
| | - Jun Cai
- The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, People's Republic of China.,Kosair Children's Hospital Research Institute at the Department of Pediatrics, University of Louisville School of Medicine, Louisville, Kentucky, 40202
| |
Collapse
|
35
|
Chen BK, Madigan NN, Hakim JS, Dadsetan M, McMahon SS, Yaszemski MJ, Windebank AJ. GDNF Schwann cells in hydrogel scaffolds promote regional axon regeneration, remyelination and functional improvement after spinal cord transection in rats. J Tissue Eng Regen Med 2017; 12:e398-e407. [DOI: 10.1002/term.2431] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Revised: 02/08/2017] [Accepted: 02/24/2017] [Indexed: 11/07/2022]
Affiliation(s)
- Bingkun K. Chen
- Department of Neurology, Mayo Clinic College of Medicine; Mayo Clinic; Rochester Minnesota
| | - Nicolas N. Madigan
- Department of Neurology, Mayo Clinic College of Medicine; Mayo Clinic; Rochester Minnesota
| | - Jeffrey S. Hakim
- Department of Neurology, Mayo Clinic College of Medicine; Mayo Clinic; Rochester Minnesota
| | - Mahrokh Dadsetan
- Department of Orthopedic Surgery; Mayo Clinic College of Medicine; Rochester Minnesota
| | - Siobhan S. McMahon
- Department of Medicine; Regenerative Medicine Institute (REMEDI), National University of Ireland; Galway
| | - Michael J. Yaszemski
- Department of Orthopedic Surgery; Mayo Clinic College of Medicine; Rochester Minnesota
| | - Anthony J. Windebank
- Department of Neurology, Mayo Clinic College of Medicine; Mayo Clinic; Rochester Minnesota
| |
Collapse
|
36
|
Sotoyama H, Iwakura Y, Oda K, Sasaoka T, Takei N, Kakita A, Enomoto H, Nawa H. Striatal hypodopamine phenotypes found in transgenic mice that overexpress glial cell line-derived neurotrophic factor. Neurosci Lett 2017. [PMID: 28645787 DOI: 10.1016/j.neulet.2017.06.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Glial cell line-derived neurotrophic factor (GDNF) positively regulates the development and maintenance of in vitro dopaminergic neurons. However, the in vivo influences of GDNF signals on the brain dopamine system are controversial and not fully defined. To address this question, we analyzed dopaminergic phenotypes of the transgenic mice that overexpress GDNF under the control of the glial Gfap promoter. Compared with wild-type, the GDNF transgenic mice contained higher levels of GDNF protein and phosphorylated RET receptors in the brain. However, there were reductions in the levels of tyrosine hydroxylase (TH), dopamine, and its metabolite homovanillic acid in the striatum of transgenic mice. The TH reduction appeared to occur during postnatal development. Immunohistochemistry revealed that striatal TH density was reduced in transgenic mice with no apparent signs of neurodegeneration. In agreement with these neurochemical traits, basal levels of extracellular dopamine and high K+-induced dopamine efflux were decreased in the striatum of transgenic mice. We also explored the influences of GDNF overexpression on lomomotor behavior. GDNF transgenic mice exhibited lower stereotypy and rearing in a novel environment compared with wild-type mice. These results suggest that chronic overexpression of GDNF in brain astrocytes exerts an opposing influence on nigrostriatal dopamine metabolism and neurotransmission.
Collapse
Affiliation(s)
- Hidekazu Sotoyama
- Department of Molecular Neurobiology, Brain Research Institute, Niigata University, Japan
| | - Yuriko Iwakura
- Department of Molecular Neurobiology, Brain Research Institute, Niigata University, Japan
| | - Kanako Oda
- Department of Comparative and Experimental Medicine, Brain Research Institute, Niigata University, Japan
| | - Toshikuni Sasaoka
- Department of Comparative and Experimental Medicine, Brain Research Institute, Niigata University, Japan
| | - Nobuyuki Takei
- Department of Molecular Neurobiology, Brain Research Institute, Niigata University, Japan
| | - Akiyoshi Kakita
- Department of Pathology, Brain Research Institute, Niigata University, Japan
| | - Hideki Enomoto
- Laboratory for Neural Differentiation and Regeneration, Graduate School of Medicine, Kobe University, Japan
| | - Hiroyuki Nawa
- Department of Molecular Neurobiology, Brain Research Institute, Niigata University, Japan.
| |
Collapse
|
37
|
Nejati-Koshki K, Mortazavi Y, Pilehvar-Soltanahmadi Y, Sheoran S, Zarghami N. An update on application of nanotechnology and stem cells in spinal cord injury regeneration. Biomed Pharmacother 2017; 90:85-92. [DOI: 10.1016/j.biopha.2017.03.035] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2017] [Revised: 03/12/2017] [Accepted: 03/14/2017] [Indexed: 02/08/2023] Open
|
38
|
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: 1.4] [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
|
39
|
Huang L, Xia B, Liu Z, Cao Q, Huang J, Luo Z. Superparamagnetic Iron Oxide Nanoparticle-Mediated Forces Enhance the Migration of Schwann Cells Across the Astrocyte-Schwann Cell Boundary In vitro. Front Cell Neurosci 2017; 11:83. [PMID: 28400720 PMCID: PMC5368970 DOI: 10.3389/fncel.2017.00083] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Accepted: 03/10/2017] [Indexed: 12/20/2022] Open
Abstract
Schwann cells (SCs) are one of the most promising cellular candidates for the treatment of spinal cord injury. However, SCs show poor migratory ability within the astrocyte-rich central nervous system (CNS) environment and exhibit only limited integration with host astrocytes. Our strategy for improving the therapeutic potential of SCs was to magnetically drive SCs to migrate across the astrocyte-SC boundary to intermingle with astrocytes. SCs were firstly magnetized with poly-L-lysine-coated superparamagnetic iron oxide nanoparticles (SPIONs). Internalization of SPIONs showed no effect upon the migration of SCs in the absence of a magnetic field (MF). In contrast, magnetized SCs exhibited enhanced migration along the direction of force in the presence of a MF. An inverted coverslip assay showed that a greater number of magnetized SCs migrated longer distances onto astrocytic monolayers under the force of a MF compared to other test groups. More importantly, a confrontation assay demonstrated that magnetized SCs intermingled with astrocytes under an applied MF. Furthermore, inhibition of integrin activation reduced the migration of magnetized SCs within an astrocyte-rich environment under an applied MF. Thus, SPION-mediated forces could act as powerful stimulants to enhance the migration of SCs across the astrocyte-SC boundary, via integrin-mediated mechanotransduction, and could represent a vital way of improving the therapeutic potential of SCs for spinal cord injuries.
Collapse
Affiliation(s)
- Liangliang Huang
- Department of Orthopaedics, Xijing Hospital, The Fourth Military Medical University Xi'an, China
| | - Bing Xia
- Department of Orthopaedics, Xijing Hospital, The Fourth Military Medical University Xi'an, China
| | - Zhongyang Liu
- Department of Orthopaedics, Xijing Hospital, The Fourth Military Medical University Xi'an, China
| | - Quanliang Cao
- State Key Laboratory of Advanced Electromagnetic Engineering and Technology, Wuhan National High Magnetic Field Center, Huazhong University of Science and Technology Wuhan, China
| | - Jinghui Huang
- Department of Orthopaedics, Xijing Hospital, The Fourth Military Medical University Xi'an, China
| | - Zhuojing Luo
- Department of Orthopaedics, Xijing Hospital, The Fourth Military Medical University Xi'an, China
| |
Collapse
|
40
|
Baez-Jurado E, Vega GG, Aliev G, Tarasov VV, Esquinas P, Echeverria V, Barreto GE. Blockade of Neuroglobin Reduces Protection of Conditioned Medium from Human Mesenchymal Stem Cells in Human Astrocyte Model (T98G) Under a Scratch Assay. Mol Neurobiol 2017; 55:2285-2300. [PMID: 28332151 DOI: 10.1007/s12035-017-0481-y] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 03/03/2017] [Indexed: 12/25/2022]
Abstract
Previous studies have indicated that paracrine factors (conditioned medium) increase wound closure and reduce reactive oxygen species in a traumatic brain injury in vitro model. Although the beneficial effects of conditioned medium from human adipose tissue-derived mesenchymal stem cells (hMSCA-CM) have been previously suggested for various neurological diseases, their actions on astrocytic cells are not well understood. In this study, we have explored the effect of hMSCA-CM on human astrocyte model (T98G cells) subjected to scratch assay. Our results indicated that hMSCA-CM improved cell viability, reduced nuclear fragmentation, attenuated the production of reactive oxygen species, and preserved mitochondrial membrane potential and ultrastructural parameters. In addition, hMSCA-CM upregulated neuroglobin in T98G cells and the genetic silencing of this protein prevented the protective action of hMSCA-CM on damaged cells, suggesting that neuroglobin is mediating, at least in part, the protective effect of hMSCA-CM. Overall, this evidence suggests that the use of hMSCA-CM is a promising therapeutic strategy for the protection of astrocytic cells in central nervous system (CNS) pathologies.
Collapse
Affiliation(s)
- Eliana Baez-Jurado
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, DC, Colombia
| | - Gina Guio Vega
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, DC, Colombia
| | - Gjumrakch Aliev
- Institute of Physiologically Active Compounds Russian Academy of Sciences, Chernogolovka, 142432, Russia
- GALLY International Biomedical Research Consulting LLC, San Antonio, TX, 78229, USA
- School of Health Science and Healthcare Administration, University of Atlanta, Johns Creek, GA, 30097, USA
| | - Vadim V Tarasov
- Institute of Pharmacy and Translational Medicine, Sechenov First Moscow State Medical University, 2-4 Bolshaya Pirogovskaya st., 119991, Moscow, Russia
| | - Paula Esquinas
- Facultad Medicina Veterinaria y Zootecnia, Universidad Nacional de Colombia, Bogotá, Colombia
| | - Valentina Echeverria
- Facultad Ciencias de la Salud, Universidad San Sebastián, Lientur 1457, 4030000, Concepción, Chile
| | - George E Barreto
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, DC, Colombia.
- Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Santiago, Chile.
| |
Collapse
|
41
|
Nori S, Nakamura M, Okano H. Plasticity and regeneration in the injured spinal cord after cell transplantation therapy. PROGRESS IN BRAIN RESEARCH 2017; 231:33-56. [DOI: 10.1016/bs.pbr.2016.12.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
42
|
Hodgetts SI, Harvey AR. Neurotrophic Factors Used to Treat Spinal Cord Injury. VITAMINS AND HORMONES 2016; 104:405-457. [PMID: 28215303 DOI: 10.1016/bs.vh.2016.11.007] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The application of neurotrophic factors as a therapy to improve morphological and behavioral outcomes after experimental spinal cord injury (SCI) has been the focus of many studies. These studies vary markedly in the type of neurotrophic factor that is delivered, the mode of administration, and the location, timing, and duration of the treatment. Generally, the majority of studies have had significant success if neurotrophic factors are applied in or close to the lesion site during the acute or the subacute phase after SCI. Comparatively fewer studies have administered neurotrophic factors in order to directly target the somata of injured neurons. The mode of delivery varies between acute injection of recombinant proteins, subacute or chronic delivery using a variety of strategies including osmotic minipumps, cell-mediated delivery, delivery using polymer release vehicles or supporting bridges of some sort, or the use of gene therapy to modify neurons, glial cells, or precursor/stem cells. In this brief review, we summarize the state of play of many of the therapies using these factors, most of which have been undertaken in rodent models of SCI.
Collapse
Affiliation(s)
- S I Hodgetts
- School of Anatomy, Physiology and Human Biology, The University of Western Australia, Perth, WA, Australia; Western Australian Neuroscience Research Institute, Perth, WA, Australia.
| | - A R Harvey
- School of Anatomy, Physiology and Human Biology, The University of Western Australia, Perth, WA, Australia; Western Australian Neuroscience Research Institute, Perth, WA, Australia
| |
Collapse
|
43
|
Wang Y, Li WY, Jia H, Zhai FG, Qu WR, Cheng YX, Liu YC, Deng LX, Guo SF, Jin ZS. KLF7-transfected Schwann cell graft transplantation promotes sciatic nerve regeneration. Neuroscience 2016; 340:319-332. [PMID: 27826105 DOI: 10.1016/j.neuroscience.2016.10.069] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 10/24/2016] [Accepted: 10/29/2016] [Indexed: 11/16/2022]
Abstract
Our former study demonstrated that Krüppel-like Factor 7 (KLF7) is a transcription factor that stimulates axonal regeneration after peripheral nerve injury. Currently, we used a gene therapy approach to overexpress KLF7 in Schwann cells (SCs) and assessed whether KLF7-transfected SCs graft could promote sciatic nerve regeneration. SCs were transfected by adeno-associated virus 2 (AAV2)-KLF7 in vitro. Mice were allografted by an acellular nerve (ANA) with either an injection of DMEM (ANA group), SCs (ANA+SCs group) or AAV2-KLF7-transfected SCs (ANA+KLF7-SCs group) to assess repair of a sciatic nerve gap. The results indicate that KLF7 overexpression promoted the proliferation of both transfected SCs and native SCs. The neurite length of the dorsal root ganglia (DRG) explants was enhanced. Several beneficial effects were detected in the ANA+KLF7-SCs group including an increase in the compound action potential amplitude, sciatic function index score, enhanced expression of PKH26-labeling transplant SCs, peripheral myelin protein 0, neurofilaments, S-100, and myelinated regeneration nerve. Additionally, HRP-labeled motoneurons in the spinal cord, CTB-labeled sensory neurons in the DRG, motor endplate density and the weight ratios of target muscles were increased by the treatment while thermal hyperalgesia was diminished. Finally, expression of KLF7, NGF, GAP43, TrkA and TrkB were enhanced in the grafted SCs, which may indicate that several signal pathways may be involved in conferring the beneficial effects from KLF7 overexpression. We concluded that KLF7-overexpressing SCs promoted axonal regeneration of the peripheral nerve and enhanced myelination, which collectively proved KLF-SCs as a novel therapeutic strategy for injured nerves.
Collapse
Affiliation(s)
- Ying Wang
- Department of Anatomy, Mudanjiang College of Medicine, Mudanjiang 157011, China
| | - Wen-Yuan Li
- Department of Anatomy, Mudanjiang College of Medicine, Mudanjiang 157011, China.
| | - Hua Jia
- Department of Anatomy, Ningxia Medical University, Yinchuan 750004, China
| | - Feng-Guo Zhai
- Department of Pharmacology, Mudanjiang College of Medicine, Mudanjiang 157011, China
| | - Wen-Rui Qu
- Hand & Foot Surgery and Reparative & Reconstructive Surgery Center, Orthopaedic Hospital of the Second Hospital of Jilin University, Changchun 130041, China
| | - Yong-Xia Cheng
- Department of Pathology, Mudanjiang College of Medicine, Mudanjiang 157011, China
| | - Yan-Cui Liu
- Department of Anatomy, Mudanjiang College of Medicine, Mudanjiang 157011, China
| | - Ling-Xiao Deng
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Su-Fen Guo
- Hand & Foot Surgery and Reparative & Reconstructive Surgery Center, Orthopaedic Hospital of the Second Hospital of Jilin University, Changchun 130041, China
| | - Zai-Shun Jin
- Hand & Foot Surgery and Reparative & Reconstructive Surgery Center, Orthopaedic Hospital of the Second Hospital of Jilin University, Changchun 130041, China
| |
Collapse
|
44
|
Önger ME, Delibaş B, Türkmen AP, Erener E, Altunkaynak BZ, Kaplan S. The role of growth factors in nerve regeneration. Drug Discov Ther 2016; 10:285-291. [PMID: 27746416 DOI: 10.5582/ddt.2016.01058] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Nerve injuries result in functional loss in the innervated organ or body parts, and recovery is difficult unless surgical treatment has been done. Different surgical treatments have been suggested for nerve repair. Tissue engineering related to growth factors has arisen as an alternative approach for triggering and improving nerve regeneration. Therefore, the aim of this review is to provide a comprehensive analysis related to growth factors as tools for optimizing the regeneration process. Studies and reviews on the use of growth factors for nerve regeneration were compiled over the course of the review. According to literature review, it may be concluded that growth factors from different sources present promising treatment related to nerve regeneration involved in neuronal differentiation, greater myelination and axonal growth and proliferation of specific cells for nerve repair.
Collapse
Affiliation(s)
- Mehmet Emin Önger
- Department of Histology and Embryology, Faculty of Medicine, Ondokuz Mayis University
| | | | | | | | | | | |
Collapse
|
45
|
Hosseini SR, Kaka G, Joghataei MT, Hooshmandi M, Sadraie SH, Yaghoobi K, Mohammadi A. Assessment of Neuroprotective Properties of Melissa officinalis in Combination With Human Umbilical Cord Blood Stem Cells After Spinal Cord Injury. ASN Neuro 2016; 8:1759091416674833. [PMID: 27815336 PMCID: PMC5098695 DOI: 10.1177/1759091416674833] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2016] [Revised: 05/17/2016] [Accepted: 07/06/2016] [Indexed: 01/05/2023] Open
Abstract
INTRODUCTION The pathophysiology of spinal cord injury (SCI) has a classically bad prognosis. It has been demonstrated that human umbilical cord blood stem cells (hUCBSCs) and Melissa officinalis (MO) are useful for the prevention of neurological disease. METHODS Thirty-six adult male rats were randomly divided into intact, sham, control (SCI), MO, hUCBSC, and MO-hUCBSC groups. Intraperitoneal injection of MO (150 mg/kg) was commenced 24 hr post-SCI and continued once a day for 14 days. Intraspinal grafting of hUCBSCs was commenced immediately in the next day. The motor and sensory functions of all animals were evaluated once a week after the commencement of SCI. Electromyography (EMG) was performed in the last day in order to measure the recruitment index. Immunohistochemistry, reverse transcription-polymerase chain reaction, and transmission electron microscopy evaluations were performed to determine the level of astrogliosis and myelination. RESULTS The results revealed that motor function (MO-hUCBSC: 15 ± 0.3, SCI: 8.2 ± 0.37, p < .001), sensory function (MO-hUCBSC: 3.57 ± 0.19, SCI: 6.38 ± 0.23, p < .001), and EMG recruitment index (MO-hUCBSC: 3.71 ± 0.18, SCI: 1.6 ± 0.1, p < .001) were significantly improved in the MO-hUCBSC group compared with SCI group. Mean cavity area (MO-hUCBSC: 0.03 ± 0.03, SCI: 0.07 ± 0.004, p < .001) was reduced and loss of lower motor neurons (MO-hUCBSC: 7.6 ± 0.43, SCI: 3 ± 0.12, p < .001) and astrogliosis density (MO-hUCBSC: 3.1 ± 0.15, SCI: 6.25 ± 1.42, p < 0.001) in the ventral horn of spinal cord were prevented in MO-hUCBSC group compared with SCI group. CONCLUSION The results revealed that the combination of MO and hUCBSCs in comparison with the control group has neuroprotective effects in SCI.
Collapse
Affiliation(s)
| | - Gholamreza Kaka
- Neuroscience Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | | | - Mehdi Hooshmandi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed Homayoon Sadraie
- Department of Anatomy, School of Medicine, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Kayvan Yaghoobi
- Neuroscience Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Alireza Mohammadi
- Neuroscience Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
46
|
Combination of grafted Schwann cells and lentiviral-mediated prevention of glial scar formation improve recovery of spinal cord injured rats. J Chem Neuroanat 2016; 76:48-60. [DOI: 10.1016/j.jchemneu.2015.12.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Revised: 11/26/2015] [Accepted: 12/25/2015] [Indexed: 01/03/2023]
|
47
|
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: 45] [Impact Index Per Article: 5.0] [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
|
48
|
Qiu DL, Wang TH. The Expression implication of GDNF in ventral horn and associated remote cortex in rhesus monkeys with hemisected spinal cord injury. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2016; 19:970-976. [PMID: 27803784 PMCID: PMC5080427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
OBJECTIVES Glial cell line-derived neurotrophic factor (GDNF) can effectively promote axonal regeneration, limit axonal retraction, and produce a statistically significant improvement in motor recovery after spinal cord injury (SCI). However, the role in primate animals with SCI is not fully cognized. MATERIALS AND METHODS 18 healthy juvenile rhesuses were divided randomly into six groups, observed during the periods of 24 hr, 7 days, 14 days, 1 month, 2 months, and 3 months after T11 hemisecting. The GDNF localization, changes in the injured region, and the remote associate cortex were detected by immunohistochemical staining. RESULTS Immunohistochemical staining showed that GDNF was located in the cytoplasm and the neurite of the neurons. Following SCI, the number of GDNF positive neurons in the ventral horn and the caudal part near the lesion area were apparently reduced at detected time points (P<0.05). Moreover, the number in the rostral part of the ventral horn in 7 day, 14 day, and 1 month groups were fewer than those in the caudal part. Importantly, in the contralateral cortex motor area, the positive neurons decreased sharply after hemi-SCI, while gradually increased and went back to normal in 3 months after hemi-SCI. CONCLUSION To sum up, GDNF disruption in neurons occurred after SCI especially in cortex motor area. Intrinsic GDNF in the spinal cord, plays an essential role in neuroplasticity. Thereafter extrinsic GDNF supplementing may be a useful strategy to promote recovery after SCI.
Collapse
Affiliation(s)
- De-Lu Qiu
- Laboratory of Anesthesia and Critical Care Medicine, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Ting-Hua Wang
- Laboratory of Anesthesia and Critical Care Medicine, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China,Corresponding author: Ting-Hua Wang. Laboratory of Anesthesia and Critical Care Medicine, Translational Neuroscience Center, West China Hospital, Sichuan university, Chengdu, Sichuan, 610041, China. Tel/Fax: +86-2885501036;
| |
Collapse
|
49
|
Lin XY, Lai BQ, Zeng X, Che MT, Ling EA, Wu W, Zeng YS. Cell Transplantation and Neuroengineering Approach for Spinal Cord Injury Treatment: A Summary of Current Laboratory Findings and Review of Literature. Cell Transplant 2016; 25:1425-38. [DOI: 10.3727/096368916x690836] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Spinal cord injury (SCI) can cause severe traumatic injury to the central nervous system (CNS). Current therapeutic effects achieved for SCI in clinical medicine show that there is still a long way to go to reach the desired goal of full or significant functional recovery. In basic medical research, however, cell transplantation, gene therapy, application of cytokines, and biomaterial scaffolds have been widely used and investigated as treatments for SCI. All of these strategies when used separately would help rebuild, to some extent, the neural circuits in the lesion area of the spinal cord. In light of this, it is generally accepted that a combined treatment may be a more effective strategy. This review focuses primarily on our recent series of work on transplantation of Schwann cells and adult stem cells, and transplantation of stem cell-derived neural network scaffolds with functional synapses. Arising from this, an artificial neural network (an exogenous neuronal relay) has been designed and fabricated by us—a biomaterial scaffold implanted with Schwann cells modified by the neurotrophin-3 (NT-3) gene and adult stem cells modified with the TrkC (receptor of NT-3) gene. More importantly, experimental evidence suggests that the novel artificial network can integrate with the host tissue and serve as an exogenous neuronal relay for signal transfer and functional improvement of SCI.
Collapse
Affiliation(s)
- Xin-Yi Lin
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat-sen University), Ministry of Education, Guangzhou, Guangdong, China
| | - Bi-Qin Lai
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat-sen University), Ministry of Education, Guangzhou, Guangdong, China
| | - Xiang Zeng
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat-sen University), Ministry of Education, Guangzhou, Guangdong, China
| | - Ming-Tian Che
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat-sen University), Ministry of Education, Guangzhou, Guangdong, China
| | - Eng-Ang Ling
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Wutian Wu
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
- State Key Laboratory of Brain and Cognitive Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
- Jinan University–Hong Kong University Joint Laboratory, GHM Institute of CNS Regeneration, Jinan University, Guangzhou, China
| | - Yuan-Shan Zeng
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat-sen University), Ministry of Education, Guangzhou, Guangdong, China
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Institute of Spinal Cord Injury, Sun Yat-sen University, Guangzhou, China
- Co-innovation Center of Neuroregeneration, Nantong, Jiangsu, China
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
50
|
Abstract
STUDY DESIGN Laboratory/animal-based proof of principle study. OBJECTIVE To validate the accuracy of a magnetic resonance imaging (MRI)-guided stereotactic system for intraspinal electrode targeting and demonstrate the feasibility of such a system for controlling implantation of intraspinal electrodes. SUMMARY OF BACKGROUND DATA Intraspinal microstimulation (ISMS) is an emerging preclinical therapy, which has shown promise for the restoration of motor function following spinal cord injury. However, targeting inaccuracy associated with existing electrode implantation techniques remains a major barrier preventing clinical translation of ISMS. METHODS System accuracy was evaluated using a test phantom comprised of nine target locations. Targeting accuracy was determined by calculating the root mean square error between MRI-generated coordinates and actual frame coordinates required to reach the target positions. System performance was further validated in an anesthetized pig model by performing MRI-guided intraspinal electrode implantation and stimulation followed by computed tomography of electrode location. Finally, system compatibility with a commercially available microelectrode array was demonstrated by implanting the array and applying a selection of stimulation amplitudes that evoked hind limb responses. RESULTS The root mean square error between actual frame coordinates and software coordinates, both acquired using the test phantom, was 1.09 ± 0.20 mm. Postoperative computed tomography in the anesthetized pig confirmed spatially accurate electrode placement relative to preoperative MRI. Additionally, MRI-guided delivery of a microwire electrode followed by ISMS evoked repeatable electromyography responses in the biceps femoris muscle. Finally, delivery of a microelectrode array produced repeatable and graded hind limb evoked movements. CONCLUSION We present a novel frame-based stereotactic system for targeting and delivery of intraspinal instrumentation. This system utilizes MRI guidance to account for variations in anatomy between subjects, thereby improving upon existing ISMS electrode implantation techniques. LEVEL OF EVIDENCE N/A.
Collapse
|