1
|
Zhang Y, Chen X, Wang X, Xu Y, Li J, Wu Y, Wang Z, Zhang S, Hu J, Qi Q. Hesperetin ameliorates spinal cord injury in rats through suppressing apoptosis, oxidative stress and inflammatory response. Eur J Pharmacol 2024; 971:176541. [PMID: 38556120 DOI: 10.1016/j.ejphar.2024.176541] [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: 01/30/2024] [Revised: 03/22/2024] [Accepted: 03/26/2024] [Indexed: 04/02/2024]
Abstract
Spinal cord injury (SCI), a fatal condition, is characterized by progressive tissue degradation and extreme functional deficits with limited treatment options. Hesperetin, a natural flavonoid with potent antioxidant, antiapoptotic and anti-inflammatory properties, has yet to be systematically investigated for its therapeutic effects on neurological damage in rat models of SCI. In this study, rats were given oral hesperetin once daily for 28 days, and their locomotion and histopathological changes were assessed. The findings demonstrated that hesperetin alleviates neurological damage caused by SCI. The observed behavioral improvement could be due to an increase in the survival rate of neurons and oligodendrocytes. This improvement further boosted the ability to repair tissue and form myelin after SCI, ultimately resulting in better neurological outcomes. Furthermore, the present study revealed that hesperetin possesses potent antioxidant capabilities in the context of SCI, reducing the levels of harmful oxygen free radicals and increasing the activity of antioxidant enzymes. Additionally, hesperetin markedly inhibited injury-induced apoptosis, as assessed by caspase-3 immunofluorescence staining and the expression level of caspase-3, indicating the ability of hesperetin to prevent cell death after SCI. Finally, after SCI, hesperetin treatment effectively reduced the expression of inflammatory factors, including IL-1β, TNFα, and NF-kB, demonstrating the anti-inflammatory effect of hesperetin. Together, our results suggest that hesperetin should be considered a valuable therapeutic aid following SCI, as its positive effects on the nervous system, including antioxidant, anti-inflammatory and antiapoptotic effects, may be crucial mechanisms through which hesperetin exerts neuroprotective effects against SCI.
Collapse
Affiliation(s)
- Yuxin Zhang
- Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical University, Bengbu, China; School of Laboratory Medicine, Bengbu Medical University, Bengbu, China
| | - Xiaojie Chen
- Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical University, Bengbu, China; School of Basic Medicine, Bengbu Medical University, Bengbu, China
| | - Xiaoxuan Wang
- School of Laboratory Medicine, Bengbu Medical University, Bengbu, China; Clinical Laboratory, Bengbu Municipal Second People Hospital, Bengbu, China
| | - Yibo Xu
- Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical University, Bengbu, China; School of Basic Medicine, Bengbu Medical University, Bengbu, China
| | - Jiaxin Li
- Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical University, Bengbu, China; School of Basic Medicine, Bengbu Medical University, Bengbu, China
| | - Yimin Wu
- Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical University, Bengbu, China; School of Basic Medicine, Bengbu Medical University, Bengbu, China
| | - Ziyao Wang
- Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical University, Bengbu, China; School of Basic Medicine, Bengbu Medical University, Bengbu, China
| | - Suhui Zhang
- Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical University, Bengbu, China; School of Basic Medicine, Bengbu Medical University, Bengbu, China
| | - Jianguo Hu
- Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical University, Bengbu, China; Department of Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical University, Bengbu, China.
| | - Qi Qi
- Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical University, Bengbu, China; School of Basic Medicine, Bengbu Medical University, Bengbu, China.
| |
Collapse
|
2
|
Pei Q, Zhao Q, Lang C, Feng S, Meng J, Tan G, Cui W, Zhang C, Luo X, Xu L, Chen J. Alleviating Severe Cytoskeletal Destruction of Spinal Motor Neurons: Another Effect of Docosahexaenoic Acid in Spinal Cord Injury. ACS Chem Neurosci 2024; 15:1456-1468. [PMID: 38472087 DOI: 10.1021/acschemneuro.3c00746] [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] [Indexed: 03/14/2024] Open
Abstract
Spinal cord injury (SCI) treatment remains a major challenge. Spinal motor neurons (MNs) are seriously injured in the early stage after SCI, but this has not received sufficient attention. Oxidative stress is known to play a crucial role in SCI pathology. Our studies demonstrated that oxidative stress can cause severe damage to the cytoskeleton of spinal MNs. Docosahexaenoic acid (DHA) has been shown to have beneficial effects on SCI, but the mechanism remains unclear, and no study has investigated the effect of DHA on oxidative stress-induced spinal MN injury. Here, we investigated the effect of DHA on spinal MN injury through in vivo and in vitro experiments, focusing on the cytoskeleton. We found that DHA not only promoted spinal MN survival but, more importantly, alleviated the severe cytoskeletal destruction of these neurons induced by oxidative stress in vitro and in mice with SCI in vivo. In addition, the mechanisms involved were investigated and elucidated. These results not only suggested a beneficial role of DHA in spinal MN cytoskeletal destruction caused by oxidative stress and SCI but also indicated the important role of the spinal MN cytoskeleton in the recovery of motor function after SCI. Our study provides new insights for the formulation of SCI treatment.
Collapse
Affiliation(s)
- Qinqin Pei
- Central laboratory, Chongqing University Three Gorges Hospital, Chongqing 404000, China
| | - Qiurong Zhao
- Central laboratory, Chongqing University Three Gorges Hospital, Chongqing 404000, China
| | - Chunhui Lang
- Chongqing Municipality Clinical Research Center for Geriatric Diseases, Chongqing University Three Gorges Hospital, Chongqing 404000, China
| | - Shilong Feng
- Department of Orthopedics, Chongqing University Three Gorges Hospital, Chongqing 404000, China
| | - Juanjuan Meng
- Central laboratory, Chongqing University Three Gorges Hospital, Chongqing 404000, China
| | - Guangjiao Tan
- Central laboratory, Chongqing University Three Gorges Hospital, Chongqing 404000, China
| | - Wei Cui
- Central laboratory, Chongqing University Three Gorges Hospital, Chongqing 404000, China
| | - Cheng Zhang
- Chongqing Municipality Clinical Research Center for Endocrinology and Metabolic Diseases, Chongqing University Three Gorges Hospital, Chongqing 404000, China
| | - Xiaohe Luo
- Central laboratory, Chongqing University Three Gorges Hospital, Chongqing 404000, China
| | - Lixin Xu
- Department of Orthopedics, Chongqing University Three Gorges Hospital, Chongqing 404000, China
- Chongqing Municipality Clinical Research Center for Geriatric Diseases, Chongqing University Three Gorges Hospital, Chongqing 404000, China
| | - Jian Chen
- Department of Orthopedics, Chongqing University Three Gorges Hospital, Chongqing 404000, China
- Chongqing Municipality Clinical Research Center for Geriatric Diseases, Chongqing University Three Gorges Hospital, Chongqing 404000, China
- Chongqing Municipality Clinical Research Center for Endocrinology and Metabolic Diseases, Chongqing University Three Gorges Hospital, Chongqing 404000, China
| |
Collapse
|
3
|
da Silva BR, de Melo Reis RA, Ribeiro-Resende VT. A Comparative Investigation of Axon-Blood Vessel Growth Interaction in the Regenerating Sciatic and Optic Nerves in Adult Mice. Mol Neurobiol 2024; 61:2215-2227. [PMID: 37864766 DOI: 10.1007/s12035-023-03705-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 10/10/2023] [Indexed: 10/23/2023]
Abstract
The vascular and the nervous systems share similarities in addition to their complex role in providing oxygen and nutrients to all cells. Both are highly branched networks that frequently grow close to one another during development. Vascular patterning and neural wiring share families of guidance cues and receptors. Most recently, this relationship has been investigated in terms of peripheral nervous system (PNS) regeneration, where nerves and blood vessels often run in parallel so endothelial cells guide the formation of the Büngner bands which support axonal regeneration. Here, we characterized the vascular response in regenerative models of the central and peripheral nervous system. After sciatic nerve crush, followed by axon regeneration, there was a significant increase in the blood vessel density 7 days after injury. In addition, the optic nerve crush model was used to evaluate intrinsic regenerative potential activated with a combined treatment that stimulated retinal ganglion cells (RGCs) regrowth. We observed that a 2-fold change in the total number of blood vessels occurred 7 days after optic nerve crush compared to the uncrushed nerve. The difference increased up to a 2.7-fold change 2 weeks after the crush. Interestingly, we did not observe differences in the total number of blood vessels 2 weeks after crush, compared to animals that had received combined treatment for regeneration and controls. Therefore, the vascular characterization showed that the increase in vascular density was not related to the efficiency of both peripheral and central axonal regeneration.
Collapse
Affiliation(s)
- Barbara Rangel da Silva
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, 21941-902, Brazil
| | - Ricardo A de Melo Reis
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, 21941-902, Brazil
| | - Victor Túlio Ribeiro-Resende
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, 21941-902, Brazil.
- Núcleo Multidisciplinar de Pesquisa em Biologia (Numpex-Bio), Campus de Duque de Caxias Geraldo Cidade, Universidade Federal do Rio de Janeiro, Duque de Caxias, Brazil.
| |
Collapse
|
4
|
Bao S, Lei Y. Motor unit activity and synaptic inputs to motoneurons in the caudal part of the injured spinal cord. J Neurophysiol 2024; 131:187-197. [PMID: 38117916 DOI: 10.1152/jn.00178.2023] [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: 05/02/2023] [Revised: 12/07/2023] [Accepted: 12/20/2023] [Indexed: 12/22/2023] Open
Abstract
Spinal cord injury (SCI) disrupts neuronal function below the lesion epicenter, causing disuse muscle atrophy. We investigated motor unit (MU) activity and synaptic inputs to motoneurons in the caudal region of the injured spinal cord. Participants with C4-C7 cervical injuries were studied. The extensor digitorum communis (EDC) muscle, which is mainly innervated by C8, was assessed for disuse muscle atrophy. Using advanced electromyography and signal-processing techniques, we examined the concurrent activation of a substantial population of MUs during force-tracking tasks. We found that in participants with SCI (n = 9), both MU discharge rates and the amplitudes of MU action potentials were significantly lower than in controls (n = 9). After SCI, MUs were recruited in a limited force range as the strength of muscle contractions increased, implying a disruption in the orderly MU recruitment pattern. Coherence analysis revealed reduced synaptic inputs to motoneurons in the delta band (0.5-5 Hz) for participants with SCI, suggesting diminished common synaptic inputs to the EDC muscle. In addition, participants with SCI exhibited greater muscle force variability. Using principal component analysis on low-frequency MU discharge rates, we found that the first common component (FCC) captured the most discharge variability in participants with SCI. The coefficients of variation (CV) of the FCC correlated with force signal CVs, suggesting force variability mainly results from common synaptic inputs to the EDC muscle after SCI. These results advance our understanding of the neurophysiology of disuse muscle atrophy in human SCI, paving the way for therapeutic interventions to restore muscle function.NEW & NOTEWORTHY This study analyzed motor unit (MU) function below the lesion epicenter in patients with spinal cord injury (SCI). We found reduced MU discharge rates and action potential amplitudes in participants with SCI compared with controls. The strength of common synaptic inputs to motoneurons was reduced in patients with SCI, with increased force variability primarily due to low-frequency oscillations of common inputs. This study enhances understanding of neurophysiological and behavioral changes in disuse muscle atrophy post-SCI.
Collapse
Affiliation(s)
- Shancheng Bao
- Department of Kinesiology & Sport Management, Texas A&M University, College Station, Texas, United States
| | - Yuming Lei
- Department of Kinesiology & Sport Management, Texas A&M University, College Station, Texas, United States
| |
Collapse
|
5
|
Chen LM, Wang F, Mishra A, Yang PF, Sengupta A, Reed JL, Gore JC. Longitudinal multiparametric MRI of traumatic spinal cord injury in animal models. Magn Reson Imaging 2023; 102:184-200. [PMID: 37343904 PMCID: PMC10528214 DOI: 10.1016/j.mri.2023.06.007] [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: 03/17/2022] [Revised: 06/14/2023] [Accepted: 06/17/2023] [Indexed: 06/23/2023]
Abstract
Multi-parametric MRI (mpMRI) technology enables non-invasive and quantitative assessments of the structural, molecular, and functional characteristics of various neurological diseases. Despite the recognized importance of studying spinal cord pathology, mpMRI applications in spinal cord research have been somewhat limited, partly due to technical challenges associated with spine imaging. However, advances in imaging techniques and improved image quality now allow longitudinal investigations of a comprehensive range of spinal cord pathological features by exploiting different endogenous MRI contrasts. This review summarizes the use of mpMRI techniques including blood oxygenation level-dependent (BOLD) functional MRI (fMRI), diffusion tensor imaging (DTI), quantitative magnetization transfer (qMT), and chemical exchange saturation transfer (CEST) MRI in monitoring different aspects of spinal cord pathology. These aspects include cyst formation and axonal disruption, demyelination and remyelination, changes in the excitability of spinal grey matter and the integrity of intrinsic functional circuits, and non-specific molecular changes associated with secondary injury and neuroinflammation. These approaches are illustrated with reference to a nonhuman primate (NHP) model of traumatic cervical spinal cord injuries (SCI). We highlight the benefits of using NHP SCI models to guide future studies of human spinal cord pathology, and demonstrate how mpMRI can capture distinctive features of spinal cord pathology that were previously inaccessible. Furthermore, the development of mechanism-based MRI biomarkers from mpMRI studies can provide clinically useful imaging indices for understanding the mechanisms by which injured spinal cords progress and repair. These biomarkers can assist in the diagnosis, prognosis, and evaluation of therapies for SCI patients, potentially leading to improved outcomes.
Collapse
Affiliation(s)
- Li Min Chen
- Vanderbilt University Institute of Imaging Science, Vanderbilt University, Nashville, TN, USA; Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA.
| | - Feng Wang
- Vanderbilt University Institute of Imaging Science, Vanderbilt University, Nashville, TN, USA; Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Arabinda Mishra
- Vanderbilt University Institute of Imaging Science, Vanderbilt University, Nashville, TN, USA; Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Pai-Feng Yang
- Vanderbilt University Institute of Imaging Science, Vanderbilt University, Nashville, TN, USA; Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Anirban Sengupta
- Vanderbilt University Institute of Imaging Science, Vanderbilt University, Nashville, TN, USA; Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jamie L Reed
- Vanderbilt University Institute of Imaging Science, Vanderbilt University, Nashville, TN, USA; Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - John C Gore
- Vanderbilt University Institute of Imaging Science, Vanderbilt University, Nashville, TN, USA; Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
6
|
Pukos N, Marion CM, Arnold WD, Noble BT, Popovich PG, McTigue DM. Chronic demyelination and myelin repair after spinal cord injury in mice: A potential link for glutamatergic axon activity. Glia 2023; 71:2096-2116. [PMID: 37208933 PMCID: PMC10330449 DOI: 10.1002/glia.24382] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 04/08/2023] [Accepted: 04/20/2023] [Indexed: 05/21/2023]
Abstract
Our prior work examining endogenous repair after spinal cord injury (SCI) in mice revealed that large numbers of new oligodendrocytes (OLs) are generated in the injured spinal cord, with peak oligodendrogenesis between 4 and 7 weeks post-injury (wpi). We also detected new myelin formation over 2 months post-injury (mpi). Our current work significantly extends these results, including quantification of new myelin through 6 mpi and concomitant examination of indices of demyelination. We also examined electrophysiological changes during peak oligogenesis and a potential mechanism driving OL progenitor cell (OPC) contact with axons. Results reveal peak in remyelination occurs during the 3rd mpi, and that myelin generation continues for at least 6 mpi. Further, motor evoked potentials significantly increased during peak remyelination, suggesting enhanced axon potential conduction. Interestingly, two indices of demyelination, nodal protein spreading and Nav1.2 upregulation, were also present chronically after SCI. Nav1.2 was expressed through 10 wpi and nodal protein disorganization was detectable throughout 6 mpi suggesting chronic demyelination, which was confirmed with EM. Thus, demyelination may continue chronically, which could trigger the long-term remyelination response. To examine a potential mechanism that may initiate post-injury myelination, we show that OPC processes contact glutamatergic axons in the injured spinal cord in an activity-dependent manner. Notably, these OPC/axon contacts were increased 2-fold when axons were activated chemogenetically, revealing a potential therapeutic target to enhance post-SCI myelin repair. Collectively, results show the surprisingly dynamic nature of the injured spinal cord over time and that the tissue may be amenable to treatments targeting chronic demyelination.
Collapse
Affiliation(s)
- Nicole Pukos
- Neuroscience Graduate Program, Ohio State University, Columbus, Ohio, USA
- Belford Center for Spinal Cord Injury, Ohio State University, Columbus, Ohio, USA
| | - Christina M Marion
- Belford Center for Spinal Cord Injury, Ohio State University, Columbus, Ohio, USA
- Department of Neuroscience, College of Medicine, Ohio State University, Columbus, Ohio, USA
| | - W David Arnold
- Department of Neuroscience, College of Medicine, Ohio State University, Columbus, Ohio, USA
- Division of Neuromuscular Disorders, Department of Neurology, Wexner Medical Center, Ohio State University, Columbus, Ohio, USA
- Department of Physical Medicine and Rehabilitation, Wexner Medical Center, Ohio State University, Columbus, Ohio, USA
- Department of Physiology and Cell Biology, College of Medicine, Ohio State University, Columbus, Ohio, USA
| | - Benjamin T Noble
- Neuroscience Graduate Program, Ohio State University, Columbus, Ohio, USA
- Belford Center for Spinal Cord Injury, Ohio State University, Columbus, Ohio, USA
| | - Phillip G Popovich
- Belford Center for Spinal Cord Injury, Ohio State University, Columbus, Ohio, USA
- Department of Neuroscience, College of Medicine, Ohio State University, Columbus, Ohio, USA
| | - Dana M McTigue
- Belford Center for Spinal Cord Injury, Ohio State University, Columbus, Ohio, USA
- Department of Neuroscience, College of Medicine, Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
7
|
Gupta S, Dutta S, Hui SP. Regenerative Potential of Injured Spinal Cord in the Light of Epigenetic Regulation and Modulation. Cells 2023; 12:1694. [PMID: 37443728 PMCID: PMC10341208 DOI: 10.3390/cells12131694] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/13/2023] [Accepted: 06/17/2023] [Indexed: 07/15/2023] Open
Abstract
A spinal cord injury is a form of physical harm imposed on the spinal cord that causes disability and, in many cases, leads to permanent mammalian paralysis, which causes a disastrous global issue. Because of its non-regenerative aspect, restoring the spinal cord's role remains one of the most daunting tasks. By comparison, the remarkable regenerative ability of some regeneration-competent species, such as some Urodeles (Axolotl), Xenopus, and some teleost fishes, enables maximum functional recovery, even after complete spinal cord transection. During the last two decades of intensive research, significant progress has been made in understanding both regenerative cells' origins and the molecular signaling mechanisms underlying the regeneration and reconstruction of damaged spinal cords in regenerating organisms and mammals, respectively. Epigenetic control has gradually moved into the center stage of this research field, which has been helped by comprehensive work demonstrating that DNA methylation, histone modifications, and microRNAs are important for the regeneration of the spinal cord. In this review, we concentrate primarily on providing a comparison of the epigenetic mechanisms in spinal cord injuries between non-regenerating and regenerating species. In addition, we further discuss the epigenetic mediators that underlie the development of a regeneration-permissive environment following injury in regeneration-competent animals and how such mediators may be implicated in optimizing treatment outcomes for spinal cord injurie in higher-order mammals. Finally, we briefly discuss the role of extracellular vesicles (EVs) in the context of spinal cord injury and their potential as targets for therapeutic intervention.
Collapse
Affiliation(s)
- Samudra Gupta
- S.N. Pradhan Centre for Neurosciences, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, India;
| | - Suman Dutta
- Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DU, UK;
| | - Subhra Prakash Hui
- S.N. Pradhan Centre for Neurosciences, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, India;
| |
Collapse
|
8
|
Ghezzi L, Bollman B, De Feo L, Piccio L, Trapp BD, Schmidt RE, Cross AH. Schwann Cell Remyelination in the Multiple Sclerosis Central Nervous System. J Transl Med 2023; 103:100128. [PMID: 36889543 PMCID: PMC10330052 DOI: 10.1016/j.labinv.2023.100128] [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: 10/10/2022] [Revised: 02/22/2023] [Accepted: 02/22/2023] [Indexed: 03/08/2023] Open
Abstract
Multiple sclerosis (MS) is a central nervous system (CNS) demyelinating disease. Failure to remyelinate successfully is common in MS lesions, often with consequent neuronal/axonal damage. CNS myelin is normally produced by oligodendroglial cells. Remyelination by Schwann cells (SchC) has been reported in spinal cord demyelination, in which SchCs are in close proximity to CNS myelin. We identified an MS cerebral lesion that was remyelinated by SchCs. This prompted us to query the extent of SchC remyelination in the brain and spinal cords of additional autopsied MS specimens. CNS tissues were obtained from the autopsies of 14 MS cases. Remyelinated lesions were identified by Luxol fast blue-periodic-acid Schiff and solochrome cyanine staining. Deparaffinized sections containing remyelinated lesions were stained with anti-glial fibrillary acid protein to identify reactive astrocytes. Glycoprotein P zero (P0) is a protein exclusive to peripheral but not CNS myelin. Areas of SchC remyelination were identified by staining with anti-P0. Myelinated regions in the index case cerebral lesion were confirmed to be of SchC origin using anti-P0 staining. Subsequently, 64 MS lesions from 14 autopsied MS cases were examined, and 23 lesions in 6 cases showed remyelination by SchCs. Lesions from the cerebrum, brainstem, and spinal cord were examined in each case. When present, SchC remyelination was most commonly located adjacent to the venules and associated with a lower surrounding density of glial fibrillary acid protein+ reactive astrocytes than areas of only oligodendroglial cell remyelination. The difference was significant only for spinal cord and brainstem lesions but not for lesions located in the brain. In conclusion, we demonstrated SchC remyelination in the cerebrum, brainstem, and spinal cord of 6 autopsied MS cases. To our knowledge, this is the first report of supratentorial SchC remyelination in MS.
Collapse
Affiliation(s)
- Laura Ghezzi
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri.
| | - Bryan Bollman
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri
| | - Luca De Feo
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri
| | - Laura Piccio
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri; Brain and Mind Centre and Charles Perkins Centre, School of Medical Sciences, Neuroscience, University of Sydney, Sydney, New South Wales, Australia
| | - Bruce D Trapp
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Robert E Schmidt
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri
| | - Anne H Cross
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
9
|
Vangansewinkel T, Lemmens S, Tiane A, Geurts N, Dooley D, Vanmierlo T, Pejler G, Hendrix S. Therapeutic administration of mouse mast cell protease 6 improves functional recovery after traumatic spinal cord injury in mice by promoting remyelination and reducing glial scar formation. FASEB J 2023; 37:e22939. [PMID: 37130013 DOI: 10.1096/fj.202201942rr] [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: 11/26/2022] [Revised: 04/06/2023] [Accepted: 04/14/2023] [Indexed: 05/03/2023]
Abstract
Traumatic spinal cord injury (SCI) most often leads to permanent paralysis due to the inability of axons to regenerate in the adult mammalian central nervous system (CNS). In the past, we have shown that mast cells (MCs) improve the functional outcome after SCI by suppressing scar tissue formation at the lesion site via mouse mast cell protease 6 (mMCP6). In this study, we investigated whether recombinant mMCP6 can be used therapeutically to improve the functional outcome after SCI. Therefore, we applied mMCP6 locally via an intrathecal catheter in the subacute phase after a spinal cord hemisection injury in mice. Our findings showed that hind limb motor function was significantly improved in mice that received recombinant mMCP6 compared with the vehicle-treated group. In contrast to our previous findings in mMCP6 knockout mice, the lesion size and expression levels of the scar components fibronectin, laminin, and axon-growth-inhibitory chondroitin sulfate proteoglycans were not affected by the treatment with recombinant mMCP6. Surprisingly, no difference in infiltration of CD4+ T cells and reactivity of Iba-1+ microglia/macrophages at the lesion site was observed between the mMCP6-treated mice and control mice. Additionally, local protein levels of the pro- and anti-inflammatory mediators IL-1β, IL-2, IL-4, IL-6, IL-10, TNF-α, IFNγ, and MCP-1 were comparable between the two treatment groups, indicating that locally applied mMCP6 did not affect inflammatory processes after injury. However, the increase in locomotor performance in mMCP6-treated mice was accompanied by reduced demyelination and astrogliosis in the perilesional area after SCI. Consistently, we found that TNF-α/IL-1β-astrocyte activation was decreased and that oligodendrocyte precursor cell (OPC) differentiation was increased after recombinant mMCP6 treatment in vitro. Mechanistically, this suggests effects of mMCP6 on reducing astrogliosis and improving (re)myelination in the spinal cord after injury. In conclusion, these data show for the first time that recombinant mMCP6 is therapeutically active in enhancing recovery after SCI.
Collapse
Affiliation(s)
- Tim Vangansewinkel
- Cardio and Organ Systems, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
- VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, Leuven, Belgium
| | - Stefanie Lemmens
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Assia Tiane
- Department of Neuroscience, Faculty of Medicine and Life Sciences, Biomedical Research Institute, Hasselt University, Hasselt, Belgium
- Department Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
- University MS Center (UMSC) Hasselt-Pelt, Hasselt, Belgium
| | - Nathalie Geurts
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Dearbhaile Dooley
- School of Medicine, Health Sciences Centre, University College Dublin, Belfield, Ireland
- UCD Conway Institute of Biomolecular & Biomedical Research University College Dublin, Belfield, Ireland
| | - Tim Vanmierlo
- Department of Neuroscience, Faculty of Medicine and Life Sciences, Biomedical Research Institute, Hasselt University, Hasselt, Belgium
- Department Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
- University MS Center (UMSC) Hasselt-Pelt, Hasselt, Belgium
| | - Gunnar Pejler
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Sven Hendrix
- Institute for Translational Medicine, Medical School Hamburg, Hamburg, Germany
| |
Collapse
|
10
|
Prasse T, Khaing ZZ, Cates LN, Dewees DM, Hyde JE, Bredow J, Hofstetter CP. A decrease in the neuroprotective effects of acute spinal cord decompression according to injury severity: introducing the concept of a ceiling effect. J Neurosurg Spine 2023; 38:299-306. [PMID: 36401546 DOI: 10.3171/2022.6.spine22383] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 06/28/2022] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Acute traumatic spinal cord injury (tSCI) is followed by a prolonged period of secondary neuroglial cell death. Neuroprotective interventions, such as surgical spinal cord decompression, aim to mitigate secondary injury. In this study, the authors explore whether the effect size of posttraumatic neuroprotective spinal cord decompression varies with injury severity. METHODS Seventy-one adult female Long Evans rats were subjected to a thoracic tSCI using a third-generation spinal contusion device. Moderate and severe tSCI were defined by recorded impact force delivered to the spinal cord. Immediately after injury (< 15 minutes), treatment cohorts underwent either a decompressive durotomy or myelotomy. Functional recovery was documented using the Basso, Beattie, and Bresnahan locomotor scale, and tissue sparing was documented using histological analysis. RESULTS Moderate and severe injuries were separated at a cutoff point of 231.8 kdyn peak impact force based on locomotor recovery at 8 weeks after injury. Durotomy improved hindlimb locomotor recovery 8 weeks after moderate trauma (p < 0.01), but not after severe trauma (p > 0.05). Myelotomy led to increased tissue sparing (p < 0.0001) and a significantly higher number of spared motor neurons (p < 0.05) in moderate trauma, but no such effect was noted in severely injured rats (p > 0.05). Within the moderate injury group, myelotomy also resulted in significantly more spared tissue when compared with durotomy-only animals (p < 0.01). CONCLUSIONS These results suggest that the neuroprotective effects of surgical spinal cord decompression decrease with increasing injury severity in a rodent tSCI model.
Collapse
Affiliation(s)
- Tobias Prasse
- 1Department of Neurological Surgery, University of Washington, Seattle, Washington
- 2Faculty of Medicine and University Hospital Cologne, Department of Orthopedics and Trauma Surgery, University of Cologne; and
| | - Zin Z Khaing
- 1Department of Neurological Surgery, University of Washington, Seattle, Washington
| | - Lindsay N Cates
- 1Department of Neurological Surgery, University of Washington, Seattle, Washington
| | - Dane M Dewees
- 1Department of Neurological Surgery, University of Washington, Seattle, Washington
| | - Jeffrey E Hyde
- 1Department of Neurological Surgery, University of Washington, Seattle, Washington
| | - Jan Bredow
- 3Department of Orthopedics and Trauma Surgery, Krankenhaus Porz am Rhein, University of Cologne, Germany
| | | |
Collapse
|
11
|
Furumiya T, Itokazu T, Nakanishi T, Yamashita T. CXCR4 signaling regulates repair Schwann cell infiltration into the spinal cord after spinal cord injury in mice. Neurosci Res 2022; 191:38-47. [PMID: 36592826 DOI: 10.1016/j.neures.2022.12.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 12/12/2022] [Accepted: 12/25/2022] [Indexed: 12/31/2022]
Abstract
Schwann cells are glial cells that myelinate neuronal axons in the peripheral nervous system (PNS). When the PNS is damaged, Schwann cells de-differentiate into p75-positive "repair Schwann cells," which contribute to neural circuit regeneration. Interestingly, Schwann cells in the dorsal roots are known to be reprogrammed to repair Schwann cells even after spinal cord injury (SCI) and then migrate into the injured spinal cord. However, the molecular mechanism underlying the migration of repair Schwann cells remains unknown. Since a recent in vitro study revealed the importance of CXCR4 signaling in Schwann cell migration, we investigated whether CXCR4 signaling is involved in the PNS-to-central nervous system (CNS) migration of repair Schwann cells after SCI. We revealed that repair Schwann cells express CXCR4, and its ligand CXCL12 is upregulated in the injured spinal cord. We also found that the pharmacological inhibition of CXCR4 signaling decreased the infiltration of repair Schwann cells. Moreover, CXCR4 agonist administration effectively increased the infiltration of repair Schwann cells along with improved motor function. These findings strongly suggest the involvement of CXCR4 signaling in the PNS-to-CNS migration of repair Schwann cells after SCI.
Collapse
Affiliation(s)
- Takeru Furumiya
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Takahide Itokazu
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan; Department of Neuro-Medical Science, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan.
| | - Toru Nakanishi
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan; Department of Neuro-Medical Science, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Toshihide Yamashita
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan; Department of Neuro-Medical Science, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan; WPI Immunology Frontier Research Center, Osaka, Japan; Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan.
| |
Collapse
|
12
|
Pathophysiology, Classification and Comorbidities after Traumatic Spinal Cord Injury. J Pers Med 2022; 12:jpm12071126. [PMID: 35887623 PMCID: PMC9323191 DOI: 10.3390/jpm12071126] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 06/26/2022] [Accepted: 06/27/2022] [Indexed: 12/25/2022] Open
Abstract
The spinal cord is a conduit within the central nervous system (CNS) that provides ongoing communication between the brain and the rest of the body, conveying complex sensory and motor information necessary for safety, movement, reflexes, and optimization of autonomic function. After a spinal cord injury (SCI), supraspinal influences on the spinal segmental control system and autonomic nervous system (ANS) are disrupted, leading to spastic paralysis, pain and dysesthesia, sympathetic blunting and parasympathetic dominance resulting in cardiac dysrhythmias, systemic hypotension, bronchoconstriction, copious respiratory secretions and uncontrolled bowel, bladder, and sexual dysfunction. This article outlines the pathophysiology of traumatic SCI, current and emerging methods of classification, and its influence on sensory/motor function, and introduces the probable comorbidities associated with SCI that will be discussed in more detail in the accompanying manuscripts of this special issue.
Collapse
|
13
|
Wang R, Zhou R, Chen Z, Gao S, Zhou F. The Glial Cells Respond to Spinal Cord Injury. Front Neurol 2022; 13:844497. [PMID: 35599739 PMCID: PMC9120539 DOI: 10.3389/fneur.2022.844497] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 03/08/2022] [Indexed: 12/24/2022] Open
Abstract
It is been over 100 years since glial cells were discovered by Virchow. Since then, a great deal of research was carried out to specify these further roles and properties of glial cells in central nervous system (CNS). As it is well-known that glial cells, such as astrocytes, microglia, oligodendrocytes (OLs), and oligodendrocyte progenitor cells (OPCs) play an important role in supporting and enabling the effective nervous system function in CNS. After spinal cord injury (SCI), these glial cells play different roles in SCI and repair. In this review, we will discuss in detail about the role of glial cells in the healthy CNS and how they respond to SCI.
Collapse
|
14
|
Progression in translational research on spinal cord injury based on microenvironment imbalance. Bone Res 2022; 10:35. [PMID: 35396505 PMCID: PMC8993811 DOI: 10.1038/s41413-022-00199-9] [Citation(s) in RCA: 65] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 11/14/2021] [Accepted: 12/22/2021] [Indexed: 02/07/2023] Open
Abstract
Spinal cord injury (SCI) leads to loss of motor and sensory function below the injury level and imposes a considerable burden on patients, families, and society. Repair of the injured spinal cord has been recognized as a global medical challenge for many years. Significant progress has been made in research on the pathological mechanism of spinal cord injury. In particular, with the development of gene regulation, cell sequencing, and cell tracing technologies, in-depth explorations of the SCI microenvironment have become more feasible. However, translational studies related to repair of the injured spinal cord have not yielded significant results. This review summarizes the latest research progress on two aspects of SCI pathology: intraneuronal microenvironment imbalance and regenerative microenvironment imbalance. We also review repair strategies for the injured spinal cord based on microenvironment imbalance, including medications, cell transplantation, exosomes, tissue engineering, cell reprogramming, and rehabilitation. The current state of translational research on SCI and future directions are also discussed. The development of a combined, precise, and multitemporal strategy for repairing the injured spinal cord is a potential future direction.
Collapse
|
15
|
Characterization of a Novel Aspect of Tissue Scarring Following Experimental Spinal Cord Injury and the Implantation of Bioengineered Type-I Collagen Scaffolds in the Adult Rat: Involvement of Perineurial-like Cells? Int J Mol Sci 2022; 23:ijms23063221. [PMID: 35328642 PMCID: PMC8954100 DOI: 10.3390/ijms23063221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 03/04/2022] [Accepted: 03/15/2022] [Indexed: 11/17/2022] Open
Abstract
Numerous intervention strategies have been developed to promote functional tissue repair following experimental spinal cord injury (SCI), including the bridging of lesion-induced cystic cavities with bioengineered scaffolds. Integration between such implanted scaffolds and the lesioned host spinal cord is critical for supporting regenerative growth, but only moderate-to-low degrees of success have been reported. Light and electron microscopy were employed to better characterise the fibroadhesive scarring process taking place after implantation of a longitudinally microstructured type-I collagen scaffold into unilateral mid-cervical resection injuries of the adult rat spinal cord. At long survival times (10 weeks post-surgery), sheets of tightly packed cells (of uniform morphology) could be seen lining the inner surface of the repaired dura mater of lesion-only control animals, as well as forming a barrier along the implant–host interface of the scaffold-implanted animals. The highly uniform ultrastructural features of these scarring cells and their anatomical continuity with the local, reactive spinal nerve roots strongly suggest their identity to be perineurial-like cells. This novel aspect of the cellular composition of reactive spinal cord tissue highlights the increasingly complex nature of fibroadhesive scarring involved in traumatic injury, and particularly in response to the implantation of bioengineered collagen scaffolds.
Collapse
|
16
|
Shang Z, Wang R, Li D, Chen J, Zhang B, Wang M, Wang X, Wanyan P. Spinal Cord Injury: A Systematic Review and Network Meta-Analysis of Therapeutic Strategies Based on 15 Types of Stem Cells in Animal Models. Front Pharmacol 2022; 13:819861. [PMID: 35359872 PMCID: PMC8964098 DOI: 10.3389/fphar.2022.819861] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 02/04/2022] [Indexed: 12/13/2022] Open
Abstract
Objective: The optimal therapeutic strategies of stem cells for spinal cord injury (SCI) are fully explored in animal studies to promote the translation of preclinical findings to clinical practice, also to provide guidance for future animal experiments and clinical studies. Methods: PubMed, Web of Science, Embase, CNKI, Wangfang, VIP, and CBM were searched from inception to September 2021. Screening of search results, data extraction, and references quality evaluation were undertaken independently by two reviewers. Results and Discussion: A total of 188 studies were included for data analysis. Results of traditional meta-analysis showed that all 15 diverse types of stem cells could significantly improve locomotor function of animals with SCI, and results of further network meta-analysis showed that adipose-derived mesenchymal stem cells had the greatest therapeutic potential for SCI. Moreover, a higher dose (≥1 × 106) of stem cell transplantation had better therapeutic effect, transplantation in the subacute phase (3–14 days, excluding 3 days) was the optimal timing, and intralesional transplantation was the optimal route. However, the evidence of current animal studies is of limited quality, and more high-quality research is needed to further explore the optimal therapeutic strategies of stem cells, while the design and implementation of experiments, as well as measurement and reporting of results for animal studies, need to be further improved and standardized to reduce the risk when the results of animal studies are translated to the clinic. Systematic Review Registration: [website], identifier [registration number].
Collapse
Affiliation(s)
- Zhizhong Shang
- The First Clinical Medical School of Lanzhou University, Lanzhou, China
| | - Ruirui Wang
- The First Clinical Medical School of Lanzhou University, Lanzhou, China
| | - Dongliang Li
- The First Clinical Medical School of Lanzhou University, Lanzhou, China
| | - Jinlei Chen
- The First Clinical Medical School of Lanzhou University, Lanzhou, China
| | - Baolin Zhang
- The First Clinical Medical School of Lanzhou University, Lanzhou, China
| | - Mingchuan Wang
- The First Clinical Medical School of Lanzhou University, Lanzhou, China
| | - Xin Wang
- The First Clinical Medical School of Lanzhou University, Lanzhou, China
- Chengren Institute of Traditional Chinese Medicine, Lanzhou, China
- Department of Spine, Changzheng Hospital, Naval Medical University, Shanghai, China
- *Correspondence: Xin Wang, ; Pingping Wanyan,
| | - Pingping Wanyan
- Gansu University of Chinese Medicine, Lanzhou, China
- The Second Hospital of Lanzhou University, Lanzhou, China
- *Correspondence: Xin Wang, ; Pingping Wanyan,
| |
Collapse
|
17
|
Kubota S, Kadone H, Shimizu Y, Koda M, Takahashi H, Miura K, Eto F, Furuya T, Sankai Y, Yamazaki M. Immediate effects of hybrid assistive limb gait training on lower limb function in a chronic myelopathy patient with postoperative late neurological deterioration. BMC Res Notes 2022; 15:89. [PMID: 35246256 PMCID: PMC8896224 DOI: 10.1186/s13104-022-05979-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 02/18/2022] [Indexed: 11/21/2022] Open
Abstract
Objective The Hybrid Assistive Limb (HAL) has recently been used to treat movement disorders. Although studies have shown its effectiveness for chronic myelopathy, the immediate effects of HAL gait training on lower limb function have not been clarified. We conducted HAL gait training and examined its immediate effects on a 69-year-old man with re-deterioration of myelopathy in the chronic phase after surgery for compression myelopathy. The HAL intervention was performed every 4 weeks for 10 total sessions. Immediately before and after each session, we analyzed the patient’s walking ability using the 10-m walk test. In the 4th HAL session, the gastrocnemius muscle activity was measured bilaterally using a synchronized motion capture-electromyogram system. Results The training effects became steady after the 2nd session. In sessions 2–10, the step length increased from 0.56 to 0.63 m (mean: 0.031 m) immediately after HAL training. The motion capture-electromyogram analyses showed that considerable amounts of gastrocnemius muscle activity were detected during the stance and swing phases before HAL training. During and immediately after HAL training, gastrocnemius activity during the swing phase was diminished. HAL gait training has an immediate effect for inducing a normal gait pattern with less spasticity in those with chronic myelopathy. Supplementary Information The online version contains supplementary material available at 10.1186/s13104-022-05979-4.
Collapse
Affiliation(s)
- Shigeki Kubota
- Department of Orthopaedic Surgery, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan.
| | - Hideki Kadone
- Center for Innovative Medicine and Engineering, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Yukiyo Shimizu
- Department of Rehabilitation Medicine, Faculty of Medicine, University of Tsukuba, Ibaraki, 305-8575, Japan
| | - Masao Koda
- Department of Orthopaedic Surgery, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Hiroshi Takahashi
- Department of Orthopaedic Surgery, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Kousei Miura
- Department of Orthopaedic Surgery, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Fumihiko Eto
- Department of Orthopaedic Surgery, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Takeo Furuya
- Department of Orthopaedic Surgery, Chiba University Graduate School of Medicine, Chiba, 260-8677, Japan
| | - Yoshiyuki Sankai
- Faculty of Engineering, Information and Systems, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8573, Japan
| | - Masashi Yamazaki
- Department of Orthopaedic Surgery, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| |
Collapse
|
18
|
Gant KL, Guest JD, Palermo AE, Vedantam A, Jimsheleishvili G, Bunge MB, Brooks AE, Anderson KD, Thomas CK, Santamaria AJ, Perez MA, Curiel R, Nash MS, Saraf-Lavi E, Pearse DD, Widerström-Noga E, Khan A, Dietrich WD, Levi AD. Phase 1 Safety Trial of Autologous Human Schwann Cell Transplantation in Chronic Spinal Cord Injury. J Neurotrauma 2022; 39:285-299. [PMID: 33757304 PMCID: PMC9360180 DOI: 10.1089/neu.2020.7590] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
A phase 1 open-label, non-randomized clinical trial was conducted to determine feasibility and safety of autologous human Schwann cell (ahSC) transplantation accompanied by rehabilitation in participants with chronic spinal cord injury (SCI). Magnetic resonance imaging (MRI) was used to screen eligible participants to estimate an individualized volume of cell suspension to be implanted. The trial incorporated standardized multi-modal rehabilitation before and after cell delivery. Participants underwent sural nerve harvest, and ahSCs were isolated and propagated in culture. The dose of culture-expanded ahSCs injected into the chronic spinal cord lesion of each individual followed a cavity-filling volume approach. Primary outcome measures for safety and trend-toward efficacy were assessed. Two participants with American Spinal Injury Association Impairment Scale (AIS) A and two participants with incomplete chronic SCI (AIS B, C) were each enrolled in cervical and thoracic SCI cohorts (n = 8 total). All participants completed the study per protocol, and no serious adverse events related to sural nerve harvest or ahSC transplantation were reported. Urinary tract infections and skin abrasions were the most common adverse events reported. One participant experienced a 4-point improvement in motor function, a 6-point improvement in sensory function, and a 1-level improvement in neurological level of injury. Follow-up MRI in the cervical (6 months) and thoracic (24 months) cohorts revealed a reduction in cyst volume after transplantation with reduced effect over time. This phase 1 trial demonstrated the feasibility and safety of ahSC transplantation combined with a multi-modal rehabilitation protocol for participants with chronic SCI.
Collapse
Affiliation(s)
- Katie L. Gant
- The Miami Project to Cure Paralysis, University of Miami, Miami, Florida, USA
- Department of Neurological Surgery, University of Miami, Miami, Florida, USA
| | - James D. Guest
- The Miami Project to Cure Paralysis, University of Miami, Miami, Florida, USA
- Department of Neurological Surgery, University of Miami, Miami, Florida, USA
- Department of Neuroscience, University of Miami, Miami, Florida, USA
| | - Anne E. Palermo
- The Miami Project to Cure Paralysis, University of Miami, Miami, Florida, USA
- Department of Neurological Surgery, University of Miami, Miami, Florida, USA
| | - Aditya Vedantam
- The Miami Project to Cure Paralysis, University of Miami, Miami, Florida, USA
- Department of Neurological Surgery, University of Miami, Miami, Florida, USA
| | - George Jimsheleishvili
- The Miami Project to Cure Paralysis, University of Miami, Miami, Florida, USA
- Department of Neurological Surgery, University of Miami, Miami, Florida, USA
| | - Mary Bartlett Bunge
- The Miami Project to Cure Paralysis, University of Miami, Miami, Florida, USA
- Department of Neurological Surgery, University of Miami, Miami, Florida, USA
- Department of Neuroscience, University of Miami, Miami, Florida, USA
- Department of Cell Biology, University of Miami, Miami, Florida, USA
- Department of Neurology, University of Miami, Miami, Florida, USA
- Department of Interdisciplinary Stem Cell Institute, University of Miami, Miami, Florida, USA
| | - Adriana E. Brooks
- The Miami Project to Cure Paralysis, University of Miami, Miami, Florida, USA
- Department of Interdisciplinary Stem Cell Institute, University of Miami, Miami, Florida, USA
| | - Kim D. Anderson
- Department of Physical Medicine and Rehabilitation, Case Western Reserve University, Metrohealth Medical Center, Cleveland, Ohio, USA
| | - Christine K. Thomas
- The Miami Project to Cure Paralysis, University of Miami, Miami, Florida, USA
- Department of Neurological Surgery, University of Miami, Miami, Florida, USA
| | - Andrea J. Santamaria
- The Miami Project to Cure Paralysis, University of Miami, Miami, Florida, USA
- Department of Neurological Surgery, University of Miami, Miami, Florida, USA
| | - Monica A. Perez
- The Miami Project to Cure Paralysis, University of Miami, Miami, Florida, USA
- Department of Neurological Surgery, University of Miami, Miami, Florida, USA
- Bruce W. Carter Department of Veterans Affairs Medical Center, Miami, Florida, USA
- Shirley Ryan AbilityLab, Northwestern University, Edward Hines Jr, VA Hospital, Chicago, Illinois, USA
| | - Rosie Curiel
- Department of Psychiatry, University of Miami, Miami, Florida, USA
| | - Mark S. Nash
- Department of Rehabilitation Medicine, University of Miami, Miami, Florida, USA
| | - Efrat Saraf-Lavi
- Department of Radiology, University of Miami, Miami, Florida, USA
| | - Damien D. Pearse
- Department of Neuroscience, University of Miami, Miami, Florida, USA
- Department of Interdisciplinary Stem Cell Institute, University of Miami, Miami, Florida, USA
- Bruce W. Carter Department of Veterans Affairs Medical Center, Miami, Florida, USA
- Shirley Ryan AbilityLab, Northwestern University, Edward Hines Jr, VA Hospital, Chicago, Illinois, USA
| | - Eva Widerström-Noga
- The Miami Project to Cure Paralysis, University of Miami, Miami, Florida, USA
- Department of Neurological Surgery, University of Miami, Miami, Florida, USA
- Department of Neuroscience, University of Miami, Miami, Florida, USA
- Department of Rehabilitation Medicine, University of Miami, Miami, Florida, USA
- Bruce W. Carter Department of Veterans Affairs Medical Center, Miami, Florida, USA
| | - Aisha Khan
- The Miami Project to Cure Paralysis, University of Miami, Miami, Florida, USA
- Department of Interdisciplinary Stem Cell Institute, University of Miami, Miami, Florida, USA
| | - W. Dalton Dietrich
- The Miami Project to Cure Paralysis, University of Miami, Miami, Florida, USA
- Department of Neurological Surgery, University of Miami, Miami, Florida, USA
- Department of Neuroscience, University of Miami, Miami, Florida, USA
- Department of Cell Biology, University of Miami, Miami, Florida, USA
- Department of Neurology, University of Miami, Miami, Florida, USA
- Department of Interdisciplinary Stem Cell Institute, University of Miami, Miami, Florida, USA
| | - Allan D. Levi
- The Miami Project to Cure Paralysis, University of Miami, Miami, Florida, USA
- Department of Neurological Surgery, University of Miami, Miami, Florida, USA
- Department of Neuroscience, University of Miami, Miami, Florida, USA
| |
Collapse
|
19
|
Leemhuis E, Giuffrida V, De Martino ML, Forte G, Pecchinenda A, De Gennaro L, Giannini AM, Pazzaglia M. Rethinking the Body in the Brain after Spinal Cord Injury. J Clin Med 2022; 11:jcm11020388. [PMID: 35054089 PMCID: PMC8780443 DOI: 10.3390/jcm11020388] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Accepted: 01/12/2022] [Indexed: 02/05/2023] Open
Abstract
Spinal cord injuries (SCI) are disruptive neurological events that severly affect the body leading to the interruption of sensorimotor and autonomic pathways. Recent research highlighted SCI-related alterations extend beyond than the expected network, involving most of the central nervous system and goes far beyond primary sensorimotor cortices. The present perspective offers an alternative, useful way to interpret conflicting findings by focusing on the deafferented and deefferented body as the central object of interest. After an introduction to the main processes involved in reorganization according to SCI, we will focus separately on the body regions of the head, upper limbs, and lower limbs in complete, incomplete, and deafferent SCI participants. On one hand, the imprinting of the body’s spatial organization is entrenched in the brain such that its representation likely lasts for the entire lifetime of patients, independent of the severity of the SCI. However, neural activity is extremely adaptable, even over short time scales, and is modulated by changing conditions or different compensative strategies. Therefore, a better understanding of both aspects is an invaluable clinical resource for rehabilitation and the successful use of modern robotic technologies.
Collapse
Affiliation(s)
- Erik Leemhuis
- Department of Psychology, Sapienza University of Rome, Via dei Marsi 78, 00185 Rome, Italy; (E.L.); (V.G.); (M.L.D.M.); (A.P.); (L.D.G.); (A.M.G.)
- Action and Body Lab, IRCCS Fondazione Santa Lucia, Via Ardeatina 306, 00179 Rome, Italy
| | - Valentina Giuffrida
- Department of Psychology, Sapienza University of Rome, Via dei Marsi 78, 00185 Rome, Italy; (E.L.); (V.G.); (M.L.D.M.); (A.P.); (L.D.G.); (A.M.G.)
- Action and Body Lab, IRCCS Fondazione Santa Lucia, Via Ardeatina 306, 00179 Rome, Italy
| | - Maria Luisa De Martino
- Department of Psychology, Sapienza University of Rome, Via dei Marsi 78, 00185 Rome, Italy; (E.L.); (V.G.); (M.L.D.M.); (A.P.); (L.D.G.); (A.M.G.)
- Action and Body Lab, IRCCS Fondazione Santa Lucia, Via Ardeatina 306, 00179 Rome, Italy
| | - Giuseppe Forte
- Department of Psychology, Sapienza University of Rome, Via dei Marsi 78, 00185 Rome, Italy; (E.L.); (V.G.); (M.L.D.M.); (A.P.); (L.D.G.); (A.M.G.)
- Action and Body Lab, IRCCS Fondazione Santa Lucia, Via Ardeatina 306, 00179 Rome, Italy
- Correspondence: (G.F.); (M.P.); Tel.: +39-6-49917633 (M.P.)
| | - Anna Pecchinenda
- Department of Psychology, Sapienza University of Rome, Via dei Marsi 78, 00185 Rome, Italy; (E.L.); (V.G.); (M.L.D.M.); (A.P.); (L.D.G.); (A.M.G.)
| | - Luigi De Gennaro
- Department of Psychology, Sapienza University of Rome, Via dei Marsi 78, 00185 Rome, Italy; (E.L.); (V.G.); (M.L.D.M.); (A.P.); (L.D.G.); (A.M.G.)
- Action and Body Lab, IRCCS Fondazione Santa Lucia, Via Ardeatina 306, 00179 Rome, Italy
| | - Anna Maria Giannini
- Department of Psychology, Sapienza University of Rome, Via dei Marsi 78, 00185 Rome, Italy; (E.L.); (V.G.); (M.L.D.M.); (A.P.); (L.D.G.); (A.M.G.)
| | - Mariella Pazzaglia
- Department of Psychology, Sapienza University of Rome, Via dei Marsi 78, 00185 Rome, Italy; (E.L.); (V.G.); (M.L.D.M.); (A.P.); (L.D.G.); (A.M.G.)
- Action and Body Lab, IRCCS Fondazione Santa Lucia, Via Ardeatina 306, 00179 Rome, Italy
- Correspondence: (G.F.); (M.P.); Tel.: +39-6-49917633 (M.P.)
| |
Collapse
|
20
|
Sardella-Silva G, Mietto BS, Ribeiro-Resende VT. Four Seasons for Schwann Cell Biology, Revisiting Key Periods: Development, Homeostasis, Repair, and Aging. Biomolecules 2021; 11:1887. [PMID: 34944531 PMCID: PMC8699407 DOI: 10.3390/biom11121887] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 12/08/2021] [Accepted: 12/10/2021] [Indexed: 01/28/2023] Open
Abstract
Like the seasons of the year, all natural things happen in stages, going through adaptations when challenged, and Schwann cells are a great example of that. During maturation, these cells regulate several steps in peripheral nervous system development. The Spring of the cell means the rise and bloom through organized stages defined by time-dependent regulation of factors and microenvironmental influences. Once matured, the Summer of the cell begins: a high energy stage focused on maintaining adult homeostasis. The Schwann cell provides many neuron-glia communications resulting in the maintenance of synapses. In the peripheral nervous system, Schwann cells are pivotal after injuries, balancing degeneration and regeneration, similarly to when Autumn comes. Their ability to acquire a repair phenotype brings the potential to reconnect axons to targets and regain function. Finally, Schwann cells age, not only by growing old, but also by imposed environmental cues, like loss of function induced by pathologies. The Winter of the cell presents as reduced activity, especially regarding their role in repair; this reflects on the regenerative potential of older/less healthy individuals. This review gathers essential information about Schwann cells in different stages, summarizing important participation of this intriguing cell in many functions throughout its lifetime.
Collapse
Affiliation(s)
- Gabriela Sardella-Silva
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil;
- Núcleo Multidisciplinar de Pesquisa em Biologia (Numpex-Bio), Campus de Duque de Caxias Geraldo Guerra Cidade, Universidade Federal do Rio de Janeiro, Duque de Caxias 25255-030, RJ, Brazil
| | - Bruno Siqueira Mietto
- Instituto de Ciências Biológicas, Universidade Federal de Juiz de Fora, Juiz de Fora 36036-900, MG, Brazil;
| | - Victor Túlio Ribeiro-Resende
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil;
- Núcleo Multidisciplinar de Pesquisa em Biologia (Numpex-Bio), Campus de Duque de Caxias Geraldo Guerra Cidade, Universidade Federal do Rio de Janeiro, Duque de Caxias 25255-030, RJ, Brazil
| |
Collapse
|
21
|
Perez JC, Gerber YN, Perrin FE. Dynamic Diversity of Glial Response Among Species in Spinal Cord Injury. Front Aging Neurosci 2021; 13:769548. [PMID: 34899275 PMCID: PMC8662749 DOI: 10.3389/fnagi.2021.769548] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 10/29/2021] [Indexed: 12/11/2022] Open
Abstract
The glial scar that forms after traumatic spinal cord injury (SCI) is mostly composed of microglia, NG2 glia, and astrocytes and plays dual roles in pathophysiological processes induced by the injury. On one hand, the glial scar acts as a chemical and physical obstacle to spontaneous axonal regeneration, thus preventing functional recovery, and, on the other hand, it partly limits lesion extension. The complex activation pattern of glial cells is associated with cellular and molecular crosstalk and interactions with immune cells. Interestingly, response to SCI is diverse among species: from amphibians and fishes that display rather limited (if any) glial scarring to mammals that exhibit a well-identifiable scar. Additionally, kinetics of glial activation varies among species. In rodents, microglia become activated before astrocytes, and both glial cell populations undergo activation processes reflected amongst others by proliferation and migration toward the injury site. In primates, glial cell activation is delayed as compared to rodents. Here, we compare the spatial and temporal diversity of the glial response, following SCI amongst species. A better understanding of mechanisms underlying glial activation and scar formation is a prerequisite to develop timely glial cell-specific therapeutic strategies that aim to increase functional recovery.
Collapse
Affiliation(s)
| | - Yannick N Gerber
- MMDN, Université de Montpellier, EPHE, INSERM, Montpellier, France
| | - Florence E Perrin
- MMDN, Université de Montpellier, EPHE, INSERM, Montpellier, France.,Institut Universitaire de France (IUF), Paris, France
| |
Collapse
|
22
|
Adhikari K, Dolma S, Mamidi T, Roy A, Pathak Z, Kumar H. Tomographic Imaging and Correlation to Quantify Vascular and Inflammatory Changes in an Experimental Spinal Cord Injury. ACS Chem Neurosci 2021; 12:3864-3872. [PMID: 34628864 DOI: 10.1021/acschemneuro.1c00390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Spinal cord injury (SCI) is a devastating condition causing the loss of sensory and motor functions. SCI pathology is multifaceted, encompassing inflammation, scarring, neuronal damage, and vascular and tissue remodeling. The dynamics of SCI rapidly transform from acute, sub-acute, and chronic phases. The rapidly changing environment necessitates the real-time monitoring of disease severity. Therefore, in this study, we used the IVIS spectrum, a noninvasive fluorescence imaging modality, to monitor the disease pathology in live animals. We used near-infrared fluorescence imaging agents including Angiosense 750 EX, a probe that detects vascular changes, and Cat B 680 FAST, a probe that detects inflammation at various day points post injury (DPI), that is, DPI-1, DPI-14, and DPI-28. We quantified the pathophysiological changes after SCI using IVIS in live animals. As a result, we observed distinct differences in the disease progression between injured and sham mice. Moreover, live imaging showed a good correlation with behavioral studies, protein expression, and immunohistological analysis. Hence, the goal of this study was to introduce a new optical imaging modality that offers a determination of disease severity and the advantage of accelerated imaging of the correlated biomarkers in a real-time and dynamic manner. This study concluded that Cat B 680 Fast and Angiosense 750 EX could be used to assess the disease severity after SCI. Furthermore, our study suggests that the noninvasive fluorescence optical imaging modality offers a unique approach in monitoring neuroinflammatory diseases in live animals.
Collapse
Affiliation(s)
- Kirti Adhikari
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Sonam Dolma
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Teena Mamidi
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Abhishek Roy
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Zarna Pathak
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Hemant Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat 382355, India
| |
Collapse
|
23
|
Shinozaki M, Nagoshi N, Nakamura M, Okano H. Mechanisms of Stem Cell Therapy in Spinal Cord Injuries. Cells 2021; 10:cells10102676. [PMID: 34685655 PMCID: PMC8534136 DOI: 10.3390/cells10102676] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 09/28/2021] [Accepted: 10/04/2021] [Indexed: 12/13/2022] Open
Abstract
Every year, 0.93 million people worldwide suffer from spinal cord injury (SCI) with irretrievable sequelae. Rehabilitation, currently the only available treatment, does not restore damaged tissues; therefore, the functional recovery of patients remains limited. The pathophysiology of spinal cord injuries is heterogeneous, implying that potential therapeutic targets differ depending on the time of injury onset, the degree of injury, or the spinal level of injury. In recent years, despite a significant number of clinical trials based on various types of stem cells, these aspects of injury have not been effectively considered, resulting in difficult outcomes of trials. In a specialty such as cancerology, precision medicine based on a patient’s characteristics has brought indisputable therapeutic advances. The objective of the present review is to promote the development of precision medicine in the field of SCI. Here, we first describe the multifaceted pathophysiology of SCI, with the temporal changes after injury, the characteristics of the chronic phase, and the subtypes of complete injury. We then detail the appropriate targets and related mechanisms of the different types of stem cell therapy for each pathological condition. Finally, we highlight the great potential of stem cell therapy in cervical SCI.
Collapse
Affiliation(s)
- Munehisa Shinozaki
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan;
| | - Narihito Nagoshi
- Department of Orthopedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan; (N.N.); (M.N.)
| | - Masaya Nakamura
- Department of Orthopedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan; (N.N.); (M.N.)
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan;
- Correspondence:
| |
Collapse
|
24
|
Richard SA, Sackey M. Elucidating the Pivotal Neuroimmunomodulation of Stem Cells in Spinal Cord Injury Repair. Stem Cells Int 2021; 2021:9230866. [PMID: 34341666 PMCID: PMC8325586 DOI: 10.1155/2021/9230866] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Revised: 07/03/2021] [Accepted: 07/17/2021] [Indexed: 12/11/2022] Open
Abstract
Spinal cord injury (SCI) is a distressing incident with abrupt onset of the motor as well as sensory dysfunction, and most often, the injury occurs as result of high-energy or velocity accidents as well as contact sports and falls in the elderly. The key challenges associated with nerve repair are the lack of self-repair as well as neurotrophic factors and primary and secondary neuronal apoptosis, as well as factors that prevent the regeneration of axons locally. Neurons that survive the initial traumatic damage may be lost due to pathogenic activities like neuroinflammation and apoptosis. Implanted stem cells are capable of differentiating into neural cells that replace injured cells as well as offer local neurotrophic factors that aid neuroprotection, immunomodulation, axonal sprouting, axonal regeneration, and remyelination. At the microenvironment of SCI, stem cells are capable of producing growth factors like brain-derived neurotrophic factor and nerve growth factor which triggers neuronal survival as well as axonal regrowth. Although stem cells have proven to be of therapeutic value in SCI, the major disadvantage of some of the cell types is the risk for tumorigenicity due to the contamination of undifferentiated cells prior to transplantation. Local administration of stem cells via either direct cellular injection into the spinal cord parenchyma or intrathecal administration into the subarachnoid space is currently the best transplantation modality for stem cells during SCI.
Collapse
Affiliation(s)
- Seidu A. Richard
- Department of Medicine, Princefield University, P.O. Box MA128, Ho, Ghana
| | - Marian Sackey
- Department of Pharmacy, Ho Teaching Hospital, P.O. Box MA-374, Ho, Ghana
| |
Collapse
|
25
|
Monje PV, Deng L, Xu XM. Human Schwann Cell Transplantation for Spinal Cord Injury: Prospects and Challenges in Translational Medicine. Front Cell Neurosci 2021; 15:690894. [PMID: 34220455 PMCID: PMC8249939 DOI: 10.3389/fncel.2021.690894] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Accepted: 05/21/2021] [Indexed: 01/18/2023] Open
Abstract
The benefits of transplanting cultured Schwann cells (SCs) for the treatment of spinal cord injury (SCI) have been systematically investigated in experimental animals since the early 1990s. Importantly, human SC (hSC) transplantation for SCI has advanced to clinical testing and safety has been established via clinical trials conducted in the USA and abroad. However, multiple barriers must be overcome to enable accessible and effective treatments for SCI patients. This review presents available information on hSC transplantation for SCI with the intention to uncover gaps in our knowledge and discuss areas for future development. To this end, we introduce the historical progression of the work that supports existing and prospective clinical initiatives and explain the reasons for the choice of hSCs while also addressing their limitations as cell therapy products. A search of the relevant literature revealed that rat SCs have served as a preclinical model of reference since the onset of investigations, and that hSC transplants are relatively understudied, possibly due to the sophisticated resources and expertise needed for the traditional processing of hSC cultures from human nerves. In turn, we reason that additional experimentation and a reexamination of the available data are needed to understand the therapeutic value of hSC transplants taking into consideration that the manufacturing of the hSCs themselves may require further development for extended uses in basic research and clinical settings.
Collapse
Affiliation(s)
- Paula V. Monje
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Lingxiao Deng
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Xiao-Ming Xu
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
26
|
Trends of Chitosan Based Delivery Systems in Neuroregeneration and Functional Recovery in Spinal Cord Injuries. POLYSACCHARIDES 2021. [DOI: 10.3390/polysaccharides2020031] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Spinal cord injury (SCI) is one of the most complicated nervous system injuries with challenging treatment and recovery. Regenerative biomaterials such as chitosan are being reported for their wide use in filling the cavities, deliver curative drugs, and also provide adsorption sites for transplanted stem cells. Biomaterial scaffolds utilizing chitosan have shown certain therapeutic effects on spinal cord injury repair with some limitations. Chitosan-based delivery in stem cell transplantation is another strategy that has shown decent success. Stem cells can be directed to differentiate into neurons or glia in vitro. Stem cell-based therapy, biopolymer chitosan delivery strategies, and scaffold-based therapeutic strategies have been advancing as a combinatorial approach for spinal cord injury repair. In this review, we summarize the recent progress in the treatment strategies of SCI due to the use of bioactivity of chitosan-based drug delivery systems. An emphasis on the role of chitosan in neural regeneration has also been highlighted.
Collapse
|
27
|
Brown AR, Martinez M. Chronic inactivation of the contralesional hindlimb motor cortex after thoracic spinal cord hemisection impedes locomotor recovery in the rat. Exp Neurol 2021; 343:113775. [PMID: 34081986 DOI: 10.1016/j.expneurol.2021.113775] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 05/13/2021] [Accepted: 05/27/2021] [Indexed: 10/21/2022]
Abstract
After incomplete spinal cord injury (SCI), cortical plasticity is involved in hindlimb locomotor recovery. Nevertheless, whether cortical activity is required for motor map plasticity and recovery remains unresolved. Here, we combined a unilateral thoracic spinal cord injury (SCI) with a cortical inactivation protocol that uncovered a functional role of contralesional cortical activity in hindlimb recovery and ipsilesional map plasticity. In adult rats, left hindlimb paralysis was induced by sectioning half of the spinal cord at the thoracic level (hemisection) and we used a continuous infusion of muscimol (GABAA agonist, 10 mM, 0.11 µl/h) delivered via implanted osmotic pump (n = 9) to chronically inactivate the contralesional hindlimb motor cortex. Hemisected rats with saline infusion served as a SCI control group (n = 8), and intact rats with muscimol infusion served as an inactivation control group (n = 6). Locomotion was assessed in an open field, on a horizontal ladder, and on a treadmill prior to and for three weeks after hemisection. Cortical inactivation after hemisection significantly impeded hindlimb locomotor recovery in all tasks and specifically disrupted the ability of rats to generate proper flexion of the affected hindlimb during stepping compared to SCI controls, with no significant effect of inactivation in intact rats. Chronic and acute (n = 4) cortical inactivation after hemisection also significantly reduced the representation of the affected hindlimb in the ipsilesional motor cortex derived with intracortical microsimulation (ICMS). Our results provide evidence that residual activity in the contralesional hindlimb motor cortex after thoracic hemisection contributes to spontaneous locomotor recovery and map plasticity.
Collapse
Affiliation(s)
- Andrew R Brown
- Département de Neurosciences Groupe de recherche sur le système nerveux central (GRSNC) and Centre Interdisciplinaire de Recherche sur le Cerveau au service de l'Apprentissage (CIRCA), Université de Montréal, Québec, Canada; CIUSSS du Nord-de-l'Île-de-Montréal, Québec, Canada
| | - Marina Martinez
- Département de Neurosciences Groupe de recherche sur le système nerveux central (GRSNC) and Centre Interdisciplinaire de Recherche sur le Cerveau au service de l'Apprentissage (CIRCA), Université de Montréal, Québec, Canada; CIUSSS du Nord-de-l'Île-de-Montréal, Québec, Canada.
| |
Collapse
|
28
|
Pizzolato C, Gunduz MA, Palipana D, Wu J, Grant G, Hall S, Dennison R, Zafonte RD, Lloyd DG, Teng YD. Non-invasive approaches to functional recovery after spinal cord injury: Therapeutic targets and multimodal device interventions. Exp Neurol 2021; 339:113612. [DOI: 10.1016/j.expneurol.2021.113612] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 12/24/2020] [Accepted: 01/11/2021] [Indexed: 12/16/2022]
|
29
|
Wang F, Zu Z, Wu TL, Yan X, Lu M, Yang PF, Byun NE, Reed JL, Gore JC, Chen LM. Sensitivity and specificity of CEST and NOE MRI in injured spinal cord in monkeys. NEUROIMAGE-CLINICAL 2021; 30:102633. [PMID: 33780866 PMCID: PMC8039857 DOI: 10.1016/j.nicl.2021.102633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 02/18/2021] [Accepted: 03/12/2021] [Indexed: 11/04/2022]
Abstract
Compare sensitivity and specificity of CEST and NOE measures from 6-pool fitting. Differentiate regional molecular signatures at and around spinal cord injury. Provide parameters that improve the diagnostic accuracy of molecular alteration. Down-sampled data acquisition can capture the characteristic molecular profile. High translational potential for clinical assessment of spinal cord injury.
Purpose The sensitivity and accuracy of chemical exchange saturation transfer (CEST) and nuclear Overhauser enhancement (NOE) effects for assessing injury-associated changes in cervical spinal cords were evaluated in squirrel monkeys. Multiple interacting pools of protons, including one identified by an NOE at −1.6 ppm relative to water (NOE(-1.6)), were derived and quantified from fitting proton Z-spectra. The effects of down-sampled data acquisitions and corrections for non-specific factors including T1, semi-solid magnetization transfer, and direct saturation of free water (DS), were investigated. The overall goal is to develop a protocol for rapid data acquisition for assessing the molecular signatures of the injured spinal cord and its surrounding regions. Methods MRI scans were recorded of anesthetized squirrel monkeys at 9.4 T, before and after a unilateral dorsal column sectioning of the cervical spinal cord. Z-spectral images at 51 different RF offsets were acquired. The amplitudes of CEST and NOE effects from multiple proton pools were quantified using a six-pool Lorenzian fitting of each Z-spectrum (MTRmfit). In addition, down-sampled data using reduced selections of RF offsets were analyzed and compared. An apparent exchange-dependent relaxation (AREXmfit) method was also used to correct for non-specific factors in quantifying regional spectra around lesion sites. Results The parametric maps from multi-pool fitting using the complete sampling data (P51e) detected unilateral changes at and around the injury. The maps derived from selected twofold down-sampled data with appropriate interpolation (P26sI51) revealed quite similar spatial distributions of different pools as those obtained using P51e at each resonance shift. Across 10 subjects, both data acquisition schemes detected significant decreases in NOE(-3.5) and NOE(-1.6) and increases in DS(0.0) and CEST(3.5) at the lesion site relative to measures of the normal tissues before injury. AREXmfit of cysts and other abnormal tissues at and around the lesion site also exhibited significant changes, especially at 3.5, −1.6 and −3.5 ppm RF offsets. Conclusion These results confirm that a reduced set of RF offsets and down sampling are adequate for CEST imaging of injured spinal cord and allow shorter imaging times and/or permit additional signal averaging. AREXmfit correction improved the accuracy of CEST and NOE measures. The results provide a rapid (~13 mins), sensitive, and accurate protocol for deriving multiple NOE and CEST effects simultaneously in spinal cord imaging at high field.
Collapse
Affiliation(s)
- Feng Wang
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, USA; Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, TN, USA
| | - Zhongliang Zu
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, USA; Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, TN, USA
| | - Tung-Lin Wu
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, USA; Department of Biomedical Engineering, Vanderbilt University, TN, USA
| | - Xinqiang Yan
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, USA; Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, TN, USA
| | - Ming Lu
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, USA; Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, TN, USA
| | - Pai-Feng Yang
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, USA; Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, TN, USA
| | - Nellie E Byun
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, USA; Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, TN, USA
| | - Jamie L Reed
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, USA; Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, TN, USA
| | - John C Gore
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, USA; Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, TN, USA; Department of Biomedical Engineering, Vanderbilt University, TN, USA
| | - Li Min Chen
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, USA; Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, TN, USA.
| |
Collapse
|
30
|
Duncan GJ, Simkins TJ, Emery B. Neuron-Oligodendrocyte Interactions in the Structure and Integrity of Axons. Front Cell Dev Biol 2021; 9:653101. [PMID: 33763430 PMCID: PMC7982542 DOI: 10.3389/fcell.2021.653101] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 02/15/2021] [Indexed: 12/12/2022] Open
Abstract
The myelination of axons by oligodendrocytes is a highly complex cell-to-cell interaction. Oligodendrocytes and axons have a reciprocal signaling relationship in which oligodendrocytes receive cues from axons that direct their myelination, and oligodendrocytes subsequently shape axonal structure and conduction. Oligodendrocytes are necessary for the maturation of excitatory domains on the axon including nodes of Ranvier, help buffer potassium, and support neuronal energy metabolism. Disruption of the oligodendrocyte-axon unit in traumatic injuries, Alzheimer's disease and demyelinating diseases such as multiple sclerosis results in axonal dysfunction and can culminate in neurodegeneration. In this review, we discuss the mechanisms by which demyelination and loss of oligodendrocytes compromise axons. We highlight the intra-axonal cascades initiated by demyelination that can result in irreversible axonal damage. Both the restoration of oligodendrocyte myelination or neuroprotective therapies targeting these intra-axonal cascades are likely to have therapeutic potential in disorders in which oligodendrocyte support of axons is disrupted.
Collapse
Affiliation(s)
- Greg J. Duncan
- Jungers Center for Neurosciences Research, Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| | - Tyrell J. Simkins
- Jungers Center for Neurosciences Research, Department of Neurology, Oregon Health & Science University, Portland, OR, United States
- Vollum Institute, Oregon Health & Science University, Portland, OR, United States
- Department of Neurology, VA Portland Health Care System, Portland, OR, United States
| | - Ben Emery
- Jungers Center for Neurosciences Research, Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| |
Collapse
|
31
|
Muthu S, Jeyaraman M, Gulati A, Arora A. Current evidence on mesenchymal stem cell therapy for traumatic spinal cord injury: systematic review and meta-analysis. Cytotherapy 2021; 23:186-197. [PMID: 33183980 DOI: 10.1016/j.jcyt.2020.09.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Revised: 09/11/2020] [Accepted: 09/14/2020] [Indexed: 02/05/2023]
Abstract
BACKGROUND AIMS The authors aim to analyze the evidence in the literature regarding the efficacy and safety of mesenchymal stem cell (MSC) therapy in human subjects with traumatic spinal cord injury (SCI) and identify its potential role in the management of SCI. METHODS The authors conducted independent and duplicate searches of electronic databases, including PubMed, Embase and the Cochrane Library, until May 2020 for studies analyzing the efficacy and safety of stem cell therapy for SCI. American Spine Injury Association (ASIA) impairment scale (AIS) grade improvement, ASIA sensorimotor score, activities of daily living score, residual urine volume, bladder function improvement, somatosensory evoked potential (SSEP) improvement and adverse reactions were the outcomes analyzed. Analysis was performed in R platform using OpenMeta[Analyst] software. RESULTS Nineteen studies involving 670 patients were included for analysis. On analysis, the intervention group showed statistically significant improvement in AIS grade (P < 0.001), ASIA sensory score (P < 0.017), light touch (P < 0.001), pinprick (P = 0.046), bladder function (P = 0.012), residual urine volume (P = 0.023) and SSEP (P = 0.002). However, no significant difference was noted in motor score (P = 0.193) or activities of daily living score (P = 0.161). Although the intervention group had a significant increase in complications (P < 0.001), no serious or permanent adverse events were reported. On subgroup analysis, low concentration of MSCs (<5 × 107 cells) and initial AIS grade A presentation showed significantly better outcomes than their counterparts. CONCLUSIONS The authors' analysis establishes the efficacy and safety of MSC transplantation in terms of improvement in AIS grade, ASIA sensory score, bladder function and electrophysiological parameters like SSEP compared with controls, without major adverse events. However, further research is needed to standardize dose, timing, route and source of MSCs used for transplantation.
Collapse
Affiliation(s)
- Sathish Muthu
- Government Hospital, Velayuthampalayam, Karur, Tamil Nadu, India; Orthopaedic Research Group, Coimbatore, Tamil Nadu, India; Indian Stem Cells Study Group, Lucknow, India.
| | - Madhan Jeyaraman
- Orthopaedic Research Group, Coimbatore, Tamil Nadu, India; Indian Stem Cells Study Group, Lucknow, India; Department of Orthopaedics, School of Medical Sciences and Research, Sharda University, Greater Noida, India
| | - Arun Gulati
- Department of Orthopaedics, Kalpana Chawla Government Medical College & Hospital, Karnal, India
| | - Arunabh Arora
- Department of Orthopaedics, School of Medical Sciences and Research, Sharda University, Greater Noida, India
| |
Collapse
|
32
|
Santamaria AJ, Benavides FD, Saraiva PM, Anderson KD, Khan A, Levi AD, Dietrich WD, Guest JD. Neurophysiological Changes in the First Year After Cell Transplantation in Sub-acute Complete Paraplegia. Front Neurol 2021; 11:514181. [PMID: 33536992 PMCID: PMC7848788 DOI: 10.3389/fneur.2020.514181] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 11/05/2020] [Indexed: 12/15/2022] Open
Abstract
Neurophysiological testing can provide quantitative information about motor, sensory, and autonomic system connectivity following spinal cord injury (SCI). The clinical examination may be insufficiently sensitive and specific to reveal evolving changes in neural circuits after severe injury. Neurophysiologic data may provide otherwise imperceptible circuit information that has rarely been acquired in biologics clinical trials in SCI. We reported a Phase 1 study of autologous purified Schwann cell suspension transplantation into the injury epicenter of participants with complete subacute thoracic SCI, observing no clinical improvements. Here, we report longitudinal electrophysiological assessments conducted during the trial. Six participants underwent neurophysiology screening pre-transplantation with three post-transplantation neurophysiological assessments, focused on the thoracoabdominal region and lower limbs, including MEPs, SSEPs, voluntarily triggered EMG, and changes in GSR. We found several notable signals not detectable by clinical exam. In all six participants, thoracoabdominal motor connectivity was detected below the clinically assigned neurological level defined by sensory preservation. Additionally, small voluntary activations of leg and foot muscles or positive lower extremity MEPs were detected in all participants. Voluntary EMG was most sensitive to detect leg motor function. The recorded MEP amplitudes and latencies indicated a more caudal thoracic level above which amplitude recovery over time was observed. In contrast, further below, amplitudes showed less improvement, and latencies were increased. Intercostal spasms observed with EMG may also indicate this thoracic “motor level.” Galvanic skin testing revealed autonomic dysfunction in the hands above the injury levels. As an open-label study, we can establish no clear link between these observations and cell transplantation. This neurophysiological characterization may be of value to detect therapeutic effects in future controlled studies.
Collapse
Affiliation(s)
- Andrea J Santamaria
- The Miami Project to Cure Paralysis, Miller School of Medicine, The University of Miami, Miami, FL, United States
| | - Francisco D Benavides
- The Miami Project to Cure Paralysis, Miller School of Medicine, The University of Miami, Miami, FL, United States
| | - Pedro M Saraiva
- The Miami Project to Cure Paralysis, Miller School of Medicine, The University of Miami, Miami, FL, United States
| | - Kimberly D Anderson
- The Miami Project to Cure Paralysis, Miller School of Medicine, The University of Miami, Miami, FL, United States.,The Department of Neurological Surgery, Miller School of Medicine, The University of Miami, Miami, FL, United States
| | - Aisha Khan
- The Miami Project to Cure Paralysis, Miller School of Medicine, The University of Miami, Miami, FL, United States.,Miller School of Medicine, The Interdisciplinary Stem Cell Institute, The University of Miami, Miami, FL, United States
| | - Allan D Levi
- The Miami Project to Cure Paralysis, Miller School of Medicine, The University of Miami, Miami, FL, United States.,The Department of Neurological Surgery, Miller School of Medicine, The University of Miami, Miami, FL, United States
| | - W Dalton Dietrich
- The Miami Project to Cure Paralysis, Miller School of Medicine, The University of Miami, Miami, FL, United States.,The Department of Neurological Surgery, Miller School of Medicine, The University of Miami, Miami, FL, United States
| | - James D Guest
- The Miami Project to Cure Paralysis, Miller School of Medicine, The University of Miami, Miami, FL, United States.,The Department of Neurological Surgery, Miller School of Medicine, The University of Miami, Miami, FL, United States
| |
Collapse
|
33
|
Chen CZ, Neumann B, Förster S, Franklin RJM. Schwann cell remyelination of the central nervous system: why does it happen and what are the benefits? Open Biol 2021; 11:200352. [PMID: 33497588 PMCID: PMC7881176 DOI: 10.1098/rsob.200352] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 01/07/2021] [Indexed: 12/18/2022] Open
Abstract
Myelin sheaths, by supporting axonal integrity and allowing rapid saltatory impulse conduction, are of fundamental importance for neuronal function. In response to demyelinating injuries in the central nervous system (CNS), oligodendrocyte progenitor cells (OPCs) migrate to the lesion area, proliferate and differentiate into new oligodendrocytes that make new myelin sheaths. This process is termed remyelination. Under specific conditions, demyelinated axons in the CNS can also be remyelinated by Schwann cells (SCs), the myelinating cell of the peripheral nervous system. OPCs can be a major source of these CNS-resident SCs-a surprising finding given the distinct embryonic origins, and physiological compartmentalization of the peripheral and central nervous system. Although the mechanisms and cues governing OPC-to-SC differentiation remain largely undiscovered, it might nevertheless be an attractive target for promoting endogenous remyelination. This article will (i) review current knowledge on the origins of SCs in the CNS, with a particular focus on OPC to SC differentiation, (ii) discuss the necessary criteria for SC myelination in the CNS and (iii) highlight the potential of using SCs for myelin regeneration in the CNS.
Collapse
Affiliation(s)
| | | | | | - Robin J. M. Franklin
- Wellcome-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0AH, UK
| |
Collapse
|
34
|
Khaing ZZ, Cates LN, Dewees DM, Hyde JE, Gaing A, Birjandian Z, Hofstetter CP. Effect of Durotomy versus Myelotomy on Tissue Sparing and Functional Outcome after Spinal Cord Injury. J Neurotrauma 2020; 38:746-755. [PMID: 33121382 DOI: 10.1089/neu.2020.7297] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Various surgical strategies have been developed to alleviate elevated intraspinal pressure (ISP) following acute traumatic spinal cord injury (tSCI). Surgical decompression of either the dural (durotomy) or the dural and pial (myelotomy) lining of the spinal cord has been proposed. However, a direct comparison of these two strategies is lacking. Here, we compare the histological and functional effects of durotomy alone and durotomy plus myelotomy in a rodent model of acute thoracic tSCI. Our results indicate that tSCI causes local tissue edema and significantly elevates ISP (7.4 ± 0.3 mmHg) compared with physiological ISP (1.7 ± 0.4 mmHg; p < 0.001). Both durotomy alone and durotomy plus myelotomy effectively mitigate elevated local ISP (p < 0.001). Histological examination at 10 weeks after tSCI revealed that durotomy plus myelotomy promoted spinal tissue sparing by 13.7% compared with durotomy alone, and by 25.9% compared with tSCI-only (p < 0.0001). Both types of decompression surgeries elicited a significant beneficial impact on gray matter sparing (p < 0.01). Impressively, durotomy plus myelotomy surgery increased preservation of motor neurons by 174.3% compared with tSCI-only (p < 0.05). Durotomy plus myelotomy surgery also significantly promoted recovery of hindlimb locomotor function in an open-field test (p < 0.001). Interestingly, only durotomy alone resulted in favorable recovery of bladder and Ladder Walk performance. Combined, our data suggest that durotomy plus myelotomy following acute tSCI facilitates tissue sparing and recovery of locomotor function. In the future, biomarkers identifying spinal cord injuries that can benefit from either durotomy alone or durotomy plus myelotomy need to be developed.
Collapse
Affiliation(s)
- Zin Z Khaing
- Department of Neurological Surgery, The University of Washington, Seattle, Washington, USA
| | - Lindsay N Cates
- Department of Neurological Surgery, The University of Washington, Seattle, Washington, USA
| | - Dane M Dewees
- Department of Neurological Surgery, The University of Washington, Seattle, Washington, USA
| | - Jeffrey E Hyde
- Department of Neurological Surgery, The University of Washington, Seattle, Washington, USA
| | - Ashley Gaing
- Department of Neurological Surgery, The University of Washington, Seattle, Washington, USA
| | - Zeinab Birjandian
- Department of Neurological Surgery, The University of Washington, Seattle, Washington, USA
| | - Christoph P Hofstetter
- Department of Neurological Surgery, The University of Washington, Seattle, Washington, USA
| |
Collapse
|
35
|
Tsata V, Kroehne V, Wehner D, Rost F, Lange C, Hoppe C, Kurth T, Reinhardt S, Petzold A, Dahl A, Loeffler M, Reimer MM, Brand M. Reactive oligodendrocyte progenitor cells (re-)myelinate the regenerating zebrafish spinal cord. Development 2020; 147:dev193946. [PMID: 33158923 DOI: 10.1242/dev.193946] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Accepted: 10/28/2020] [Indexed: 12/11/2022]
Abstract
Spinal cord injury (SCI) results in loss of neurons, oligodendrocytes and myelin sheaths, all of which are not efficiently restored. The scarcity of oligodendrocytes in the lesion site impairs re-myelination of spared fibres, which leaves axons denuded, impedes signal transduction and contributes to permanent functional deficits. In contrast to mammals, zebrafish can functionally regenerate the spinal cord. Yet, little is known about oligodendroglial lineage biology and re-myelination capacity after SCI in a regeneration-permissive context. Here, we report that, in adult zebrafish, SCI results in axonal, oligodendrocyte and myelin sheath loss. We find that OPCs, the oligodendrocyte progenitor cells, survive the injury, enter a reactive state, proliferate and differentiate into oligodendrocytes. Concomitantly, the oligodendrocyte population is re-established to pre-injury levels within 2 weeks. Transcriptional profiling revealed that reactive OPCs upregulate the expression of several myelination-related genes. Interestingly, global reduction of axonal tracts and partial re-myelination, relative to pre-injury levels, persist at later stages of regeneration, yet are sufficient for functional recovery. Taken together, these findings imply that, in the zebrafish spinal cord, OPCs replace lost oligodendrocytes and, thus, re-establish myelination during regeneration.
Collapse
Affiliation(s)
- Vasiliki Tsata
- Center for Regenerative Therapies Dresden TU Dresden (CRTD) and Cluster of Excellence, Physics of Life (PoL), TU Dresden, Dresden 01307, Germany
| | - Volker Kroehne
- Center for Regenerative Therapies Dresden TU Dresden (CRTD) and Cluster of Excellence, Physics of Life (PoL), TU Dresden, Dresden 01307, Germany
| | - Daniel Wehner
- Center for Regenerative Therapies Dresden TU Dresden (CRTD) and Cluster of Excellence, Physics of Life (PoL), TU Dresden, Dresden 01307, Germany
- Max Planck Institute for the Science of Light, Erlangen 91058, Germany
- Max-Planck-Zentrum für Physik und Medizin, Erlangen 91058, Germany
| | - Fabian Rost
- Center for Regenerative Therapies Dresden TU Dresden (CRTD) and Cluster of Excellence, Physics of Life (PoL), TU Dresden, Dresden 01307, Germany
- Center for Information Services and High Performance Computing, TU Dresden, Dresden 01062, Germany
| | - Christian Lange
- Center for Regenerative Therapies Dresden TU Dresden (CRTD) and Cluster of Excellence, Physics of Life (PoL), TU Dresden, Dresden 01307, Germany
| | - Cornelia Hoppe
- Center for Regenerative Therapies Dresden TU Dresden (CRTD) and Cluster of Excellence, Physics of Life (PoL), TU Dresden, Dresden 01307, Germany
| | - Thomas Kurth
- Center for Molecular and Cellular Bioengineering (CMCB), TU Dresden, Technology Platform, Dresden 01307, Germany
| | - Susanne Reinhardt
- Dresden Genome Center c/o Center for Regenerative Therapies TU Dresden (CRTD), TU Dresden, Dresden 01307, Germany
| | - Andreas Petzold
- Dresden Genome Center c/o Center for Regenerative Therapies TU Dresden (CRTD), TU Dresden, Dresden 01307, Germany
| | - Andreas Dahl
- Dresden Genome Center c/o Center for Regenerative Therapies TU Dresden (CRTD), TU Dresden, Dresden 01307, Germany
| | - Markus Loeffler
- Center for Advancing Electronics Dresden (cfaed)/Dresden Center for Nanoanalysis (DCN), TU Dresden, Dresden 01062, Germany
| | - Michell M Reimer
- Center for Regenerative Therapies Dresden TU Dresden (CRTD) and Cluster of Excellence, Physics of Life (PoL), TU Dresden, Dresden 01307, Germany
| | - Michael Brand
- Center for Regenerative Therapies Dresden TU Dresden (CRTD) and Cluster of Excellence, Physics of Life (PoL), TU Dresden, Dresden 01307, Germany
| |
Collapse
|
36
|
Yao L, Shippy T, Li Y. Genetic analysis of the molecular regulation of electric fields-guided glia migration. Sci Rep 2020; 10:16821. [PMID: 33033380 PMCID: PMC7546725 DOI: 10.1038/s41598-020-74085-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Accepted: 08/31/2020] [Indexed: 11/09/2022] Open
Abstract
In a developing nervous system, endogenous electric field (EF) influence embryonic growth. We reported the EF-directed migration of both rat Schwann cells (SCs) and oligodendrocyte precursor cells (OPCs) and explored the molecular mechanism using RNA-sequencing assay. However, previous studies revealed the differentially expressed genes (DEGs) associated with EF-guided migration of SCs or OPCs alone. In this study, we performed joint differential expression analysis on the RNA-sequencing data from both cell types. We report a number of significantly enriched gene ontology (GO) terms that are related to the cytoskeleton, cell adhesion, and cell migration. Of the DEGs associated with these terms, nine up-regulated DEGs and 32 down-regulated DEGs showed the same direction of effect in both SCs and OPCs stimulated with EFs, while the remaining DEGs responded differently. Thus, our study reveals the similarities and differences in gene expression and cell migration regulation of different glial cell types in response to EF stimulation.
Collapse
Affiliation(s)
- Li Yao
- Department of Biological Sciences, Wichita State University, 1845 Fairmount Street, Wichita, KS, 67260, USA.
| | - Teresa Shippy
- Bioinformatics Specialist, KSU Bioinformatics Center, Kansas State University, Manhattan, KS, 66506, USA
| | - Yongchao Li
- Department of Biological Sciences, Wichita State University, 1845 Fairmount Street, Wichita, KS, 67260, USA
| |
Collapse
|
37
|
Pukos N, McTigue DM. Delayed short-term tamoxifen treatment does not promote remyelination or neuron sparing after spinal cord injury. PLoS One 2020; 15:e0235232. [PMID: 32735618 PMCID: PMC7394399 DOI: 10.1371/journal.pone.0235232] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 06/10/2020] [Indexed: 12/18/2022] Open
Abstract
The tamoxifen-dependent Cre/lox system in transgenic mice has become an important research tool across all scientific disciplines for manipulating gene expression in specific cell types. In these mouse models, Cre-recombination is not induced until tamoxifen is administered, which allows researchers to have temporal control of genetic modifications. Interestingly, tamoxifen has been identified as a potential therapy for spinal cord injury (SCI) and traumatic brain injury patients due to its neuroprotective properties. It is also reparative in that it stimulates oligodendrocyte differentiation and remyelination after toxin-induced demyelination. However, it is unknown whether tamoxifen is neuroprotective and neuroreparative when administration is delayed after SCI. To properly interpret data from transgenic mice in which tamoxifen treatment is delayed after SCI, it is necessary to identify the effects of tamoxifen alone on anatomical and functional recovery. In this study, female and male mice received a moderate mid-thoracic spinal cord contusion. Mice were then gavaged with corn oil or a high dose of tamoxifen from 19-22 days post-injury, and sacrificed 42 days post-injury. All mice underwent behavioral testing for the duration of the study, which revealed that tamoxifen treatment did not impact hindlimb motor recovery. Similarly, histological analyses revealed that tamoxifen had no effect on white matter sparing, total axon number, axon sprouting, glial reactivity, cell proliferation, oligodendrocyte number, or myelination, but tamoxifen did decrease the number of neurons in the dorsal and ventral horn. Semi-thin sections confirmed that axon demyelination and remyelination were unaffected by tamoxifen. Sex-specific responses to tamoxifen were also assessed, and there were no significant differences between female and male mice. These data suggest that delayed tamoxifen administration after SCI does not change functional recovery or improve tissue sparing in female or male mice.
Collapse
Affiliation(s)
- Nicole Pukos
- Neuroscience Graduate Program, The Ohio State University, Columbus, OH, United States of America
- Belford Center for Spinal Cord Injury, The Ohio State University, Columbus, OH, United States of America
| | - Dana M. McTigue
- Belford Center for Spinal Cord Injury, The Ohio State University, Columbus, OH, United States of America
- Department of Neuroscience, Wexner Medical Center, Ohio State University, Columbus, OH, United States of America
| |
Collapse
|
38
|
Fischer I, Dulin JN, Lane MA. Transplanting neural progenitor cells to restore connectivity after spinal cord injury. Nat Rev Neurosci 2020; 21:366-383. [PMID: 32518349 PMCID: PMC8384139 DOI: 10.1038/s41583-020-0314-2] [Citation(s) in RCA: 144] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/30/2020] [Indexed: 12/12/2022]
Abstract
Spinal cord injury remains a scientific and therapeutic challenge with great cost to individuals and society. The goal of research in this field is to find a means of restoring lost function. Recently we have seen considerable progress in understanding the injury process and the capacity of CNS neurons to regenerate, as well as innovations in stem cell biology. This presents an opportunity to develop effective transplantation strategies to provide new neural cells to promote the formation of new neuronal networks and functional connectivity. Past and ongoing clinical studies have demonstrated the safety of cell therapy, and preclinical research has used models of spinal cord injury to better elucidate the underlying mechanisms through which donor cells interact with the host and thus increase long-term efficacy. While a variety of cell therapies have been explored, we focus here on the use of neural progenitor cells obtained or derived from different sources to promote connectivity in sensory, motor and autonomic systems.
Collapse
Affiliation(s)
- Itzhak Fischer
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, USA.
| | - Jennifer N Dulin
- Department of Biology, Texas A&M University, College Station, TX, USA
| | - Michael A Lane
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, USA
| |
Collapse
|
39
|
O'Hare Doig RL, Santhakumar S, Fehily B, Raja S, Solomon T, Bartlett CA, Fitzgerald M, Hodgetts SI. Acute Cellular and Functional Changes With a Combinatorial Treatment of Ion Channel Inhibitors Following Spinal Cord Injury. Front Mol Neurosci 2020; 13:85. [PMID: 32670018 PMCID: PMC7331598 DOI: 10.3389/fnmol.2020.00085] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 04/24/2020] [Indexed: 01/12/2023] Open
Abstract
Reducing the extent of secondary degeneration following spinal cord injury (SCI) is necessary to preserve function, but treatment options have thus far been limited. A combination of the ion channel inhibitors Lomerizine (Lom), YM872 and oxATP, to inhibit voltage-gated Ca2+ channels, Ca2+ permeable AMPA receptors, and purinergic P2X7 receptors respectively, effectively limits secondary consequences of injury in in vitro and in vivo models of CNS injury. Here, we investigated the efficacy of these inhibitors in a clinically relevant model of SCI. Fischer (F344) rats were subjected to a moderate (150 kD) contusive SCI at thoracic level T10 and assessed at 2 weeks or 10 weeks post-injury. Lom was delivered orally twice daily and YM872 and oxATP were delivered via osmotic mini-pump implanted at the time of SCI until 2 weeks following injury. Open field locomotion analysis revealed that treatment with the three inhibitors in combination improved the rate of functional recovery of the hind limb (compared to controls) as early as 1-day post-injury, with beneficial effects persisting to 14 days post-injury, while all three inhibitors were present. At 2 weeks following combinatorial treatment, the functional improvement was associated with significantly decreased cyst size, increased immunoreactivity of β-III tubulin+ve axons, myelin basic protein, and reduced lipid peroxidation by-products, and increased CC1+ve oligodendrocytes and NG2+ve/PDGFα+ve oligodendrocyte progenitor cell densities, compared to vehicle-treated SCI animals. The combination of Lom, oxATP, and YM872 shows preclinical promise for control of secondary degeneration following SCI, and further investigation of long-term sustained treatment is warranted.
Collapse
Affiliation(s)
- Ryan L O'Hare Doig
- Experimental and Regenerative Neurosciences, School of Biological Sciences, The University of Western Australia, Crawley, WA, Australia.,Experimental and Regenerative Neurosciences, School of Human Sciences, The University of Western Australia, Crawley, WA, Australia.,Neil Sachse Centre for Spinal Cord Research, South Australian Health and Medical Research Institute, Adelaide, SA, Australia.,Adelaide Spinal Research Group, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, Australia
| | - Sreya Santhakumar
- Experimental and Regenerative Neurosciences, School of Human Sciences, The University of Western Australia, Crawley, WA, Australia.,Perron Institute for Neurological and Translational Science, Nedlands, WA, Australia
| | - Brooke Fehily
- Experimental and Regenerative Neurosciences, School of Biological Sciences, The University of Western Australia, Crawley, WA, Australia
| | - Sushmitha Raja
- Experimental and Regenerative Neurosciences, School of Biological Sciences, The University of Western Australia, Crawley, WA, Australia
| | - Tanya Solomon
- Experimental and Regenerative Neurosciences, School of Biological Sciences, The University of Western Australia, Crawley, WA, Australia
| | - Carole A Bartlett
- Experimental and Regenerative Neurosciences, School of Biological Sciences, The University of Western Australia, Crawley, WA, Australia
| | - Melinda Fitzgerald
- Experimental and Regenerative Neurosciences, School of Biological Sciences, The University of Western Australia, Crawley, WA, Australia.,Perron Institute for Neurological and Translational Science, Nedlands, WA, Australia.,Curtin Health Innovation Research Institute, Curtin University, Nedlands, WA, Australia
| | - Stuart I Hodgetts
- Experimental and Regenerative Neurosciences, School of Human Sciences, The University of Western Australia, Crawley, WA, Australia.,Perron Institute for Neurological and Translational Science, Nedlands, WA, Australia
| |
Collapse
|
40
|
Wu TL, Byun NE, Wang F, Mishra A, Janve VA, Chen LM, Gore JC. Longitudinal assessment of recovery after spinal cord injury with behavioral measures and diffusion, quantitative magnetization transfer and functional magnetic resonance imaging. NMR IN BIOMEDICINE 2020; 33:e4216. [PMID: 31943383 PMCID: PMC7155919 DOI: 10.1002/nbm.4216] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 10/08/2019] [Accepted: 10/15/2019] [Indexed: 05/09/2023]
Abstract
Spinal cord injuries (SCIs) are a leading cause of disability and can severely impact the quality of life. However, to date, the processes of spontaneous repair of damaged spinal cord remain incompletely understood, partly due to a lack of appropriate longitudinal tracking methods. Noninvasive, multiparametric magnetic resonance imaging (MRI) provides potential biomarkers for the comprehensive evaluation of spontaneous repair after SCI. In this study in rats, a clinically relevant contusion injury was introduced at the lumbar level that impairs both hindlimb motor and sensory functions. Quantitative MRI measurements were acquired at baseline and serially post-SCI for up to 2 wk. The progressions of injury and spontaneous recovery in both white and gray matter were tracked longitudinally using pool-size ratio (PSR) measurements derived from quantitative magnetization transfer (qMT) methods, measurements of water diffusion parameters using diffusion tensor imaging (DTI) and intrasegment functional connectivity derived from resting state functional MRI. Changes in these quantitative imaging measurements were correlated with behavioral readouts. We found (a) a progressive decrease in PSR values within 2 wk post-SCI, indicating a progressive demyelination at the center of the injury that was validated with histological staining, (b) PSR correlated closely with fractional anisotropy and transverse relaxation of free water, but did not show significant correlations with behavioral recovery, and (c) preliminary evidence that SCI induced a decrease in functional connectivity between dorsal horns below the injury site at 24 h. Findings from this study not only confirm the value of qMT and DTI methods for assessing the myelination state of injured spinal cord but indicate that they may also have further implications on whether therapies targeted towards remyelination may be appropriate. Additionally, a better understanding of changes after SCI provides valuable information to guide and assess interventions.
Collapse
Affiliation(s)
- Tung-Lin Wu
- Vanderbilt University Institute of Imaging Science, Nashville, TN, 37232, United States
- Biomedical Engineering, Vanderbilt University, Nashville, TN, 37232, United States
| | - Nellie E. Byun
- Vanderbilt University Institute of Imaging Science, Nashville, TN, 37232, United States
- Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, 37232, United States
| | - Feng Wang
- Vanderbilt University Institute of Imaging Science, Nashville, TN, 37232, United States
- Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, 37232, United States
| | - Arabinda Mishra
- Vanderbilt University Institute of Imaging Science, Nashville, TN, 37232, United States
- Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, 37232, United States
| | - Vaibhav A. Janve
- Vanderbilt University Institute of Imaging Science, Nashville, TN, 37232, United States
- Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, 37232, United States
| | - Li Min Chen
- Vanderbilt University Institute of Imaging Science, Nashville, TN, 37232, United States
- Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, 37232, United States
| | - John C. Gore
- Vanderbilt University Institute of Imaging Science, Nashville, TN, 37232, United States
- Biomedical Engineering, Vanderbilt University, Nashville, TN, 37232, United States
- Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, 37232, United States
- Physics and Astronomy, Vanderbilt University, Nashville, TN, 37232, United States
- Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, United States
| |
Collapse
|
41
|
Garcia-Diaz B, Baron-Van Evercooren A. Schwann cells: Rescuers of central demyelination. Glia 2020; 68:1945-1956. [PMID: 32027054 DOI: 10.1002/glia.23788] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 01/15/2020] [Accepted: 01/23/2020] [Indexed: 12/31/2022]
Abstract
The presence of peripheral myelinating cells in the central nervous system (CNS) has gained the neurobiologist attention over the years. Despite the confirmed presence of Schwann cells in the CNS in pathological conditions, and the long list of their beneficial effects on central remyelination, the cues that impede or allow Schwann cells to successfully conquer and remyelinate central axons remain partially undiscovered. A better knowledge of these factors stands out as crucial to foresee a rational therapeutic approach for the use of Schwann cells in CNS repair. Here, we review the diverse origins of Schwann cells into the CNS, both peripheral and central, as well as the CNS components that inhibit Schwann survival and migration into the central parenchyma. Namely, we analyze the astrocyte- and the myelin-derived components that restrict Schwann cells into the CNS. Finally, we highlight the unveiled mode of invasion of these peripheral cells through the central environment, using blood vessels as scaffolds to pave their ways toward demyelinated lesions. In short, this review presents the so far uncovered knowledge of this complex CNS-peripheral nervous system (PNS) relationship.
Collapse
Affiliation(s)
- Beatriz Garcia-Diaz
- Unidad de Gestión Clínica de Neurociencias, IBIMA, Hospital Regional Universitario de Málaga, Universidad de Málaga, Málaga, Spain.,Institut du Cerveau et de la Moelle Epinière-Groupe Hospitalier Pitié-Salpêtrière, INSERM, U1127, CNRS, Paris, France.,Sorbonne Universités, Université Pierre et Marie Curie Paris 06, Paris, France
| | - Anne Baron-Van Evercooren
- Institut du Cerveau et de la Moelle Epinière-Groupe Hospitalier Pitié-Salpêtrière, INSERM, U1127, CNRS, Paris, France.,Sorbonne Universités, Université Pierre et Marie Curie Paris 06, Paris, France
| |
Collapse
|
42
|
Abdullahi D, Ahmad Annuar A, Sanusi J. Neuroprotective potential of Spirulina platensis on lesioned spinal cord corticospinal tract under experimental conditions in rat models. Ultrastruct Pathol 2019; 43:273-289. [PMID: 31779507 DOI: 10.1080/01913123.2019.1695693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Spinal cord injury (SCI) results from penetrating or compressive traumatic injury to the spine in humans or by the surgical compression of the spinal cord in experimental animals. In this study, the neuroprotective potential of Spirulina platensis was investigated on ultrastructural and functional recovery of the spinal cord following surgical-induced injury. Twenty-four Sprague-Dawley rats were divided into three groups; sham group, control (trauma) group, and experimental (S. platensis) group (180 mg/kg) of eight rats each. For each group, the rats were then subdivided into two groups to allow measurement at two different timepoints (day 14 and 28) for the microscopic analysis. Rats in the control and experimental S. platensis groups were subjected to partial crush injury at the level of T12 with Inox number 2 modified forceps by compressing on the spinal cord for 30 s. Pairwise comparisons of ultrastructural grading mean scores difference between the control and experimental S. platensis groups reveals that there were significant differences on the axonal ultrastructure, myelin sheath and BBB Score on Day 28; these correlate with the functional locomotor recovery at this timepoint. The results suggest that supplementation with S. platensis induces functional recovery and effective preservation of the spinal cord ultrastructure after SCI. These findings will open new potential avenue for further research into the mechanism of S. platensis-mediated spinal cord repair.
Collapse
Affiliation(s)
- Dauda Abdullahi
- Department of Anatomy, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia.,Department of Human Anatomy, College of Medical Sciences, Abubakar Tafawa Balewa University Bauchi, Bauchi, Nigeria
| | - Azlina Ahmad Annuar
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Junedah Sanusi
- Department of Anatomy, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
43
|
Stierli S, Imperatore V, Lloyd AC. Schwann cell plasticity-roles in tissue homeostasis, regeneration, and disease. Glia 2019; 67:2203-2215. [PMID: 31215712 DOI: 10.1002/glia.23643] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 05/14/2019] [Accepted: 05/14/2019] [Indexed: 12/12/2022]
Abstract
How tissues are maintained over a lifetime and repaired following injury are fundamental questions in biology with a disruption to these processes underlying pathologies such as cancer and degenerative disorders. It is becoming increasingly clear that each tissue has a distinct mechanism to maintain homeostasis and respond to injury utilizing different types of stem/progenitor cell populations depending on the insult and/or with a contribution from more differentiated cells that are able to dedifferentiate to aid tissue regeneration. Peripheral nerves are highly quiescent yet show remarkable regenerative capabilities. Remarkably, there is no evidence for a classical stem cell population, rather all cell-types within the nerve are able to proliferate to produce new nerve tissue. Co-ordinating the regeneration of this tissue are Schwann cells (SCs), the main glial cells of the peripheral nervous system. SCs exist in architecturally stable structures that can persist for the lifetime of an animal, however, they are not postmitotic, in that following injury they are reprogrammed at high efficiency to a progenitor-like state, with these cells acting to orchestrate the nerve regeneration process. During nerve regeneration, SCs show little plasticity, maintaining their identity in the repaired tissue. However, once free of the nerve environment they appear to exhibit increased plasticity with reported roles in the repair of other tissues. In this review, we will discuss the mechanisms underlying the homeostasis and regeneration of peripheral nerves and how reprogrammed progenitor-like SCs have broader roles in the repair of other tissues with implications for pathologies such as cancer.
Collapse
Affiliation(s)
- Salome Stierli
- MRC LMCB, University College London, Gower Street, London, WC1E 6BT, UK
| | | | - Alison C Lloyd
- MRC LMCB, University College London, Gower Street, London, WC1E 6BT, UK
| |
Collapse
|
44
|
Garcia-Diaz B, Bachelin C, Coulpier F, Gerschenfeld G, Deboux C, Zujovic V, Charnay P, Topilko P, Baron-Van Evercooren A. Blood vessels guide Schwann cell migration in the adult demyelinated CNS through Eph/ephrin signaling. Acta Neuropathol 2019; 138:457-476. [PMID: 31011859 PMCID: PMC6689289 DOI: 10.1007/s00401-019-02011-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 03/27/2019] [Accepted: 04/07/2019] [Indexed: 12/30/2022]
Abstract
Schwann cells (SC) enter the central nervous system (CNS) in pathophysiological conditions. However, how SC invade the CNS to remyelinate central axons remains undetermined. We studied SC migratory behavior ex vivo and in vivo after exogenous transplantation in the demyelinated spinal cord. The data highlight for the first time that SC migrate preferentially along blood vessels in perivascular extracellular matrix (ECM), avoiding CNS myelin. We demonstrate in vitro and in vivo that this migration route occurs by virtue of a dual mode of action of Eph/ephrin signaling. Indeed, EphrinB3, enriched in myelin, interacts with SC Eph receptors, to drive SC away from CNS myelin, and triggers their preferential adhesion to ECM components, such as fibronectin via integrinβ1 interactions. This complex interplay enhances SC migration along the blood vessel network and together with lesion-induced vascular remodeling facilitates their timely invasion of the lesion site. These novel findings elucidate the mechanism by which SC invade and contribute to spinal cord repair.
Collapse
|
45
|
Pukos N, Goodus MT, Sahinkaya FR, McTigue DM. Myelin status and oligodendrocyte lineage cells over time after spinal cord injury: What do we know and what still needs to be unwrapped? Glia 2019; 67:2178-2202. [PMID: 31444938 DOI: 10.1002/glia.23702] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 07/29/2019] [Accepted: 07/30/2019] [Indexed: 01/04/2023]
Abstract
Spinal cord injury (SCI) affects over 17,000 individuals in the United States per year, resulting in sudden motor, sensory and autonomic impairments below the level of injury. These deficits may be due at least in part to the loss of oligodendrocytes and demyelination of spared axons as it leads to slowed or blocked conduction through the lesion site. It has long been accepted that progenitor cells form new oligodendrocytes after SCI, resulting in the acute formation of new myelin on demyelinated axons. However, the chronicity of demyelination and the functional significance of remyelination remain contentious. Here we review work examining demyelination and remyelination after SCI as well as the current understanding of oligodendrocyte lineage cell responses to spinal trauma, including the surprisingly long-lasting response of NG2+ oligodendrocyte progenitor cells (OPCs) to proliferate and differentiate into new myelinating oligodendrocytes for months after SCI. OPCs are highly sensitive to microenvironmental changes, and therefore respond to the ever-changing post-SCI milieu, including influx of blood, monocytes and neutrophils; activation of microglia and macrophages; changes in cytokines, chemokines and growth factors such as ciliary neurotrophic factor and fibroblast growth factor-2; glutamate excitotoxicity; and axon degeneration and sprouting. We discuss how these changes relate to spontaneous oligodendrogenesis and remyelination, the evidence for and against demyelination being an important clinical problem and if remyelination contributes to motor recovery.
Collapse
Affiliation(s)
- Nicole Pukos
- Neuroscience Graduate Program, Ohio State University, Columbus, Ohio.,Belford Center for Spinal Cord Injury, Ohio State University, Columbus, Ohio
| | - Matthew T Goodus
- Belford Center for Spinal Cord Injury, Ohio State University, Columbus, Ohio.,Department of Neuroscience, Wexner Medical Center, Ohio State University, Columbus, Ohio
| | - Fatma R Sahinkaya
- Neuroscience Graduate Program, Ohio State University, Columbus, Ohio
| | - Dana M McTigue
- Belford Center for Spinal Cord Injury, Ohio State University, Columbus, Ohio.,Department of Neuroscience, Wexner Medical Center, Ohio State University, Columbus, Ohio
| |
Collapse
|
46
|
Duncan GJ, Manesh SB, Hilton BJ, Assinck P, Plemel JR, Tetzlaff W. The fate and function of oligodendrocyte progenitor cells after traumatic spinal cord injury. Glia 2019; 68:227-245. [PMID: 31433109 DOI: 10.1002/glia.23706] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 07/24/2019] [Accepted: 08/01/2019] [Indexed: 12/27/2022]
Abstract
Oligodendrocyte progenitor cells (OPCs) are the most proliferative and dispersed population of progenitor cells in the adult central nervous system, which allows these cells to rapidly respond to damage. Oligodendrocytes and myelin are lost after traumatic spinal cord injury (SCI), compromising efficient conduction and, potentially, the long-term health of axons. In response, OPCs proliferate and then differentiate into new oligodendrocytes and Schwann cells to remyelinate axons. This culminates in highly efficient remyelination following experimental SCI in which nearly all intact demyelinated axons are remyelinated in rodent models. However, myelin regeneration comprises only one role of OPCs following SCI. OPCs contribute to scar formation after SCI and restrict the regeneration of injured axons. Moreover, OPCs alter their gene expression following demyelination, express cytokines and perpetuate the immune response. Here, we review the functional contribution of myelin regeneration and other recently uncovered roles of OPCs and their progeny to repair following SCI.
Collapse
Affiliation(s)
- Greg J Duncan
- Department of Neurology, Jungers Center for Neurosciences Research, Oregon Health and Science University, Portland, Oregon
| | - Sohrab B Manesh
- Graduate Program in Neuroscience, International Collaboration on Repair Discoveries (ICORD), University of British Columbia (UBC), Vancouver, British Columbia, Canada
| | - Brett J Hilton
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Bonn, Germany
| | - Peggy Assinck
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
| | - Jason R Plemel
- Department of Medicine, Division of Neurology, Neuroscience and Mental Health Institute, University of Alberta, Calgary, Alberta, Canada
| | - Wolfram Tetzlaff
- Graduate Program in Neuroscience, International Collaboration on Repair Discoveries (ICORD), University of British Columbia (UBC), Vancouver, British Columbia, Canada.,Departments of Zoology and Surgery, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
47
|
Spatiotemporal trajectories of quantitative magnetization transfer measurements in injured spinal cord using simplified acquisitions. NEUROIMAGE-CLINICAL 2019; 23:101921. [PMID: 31491830 PMCID: PMC6639592 DOI: 10.1016/j.nicl.2019.101921] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 06/04/2019] [Accepted: 06/30/2019] [Indexed: 12/19/2022]
Abstract
Purpose This study aims to systematically evaluate the accuracy and precision of pool size ratio (PSR) measurements from quantitative magnetization transfer (qMT) acquisitions using simplified models in the context of assessing injury-associated spatiotemporal changes in spinal cords of non-human primates. This study also aims to characterize changes in the spinal tissue pathology in individual subjects, both regionally and longitudinally, in order to demonstrate the relationship between regional tissue compositional changes and sensorimotor behavioral recovery after cervical spinal cord injury (SCI). Methods MRI scans were recorded on anesthetized monkeys at 9.4 T, before and serially after a unilateral section of the dorsal column tract. Images were acquired following saturating RF pulses at different offset frequencies. Models incorporating two pools of protons but with differing numbers of variable parameters were used to fit the data to derive qMT parameters. The results using different amounts of measured data and assuming different numbers of variable model parameters were compared. Behavioral impairments and recovery were assessed by a food grasping-retrieving task. Histological sections were obtained post mortem for validation of the injury. Results QMT fitting provided maps of pool size ratio (PSR), the relative amounts of immobilized protons exchanging magnetization compared to the “free” water. All the selected modeling approaches detected a lesion/cyst at the site of injury as significant reductions in PSR values. The regional contrasts in the PSR maps obtained using the different fittings varied, but the 2-parameter fitting results showed strong positive correlations with results from 5-parameter modeling. 2-parameter fitting results with modest (>3) RF offsets showed comparable sensitivity for detecting demyelination in white matter and loss of macromolecules in gray matter around lesion sites compared to 5-parameter fitting with fully-sampled data acquisitions. Histology confirmed that decreases of PSR corresponded to regional demyelination around lesion sites, especially when demyelination occurred along the dorsal column on the injury side. Longitudinally, PSR values of injured dorsal column tract and gray matter horns exhibited remarkable recovery that associated with behavioral improvement. Conclusion Simplified qMT modeling approaches provide efficient and sensitive means to detect and characterize injury-associated demyelination in white matter tracts and loss of macromolecules in gray matter and to monitor its recovery over time. Simplified 2-parameter and fully sampled 5-parameter qMT modeling achieved comparable accuracy and precision of PSR values. Successfully tracked and differentiated myelination states of specific WM tracts and macromolecular changes in GM horns. Recovery of WM and GM pathology assessed by qMT correlated with improvements in hand uses after injury. High translational potential for clinical studies of human patients with spinal cord injury.
Collapse
|
48
|
Transplantation of miR‐219 overexpressed human endometrial stem cells encapsulated in fibrin hydrogel in spinal cord injury. J Cell Physiol 2019; 234:18887-18896. [DOI: 10.1002/jcp.28527] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 02/09/2019] [Accepted: 02/14/2019] [Indexed: 12/31/2022]
|
49
|
Zhang Q, Shi B, Ding J, Yan L, Thawani JP, Fu C, Chen X. Polymer scaffolds facilitate spinal cord injury repair. Acta Biomater 2019; 88:57-77. [PMID: 30710714 DOI: 10.1016/j.actbio.2019.01.056] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 01/10/2019] [Accepted: 01/28/2019] [Indexed: 12/23/2022]
Abstract
During the past decades, improving patient neurological recovery following spinal cord injury (SCI) has remained a challenge. An effective treatment for SCI would not only reduce fractured elements and isolate developing local glial scars to promote axonal regeneration but also ameliorate secondary effects, including inflammation, apoptosis, and necrosis. Three-dimensional (3D) scaffolds provide a platform in which these mechanisms can be addressed in a controlled manner. Polymer scaffolds with favorable biocompatibility and appropriate mechanical properties have been engineered to minimize cicatrization, customize drug release, and ensure an unobstructed space to promote cell growth and differentiation. These properties make polymer scaffolds an important potential therapeutic platform. This review highlights the recent developments in polymer scaffolds for SCI engineering. STATEMENT OF SIGNIFICANCE: How to improve the efficacy of neurological recovery after spinal cord injury (SCI) is always a challenge. Tissue engineering provides a promising strategy for SCI repair, and scaffolds are one of the most important elements in addition to cells and inducing factors. The review highlights recent development and future prospects in polymer scaffolds for SCI therapy. The review will guide future studies by outlining the requirements and characteristics of polymer scaffold technologies employed against SCI. Additionally, the peculiar properties of polymer materials used in the therapeutic process of SCI also have guiding significance to other tissue engineering approaches.
Collapse
|
50
|
Alizadeh A, Dyck SM, Karimi-Abdolrezaee S. Traumatic Spinal Cord Injury: An Overview of Pathophysiology, Models and Acute Injury Mechanisms. Front Neurol 2019; 10:282. [PMID: 30967837 PMCID: PMC6439316 DOI: 10.3389/fneur.2019.00282] [Citation(s) in RCA: 635] [Impact Index Per Article: 127.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 03/05/2019] [Indexed: 12/11/2022] Open
Abstract
Traumatic spinal cord injury (SCI) is a life changing neurological condition with substantial socioeconomic implications for patients and their care-givers. Recent advances in medical management of SCI has significantly improved diagnosis, stabilization, survival rate and well-being of SCI patients. However, there has been small progress on treatment options for improving the neurological outcomes of SCI patients. This incremental success mainly reflects the complexity of SCI pathophysiology and the diverse biochemical and physiological changes that occur in the injured spinal cord. Therefore, in the past few decades, considerable efforts have been made by SCI researchers to elucidate the pathophysiology of SCI and unravel the underlying cellular and molecular mechanisms of tissue degeneration and repair in the injured spinal cord. To this end, a number of preclinical animal and injury models have been developed to more closely recapitulate the primary and secondary injury processes of SCI. In this review, we will provide a comprehensive overview of the recent advances in our understanding of the pathophysiology of SCI. We will also discuss the neurological outcomes of human SCI and the available experimental model systems that have been employed to identify SCI mechanisms and develop therapeutic strategies for this condition.
Collapse
Affiliation(s)
- Arsalan Alizadeh
- Regenerative Medicine Program, Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, Spinal Cord Research Center, University of Manitoba, Winnipeg, MB, Canada
| | - Scott Matthew Dyck
- Regenerative Medicine Program, Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, Spinal Cord Research Center, University of Manitoba, Winnipeg, MB, Canada
| | - Soheila Karimi-Abdolrezaee
- Regenerative Medicine Program, Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, Spinal Cord Research Center, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|