1
|
Chen RY, Lee KZ. Therapeutic Efficacy of Hemodynamic Management Using Norepinephrine on Cardiorespiratory Function Following Cervical Spinal Cord Contusion in Rats. J Neurotrauma 2024. [PMID: 39661956 DOI: 10.1089/neu.2024.0342] [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: 12/13/2024] Open
Abstract
Cervical spinal cord injury usually leads to cardiorespiratory dysfunction due to interruptions of the supraspinal pathways innervating the phrenic motoneurons and thoracic sympathetic preganglionic neurons. Although clinical guidelines recommend maintaining the mean arterial pressure within 85-90 mmHg during the first week of injury, there is no pre-clinical evidence from animal models to prove the therapeutic efficacy of hemodynamic management. Accordingly, the present study was designed to investigate the therapeutic efficacy of hemodynamic management in rats with cervical spinal cord contusion. Adult male rats underwent cervical spinal cord contusion and the implantation of osmotic pumps filled with saline or norepinephrine (NE) (125 μg/(kg·h) for 1 week). The cardiorespiratory function of unanesthetized rats was examined using a non-invasive blood pressure analyzer and double-chamber plethysmography. Cervical spinal cord contusion caused a long-term reduction in the mean arterial pressure and tidal volume. This hypotensive response was significantly reversed in contused rats receiving NE (1 day: 88 ± 19 mmHg; 2 weeks: 96 ± 13 mmHg) compared with contused rats receiving saline (1 day: 72 ± 15 mmHg; 2 weeks: 82 ± 10 mmHg). NE also significantly improved the tidal volume 1 day post-injury (contused + NE: 0.7 ± 0.2 mL; contused + saline: 0.5 ± 0.1 mL). Immunofluorescence staining results revealed that injury-induced reductions of noradrenergic and glutamatergic fibers within the thoracic spinal cord were significantly improved by NE. These results provided the evidence demonstrating that hemodynamic management using NE significantly improves cardiorespiratory function by alleviating neural pathway damage after cervical spinal cord contusion.
Collapse
Affiliation(s)
- Rui-Yi Chen
- Department of Biological Sciences, College of Science, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Kun-Ze Lee
- Department of Biological Sciences, College of Science, National Sun Yat-sen University, Kaohsiung, Taiwan
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
2
|
Sen S, Parihar N, Patil PM, Upadhyayula SM, Pemmaraju DB. Revisiting the Emerging Role of Light-Based Therapies in the Management of Spinal Cord Injuries. Mol Neurobiol 2024:10.1007/s12035-024-04658-8. [PMID: 39658774 DOI: 10.1007/s12035-024-04658-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Accepted: 12/02/2024] [Indexed: 12/12/2024]
Abstract
The surge in spinal cord injuries (SCI) attracted many neurobiologists to explore the underlying complex pathophysiology and to offer better therapeutic outcomes. The multimodal approaches to therapy in SCI have proven to be effective but to a limited extent. The clinical basics involve invasive procedures and limited therapeutic interventions, and most preclinical studies and formulations are yet to be translated due to numerous factors. In recent years, photobiomodulation therapy (PBMT) has found many applications in various medical fields. In most PBMT, studies on SCI have employed laser sources in experimental animal models as a non-invasive source. PBMT has been applied in numerous facets of SCI pathophysiology, especially attenuation of neuroinflammatory cascades, enhanced neuronal regeneration, reduced apoptosis and gliosis, and increased behavioral recovery within a short span. Although PBMT is specific in modulating mitochondrial bioenergetics, innumerous molecular pathways such as JAK-STAT, PI3K-AKT, NF-κB, MAPK, JNK/TLR/MYD88, ERK/CREB, TGF-β/SMAD, GSK3β-AKT-β-catenin, and AMPK/PGC-1α/TFAM signaling pathways have been or are yet to be exploited. PMBT has been effective not only in cell-specific actions in SCI such as astrocyte activation or microglial polarization or alterations in neuronal pathology but also modulated overall pathobiology in SCI animals such as rapid behavioral recovery. The goal of this review is to summarize research that has used PBMT for various models of SCI in different animals, including clarifying its mechanisms and prospective molecular pathways that may be utilized for better therapeutic outcomes.
Collapse
Affiliation(s)
- Santimoy Sen
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, Assam, India
| | - Nidhi Parihar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, Assam, India
| | - Prathamesh Mahadev Patil
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, Assam, India
| | - Suryanarayana Murty Upadhyayula
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, Assam, India
| | - Deepak B Pemmaraju
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, Assam, India.
| |
Collapse
|
3
|
Lancaster MA. Pluripotent stem cell-derived organoids: A brief history of curiosity-led discoveries. Bioessays 2024; 46:e2400105. [PMID: 39101295 PMCID: PMC11589667 DOI: 10.1002/bies.202400105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/27/2024] [Accepted: 07/01/2024] [Indexed: 08/06/2024]
Abstract
Organoids are quickly becoming an accepted model for understanding human biology and disease. Pluripotent stem cells (PSC) provide a starting point for many organs and enable modeling of the embryonic development and maturation of such organs. The foundation of PSC-derived organoids can be found in elegant developmental studies demonstrating the remarkable ability of immature cells to undergo histogenesis even when taken out of the embryo context. PSC-organoids are an evolution of earlier methods such as embryoid bodies, taken to a new level with finer control and in some cases going beyond tissue histogenesis to organ-like morphogenesis. But many of the discoveries that led to organoids were not necessarily planned, but rather the result of inquisitive minds with freedom to explore. Protecting such curiosity-led research through flexible funding will be important going forward if we are to see further ground-breaking discoveries.
Collapse
|
4
|
Lee KZ, Liu TT, Chen RY. Therapeutic efficacy of adrenergic agents on systemic and spinal hemodynamics in an acute cervical spinal cord injury rodent model. Spine J 2024; 24:1964-1980. [PMID: 38679076 DOI: 10.1016/j.spinee.2024.04.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 04/19/2024] [Accepted: 04/23/2024] [Indexed: 05/01/2024]
Abstract
BACKGROUND Cervical spinal cord injury usually results in cardiorespiratory dysfunctions due to interruptions of the bulbospinal pathways innervating the cervical phrenic motoneurons and thoracic sympathetic preganglionic neurons. PURPOSE The present study aimed to evaluate the therapeutic effects of adrenergic agents on systemic and spinal hemodynamics during acute cervical spinal cord injury. STUDY DESIGN In vivo animal study. METHODS The cardiorespiratory function and spinal cord blood flow and oxygenation level were monitored in response to cervical spinal cord contusion and intravenous infusion of three types of adrenergic agents (phenylephrine, dobutamine, and norepinephrine). RESULTS Cervical spinal cord contusion resulted in immediate reduction of respiratory airflow, arterial blood pressure, and spinal cord blood flow. The arterial blood pressure and spinal cord blood flow remained lower than the preinjury value in contused animals infused with saline at 60 min postinjury. Infusion of phenylephrine (500, 1000, and 2000 μg/kg) and norepinephrine (125, 250, and 500 μg/kg) significantly increased the arterial blood pressure, while only norepinephrine augmented the spinal cord blood flow. Conversely, dobutamine (1000 and 2000 μg/kg) reduced both arterial blood pressure and spinal cord blood flow. Notably, administration of adrenergic agents tended to increase spinal cord hemorrhage in contused animals. CONCLUSIONS Infusion of norepinephrine can effectively maintain the blood pressure and improve spinal cord blood flow during acute spinal cord injury. CLINICAL SIGNIFICANCE Norepinephrine may be a superior medicine for hemodynamic management; however, the potential hemorrhage should be considered when utilizing the vasopressor to regulate systemic and spinal hemodynamics at the acute injured stage.
Collapse
Affiliation(s)
- Kun-Ze Lee
- Department of Biological Sciences, National Sun Yat-sen University, No. 70, Lien-Hai Rd., Kaohsiung city 804, Taiwan; Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, No. 100, Shih-Chuan 1st Rd., Kaohsiung city 807, Taiwan.
| | - Tzu-Ting Liu
- Department of Biological Sciences, National Sun Yat-sen University, No. 70, Lien-Hai Rd., Kaohsiung city 804, Taiwan
| | - Rui-Yi Chen
- Department of Biological Sciences, National Sun Yat-sen University, No. 70, Lien-Hai Rd., Kaohsiung city 804, Taiwan
| |
Collapse
|
5
|
Li Q, Sandoval A, Moth J, Shang J, Liew JY, Dunn T, Yang Z, Su J, Henwood M, Williams P, Chen B. Reduction of prolonged excitatory neuron swelling after spinal cord injury improves locomotor recovery in mice. Sci Transl Med 2024; 16:eadn7095. [PMID: 39321270 DOI: 10.1126/scitranslmed.adn7095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 04/09/2024] [Accepted: 09/05/2024] [Indexed: 09/27/2024]
Abstract
Spinal cord injury (SCI) results in acute damage and triggers secondary injury responses with sustained neuronal loss and dysfunction. However, the underlying mechanisms for these delayed neuronal pathologies are not entirely understood. SCI results in the swelling of spinal neurons, but the contribution of cell swelling to neuronal loss and functional deficits after SCI has not been systematically characterized. In this study, we devised a three-dimensional image analysis pipeline to evaluate spinal neurons, examining their types, quantities, volumes, and spatial distribution in a double-lateral hemisection SCI mouse model. We found that both excitatory and inhibitory neurons swell and are lost, albeit with distinct temporal patterns. Inhibitory neurons demonstrated marked swelling and decline in number on day 2 after SCI, which resolved by day 14. In contrast, excitatory neurons maintained persistent swelling and continued cell loss for at least 35 days after SCI in mice. Excitatory neurons exhibited sustained expression of the Na+-K+-Cl- cotransporter 1 (NKCC1), whereas inhibitory neurons down-regulated the protein by day 14 after SCI. Treatment with a Food and Drug Administration-approved NKCC1 inhibitor, bumetanide, mitigated swelling of excitatory neurons and reduced their loss in the secondary injury phase after SCI. The administration of bumetanide after SCI in mouse improved locomotor recovery, with functional benefits persisting for at least 4 weeks after treatment cessation. This study advances our understanding of SCI-related pathology and introduces bumetanide as a potential treatment to mitigate sustained neuronal swelling and enhance recovery after SCI.
Collapse
Affiliation(s)
- Qiang Li
- Department of Neurobiology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Alfredo Sandoval
- Department of Neurobiology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - John Moth
- Department of Anesthesiology, Houston Methodist Hospital, Houston, TX 77030, USA
| | - Junkui Shang
- Department of Neurobiology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Jia Yi Liew
- Department of Neurobiology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Tiffany Dunn
- Department of Neurobiology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Zhiyun Yang
- F. M. Kirby Neurobiology Center, Boston Children's Hospital and Departments of Neurology and Ophthalmology, Harvard Medical School, Boston, MA 02115, USA
| | - Junfeng Su
- F. M. Kirby Neurobiology Center, Boston Children's Hospital and Departments of Neurology and Ophthalmology, Harvard Medical School, Boston, MA 02115, USA
| | - Melissa Henwood
- Department of Neurobiology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Philip Williams
- Department of Ophthalmology and Visual Sciences and Department of Neuroscience, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Bo Chen
- Department of Neurobiology, University of Texas Medical Branch, Galveston, TX 77555, USA
| |
Collapse
|
6
|
Gu J, Cai X, Raza F, Zafar H, Chu B, Yuan H, Wang T, Wang J, Feng X. Preparation of a minocycline polymer micelle thermosensitive gel and its application in spinal cord injury. NANOSCALE ADVANCES 2024:d4na00625a. [PMID: 39355839 PMCID: PMC11440374 DOI: 10.1039/d4na00625a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 09/15/2024] [Indexed: 10/03/2024]
Abstract
Neuroprotection is an important approach for the treatment of spinal cord injury (SCI). Minocycline (MC), a known neuroprotective agent, has been utilized for SCI treatment, but its therapeutic effect is limited by instability and low bioavailability. Herein, we developed an innovative micellar thermosensitive hydrogel (MCPP-M-gel) that encapsulates MC in polyethylene glycol (PEG)-poly(lactide-co-glycolic acid) (PLGA) micelles to enhance its therapeutic efficacy in a rat model of SCI. The micelles were synthesized via the thin-film hydration method and characterized for encapsulation efficiency, particle size, zeta potential, and polydispersity index (PDI). MCPP-M-gel demonstrated favorable physico-mechanical properties and extended MC release over 72 hours in vitro without cytotoxic effects on neural crest-derived ectoderm mesenchymal stem cells (EMSCs). Thereafter, MC, MCPP-M, MCPP-M-gel and a blank micellar thermosensitive gel were injected into the injured site of SCI rats. Histopathological evaluation demonstrated that MCPP-M-gel could promote neuronal regeneration at the injured site of the SC after 28 days. Immunofluorescence techniques revealed that MCPP-M-gel increased the expression of neuronal class III β-tubulin (Tuj1), myelin basic protein (MBP), growth-associated protein 43 (GAP43), neurofilament protein-200 (NF-200) and nestin as well as reduced glial-fibrillary acidic protein (GFAP) expression in damaged areas of the SC. In conclusion, this study innovatively developed MCPP-M-gel based on a PEG-PLGA copolymer as a biomaterial, laying a solid foundation for further research and application of MCPP-M-gel in SCI models or other neurodegenerative diseases.
Collapse
Affiliation(s)
- Jun Gu
- School of Medicine, Yangzhou University Yangzhou Jiangsu 225009 China
- Department of Orthopedics, Xishan People's Hospital Wuxi Jiangsu 204105 China
| | - Xiaohu Cai
- School of Medicine, Yangzhou University Yangzhou Jiangsu 225009 China
- Department of Rehabilitation, Xishan People's Hospital Wuxi Jiangsu 204105 China
| | - Faisal Raza
- School of Pharmacy, Shanghai Jiao Tong University Shanghai 200240 China
| | - Hajra Zafar
- School of Pharmacy, Shanghai Jiao Tong University Shanghai 200240 China
| | - Bo Chu
- Department of Orthopedics, Xishan People's Hospital Wuxi Jiangsu 204105 China
| | - Haitao Yuan
- Department of Orthopedics, Xishan People's Hospital Wuxi Jiangsu 204105 China
| | - Tianqi Wang
- Department of Orthopedics, Xishan People's Hospital Wuxi Jiangsu 204105 China
| | - Jiapeng Wang
- School of Pharmacy, Jiangsu University Zhenjiang Jiangsu 212013 China
| | - Xiaojun Feng
- School of Medicine, Yangzhou University Yangzhou Jiangsu 225009 China
- Department of Orthopedics, Xishan People's Hospital Wuxi Jiangsu 204105 China
| |
Collapse
|
7
|
Jiang F, Joshi H, Badhiwala JH, Wilson JRF, Lenke LG, Shaffrey CI, Cheung KMC, Carreon LY, Dekutoski MB, Schwab FJ, Boachie-Adjei O, Kebaish KM, Ames CP, Berven SH, Qiu Y, Matsuyama Y, Dahl BT, Mehdian H, Pellisé F, Lewis SJ, Fehlings MG. Spinal cord injury in high-risk complex adult spinal deformity surgery: review of incidence and outcomes from the Scoli-RISK-1 study. Spinal Cord Ser Cases 2024; 10:59. [PMID: 39153987 PMCID: PMC11330517 DOI: 10.1038/s41394-024-00673-y] [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: 07/18/2024] [Revised: 07/29/2024] [Accepted: 08/02/2024] [Indexed: 08/19/2024] Open
Abstract
STUDY DESIGN Clinical case series. OBJECTIVE To describe the cause, treatment and outcome of 6 cases of perioperative spinal cord injury (SCI) in high-risk adult deformity surgery. SETTING Adult spinal deformity patients were enrolled in the multi-center Scoli-RISK-1 cohort study. METHODS A total of 272 patients who underwent complex adult deformity surgery were enrolled in the prospective, multi-center Scoli-RISK-1 cohort study. Clinical follow up data were available up to a maximum of 2 years after index surgery. Cases of perioperative SCI were identified and an extensive case review was performed. RESULTS Six individuals with SCI were identified from the Scoli-RISK-1 database (2.2%). Two cases occurred intraoperatively and four cases occurred postoperatively. The first case was an incomplete SCI due to a direct intraoperative insult and was treated postoperatively with Riluzole. The second SCI case was caused by a compression injury due to overcorrection of the deformity. Three cases of incomplete SCI occurred; one case of postoperative hematoma, one case of proximal junctional kyphosis (PJK) and one case of adjacent segment disc herniation. All cases of post-operative incomplete SCI were managed with revision decompression and resulted in excellent clinical recovery. One case of incomplete SCI resulted from infection and PJK. The patient's treatment was complicated by a delay in revision and the patient suffered persistent neurological deficits up to six weeks following the onset of SCI. CONCLUSION Despite the low incidence in high-risk adult deformity surgeries, perioperative SCI can result in devastating consequences. Thus, appropriate postoperative care, follow up and timely management of SCI are essential.
Collapse
Affiliation(s)
- Fan Jiang
- Department of Surgery and Spine Program, University of Toronto, Toronto, ON, Canada
| | - Hetshree Joshi
- Department of Surgery and Spine Program, University of Toronto, Toronto, ON, Canada
| | - Jetan H Badhiwala
- Department of Surgery and Spine Program, University of Toronto, Toronto, ON, Canada
| | - Jamie R F Wilson
- Department of Surgery and Spine Program, University of Toronto, Toronto, ON, Canada
- Department of Neurosurgery, University of Nebraska Medical Center, Omaha, NE, USA
| | - Lawrence G Lenke
- Department of Orthopedic Surgery, The Spine Hospital, Columbia University Medical Center, New York, NY, USA
| | | | - Kenneth M C Cheung
- Department of Orthopaedics and Traumatology, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | | | | | - Frank J Schwab
- Spine Service, Hospital for Special Surgery, New York, NY, USA
| | | | - Khaled M Kebaish
- Department of Orthopaedic Surgery, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Christopher P Ames
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Sigurd H Berven
- Department of Orthopaedic Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Yong Qiu
- Spine Surgery, Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Yukihiro Matsuyama
- Department of Orthopedic Surgery, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Benny T Dahl
- Division of Orthopedic Surgery, Texas Children's Hospital, Baylor College of Medicine, Houston & Rigshospitalet, National University of Denmark, Copenhagen, Denmark
| | - Hossein Mehdian
- The Centre for Spinal Studies and Surgery, Queen's Medical Centre, Nottingham University Hospitals, Nottingham, UK
| | - Ferran Pellisé
- Hospital Universitari de la Vall d'Hebron, Barcelona, Spain
| | - Stephen J Lewis
- Department of Surgery and Spine Program, University of Toronto, Toronto, ON, Canada
| | - Michael G Fehlings
- Department of Surgery and Spine Program, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
8
|
Saraswat Ohri S, Forston MD, Myers SA, Brown BL, Andres KR, Howard RM, Gao Y, Liu Y, Cavener DR, Hetman M, Whittemore SR. Oligodendrocyte-selective deletion of the eIF2α kinase Perk/Eif2ak3 limits functional recovery after spinal cord injury. Glia 2024; 72:1259-1272. [PMID: 38587137 DOI: 10.1002/glia.24525] [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: 02/01/2023] [Revised: 02/13/2024] [Accepted: 03/04/2024] [Indexed: 04/09/2024]
Abstract
After spinal cord injury (SCI), re-establishing cellular homeostasis is critical to optimize functional recovery. Central to that response is PERK signaling, which ultimately initiates a pro-apoptotic response if cellular homeostasis cannot be restored. Oligodendrocyte (OL) loss and white matter damage drive functional consequences and determine recovery potential after thoracic contusive SCI. We examined acute (<48 h post-SCI) and chronic (6 weeks post-SCI) effects of conditionally deleting Perk from OLs prior to SCI. While Perk transcript is expressed in many types of cells in the adult spinal cord, its levels are disproportionately high in OL lineage cells. Deletion of OL-Perk prior to SCI resulted in: (1) enhanced acute phosphorylation of eIF2α, a major PERK substrate and the critical mediator of the integrated stress response (ISR), (2) enhanced acute expression of the downstream ISR genes Atf4, Ddit3/Chop, and Tnfrsf10b/Dr5, (3) reduced acute OL lineage-specific Olig2 mRNA, but not neuronal or astrocytic mRNAs, (4) chronically decreased OL content in the spared white matter at the injury epicenter, (5) impaired hindlimb locomotor recovery, and (6) reduced chronic epicenter white matter sparing. Cultured primary OL precursor cells with reduced PERK expression and activated ER stress response showed: (1) unaffected phosphorylation of eIF2α, (2) enhanced ISR gene induction, and (3) increased cytotoxicity. Therefore, OL-Perk deficiency exacerbates ISR signaling and potentiates white matter damage after SCI. The latter effect is likely mediated by increased loss of Perk-/- OLs.
Collapse
Affiliation(s)
- Sujata Saraswat Ohri
- Kentucky Spinal Cord Injury Research Center, Louisville, Kentucky, USA
- Department of Neurological Surgery, University of Louisville School of Medicine, Louisville, Kentucky, USA
- Department of Interdisciplinary Program in Translational Neuroscience, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Michael D Forston
- Kentucky Spinal Cord Injury Research Center, Louisville, Kentucky, USA
- Department of Anatomical Sciences & Neurobiology, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Scott A Myers
- Kentucky Spinal Cord Injury Research Center, Louisville, Kentucky, USA
| | - Brandon L Brown
- Kentucky Spinal Cord Injury Research Center, Louisville, Kentucky, USA
- Department of Interdisciplinary Program in Translational Neuroscience, University of Louisville School of Medicine, Louisville, Kentucky, USA
- Department of Anatomical Sciences & Neurobiology, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Kariena R Andres
- Kentucky Spinal Cord Injury Research Center, Louisville, Kentucky, USA
| | - Russell M Howard
- Kentucky Spinal Cord Injury Research Center, Louisville, Kentucky, USA
| | - Yonglin Gao
- Kentucky Spinal Cord Injury Research Center, Louisville, Kentucky, USA
| | - Yu Liu
- Kentucky Spinal Cord Injury Research Center, Louisville, Kentucky, USA
| | - Douglas R Cavener
- Department of Biology, Penn State University, University Park, Pennsylvania, USA
| | - Michal Hetman
- Kentucky Spinal Cord Injury Research Center, Louisville, Kentucky, USA
- Department of Neurological Surgery, University of Louisville School of Medicine, Louisville, Kentucky, USA
- Department of Interdisciplinary Program in Translational Neuroscience, University of Louisville School of Medicine, Louisville, Kentucky, USA
- Department of Anatomical Sciences & Neurobiology, University of Louisville School of Medicine, Louisville, Kentucky, USA
- Department of Pharmacology & Toxicology, University of Louisville School of Medicine, Louisville, Kentucky, USA
- M.D./Ph.D. Program, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Scott R Whittemore
- Kentucky Spinal Cord Injury Research Center, Louisville, Kentucky, USA
- Department of Neurological Surgery, University of Louisville School of Medicine, Louisville, Kentucky, USA
- Department of Interdisciplinary Program in Translational Neuroscience, University of Louisville School of Medicine, Louisville, Kentucky, USA
- Department of Anatomical Sciences & Neurobiology, University of Louisville School of Medicine, Louisville, Kentucky, USA
- Department of Pharmacology & Toxicology, University of Louisville School of Medicine, Louisville, Kentucky, USA
- M.D./Ph.D. Program, University of Louisville School of Medicine, Louisville, Kentucky, USA
| |
Collapse
|
9
|
Ko CC, Lee PH, Lee JS, Lee KZ. Spinal decompression surgery may alleviate vasopressor-induced spinal hemorrhage and extravasation during acute cervical spinal cord injury in rats. Spine J 2024; 24:519-533. [PMID: 37793474 DOI: 10.1016/j.spinee.2023.09.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 09/19/2023] [Accepted: 09/26/2023] [Indexed: 10/06/2023]
Abstract
BACKGROUND Cervical spinal injury often disrupts the supraspinal vasomotor pathways projecting to the thoracic sympathetic preganglionic neurons, leading to cardiovascular dysfunction. The current guideline is to maintain the mean arterial blood pressure at 85 to 90 mmHg using a vasopressor during the first week of the injury. Some studies have demonstrated that this treatment might be beneficial to alleviate secondary injury and improve neurological outcomes; however, elevation of blood pressure may exacerbate spinal hemorrhage, extravasation, and edema, exacerbating the initial injury. PURPOSE The present study was designed to (1) examine whether vasopressor administration exacerbates spinal hemorrhage and extravasation; (2) evaluate whether spinal decompression surgery relieves vasopressor-induced spinal hemorrhage and extravasation. STUDY DESIGN In vivo animal study. METHODS Animals received a saline solution or a vasopressor (phenylephrine hydrochloride, 500 or 1000 μg/kg, 7 mL/kg/h) after mid-cervical contusion with or without spinal decompression (ie, incision of the dura and arachnoid mater). Spinal cord hemorrhage and extravasation were examined by expression of Evans blue within the spinal cord section. RESULTS The results demonstrated that cervical spinal contusion significantly reduced the mean arterial blood pressure and induced spinal hemorrhage and extravasation. Phenylephrine infusion significantly elevated the mean arterial blood pressure to the preinjury level within 15 to 60 minutes postcontusion; however, spinal hemorrhage and extravasation were more extensive in animals that received phenylephrine than in those that received saline. Notably, spinal decompression mitigated spinal hemorrhage and extravasation in contused rats who received phenylephrine. CONCLUSIONS These data indicate that, although phenylephrine can prevent hypotension after cervical spinal injury, it also causes excess spinal hemorrhage and extravasation. CLINICAL SIGNIFICANCE Spinal decompressive surgery seemed to minimize the side effect of phenylephrine as vasopressor treatment during acute spinal cord injury.
Collapse
Affiliation(s)
- Chia-Chen Ko
- Department of Biological Sciences, National Sun Yat-sen University, No. 70, Lien-Hai Rd., Kaohsiung city 804, Taiwan
| | - Po-Hsuan Lee
- Division of Neurosurgery, Department of Surgery, National Cheng Kung University Hospital, No. 138, Sheng-Li Rd., Tainan city 704, Taiwan
| | - Jung-Shun Lee
- Division of Neurosurgery, Department of Surgery, National Cheng Kung University Hospital, No. 138, Sheng-Li Rd., Tainan city 704, Taiwan; Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, No.1, University Rd., Tainan city 701, Taiwan; Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, No.1, University Rd., Tainan city 701, Taiwan
| | - Kun-Ze Lee
- Department of Biological Sciences, National Sun Yat-sen University, No. 70, Lien-Hai Rd., Kaohsiung city 804, Taiwan; Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, No. 100, Shih-Chuan 1st Rd., Kaohsiung city 807, Taiwan.
| |
Collapse
|
10
|
Lin YT, Gonzalez-Rothi EJ, Lee KZ. Acute Hyperoxia Improves Spinal Cord Oxygenation and Circulatory Function Following Cervical Spinal Cord Injury in Rats. CHINESE J PHYSIOL 2024; 67:27-36. [PMID: 38780270 DOI: 10.4103/ejpi.ejpi-d-23-00003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 12/06/2023] [Indexed: 05/25/2024] Open
Abstract
Spinal cord injury is associated with spinal vascular disruptions that result in spinal ischemia and tissue hypoxia. This study evaluated the therapeutic efficacy of normobaric hyperoxia on spinal cord oxygenation and circulatory function at the acute stage of cervical spinal cord injury. Adult male Sprague Dawley rats underwent dorsal cervical laminectomy or cervical spinal cord contusion. At 1-2 days after spinal surgery, spinal cord oxygenation was monitored in anesthetized and spontaneously breathing rats through optical recording of oxygen sensor foils placed on the cervical spinal cord and pulse oximetry. The arterial blood pressure, heart rate, blood gases, and peripheral oxyhemoglobin saturation were also measured under hyperoxic (50% O2) and normoxic (21% O2) conditions. The results showed that contused animals had significantly lower spinal cord oxygenation levels than uninjured animals during normoxia. Peripheral oxyhemoglobin saturation, arterial oxygen partial pressure, and mean arterial blood pressure are significantly reduced following cervical spinal cord contusion. Notably, spinal oxygenation of contused rats could be improved to a level comparable to uninjured animals under hyperoxia. Furthermore, acute hyperoxia elevated blood pressure, arterial oxygen partial pressure, and peripheral oxyhemoglobin saturation. These results suggest that normobaric hyperoxia can significantly improve spinal cord oxygenation and circulatory function in the acute phase after cervical spinal cord injury. We propose that adjuvant normobaric hyperoxia combined with other hemodynamic optimization strategies may prevent secondary damage after spinal cord injury and improve functional recovery.
Collapse
Affiliation(s)
- Yen-Ting Lin
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Elisa J Gonzalez-Rothi
- Breathing Research and Therapeutics Center, Department of Physical Therapy, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Kun-Ze Lee
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
11
|
Liu Y, Wang Y, Wang Y, Zhou J, Ding W. The growth status and functions of olfactory ensheathing cells cultured on randomly oriented and aligned type-I-collagen-based nanofibrous scaffolds. NANOTECHNOLOGY 2023; 35:035101. [PMID: 37905427 DOI: 10.1088/1361-6528/ad02a4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 10/11/2023] [Indexed: 11/02/2023]
Abstract
Aim. The potential of olfactory ensheathing cells (OECs) as a cell therapy for spinal cord reconstruction and regeneration after injury has drawn significant attention in recent years. This study attempted to investigate the influences of nano-fibrous scaffolds on the growth status and functional properties of OECs.Methods.The ultra-morphology of the scaffolds was visualized using scanning electron microscopy (SEM). To culture OECs, donated cells were subcultured and identified with p75. Cell proliferation, apoptosis, and survival rates were measured through MTT assay, Annexin-V/PI staining, and p75 cell counting, respectively. The adhesion of cells cultured on scaffolds was observed using SEM. Additionally, the functions of OECs cultured on scaffolds were assessed by testing gene expression levels through real time polymerase chain reaction.Results.The electrospun type I collagen-based nano-fibers exhibited a smooth surface and uniform distribution. It was indicated that the proliferation and survival rates of OECs cultured on both randomly oriented and aligned type I collagen-based nano-fibrous scaffolds were higher than those observed in the collagen-coated control. Conversely, apoptosis rates were lower in cells cultured on scaffolds. Furthermore, OEC adhesion was better on the scaffolds than on the control. The expression levels of target genes were significantly elevated in cells cultured on scaffolds versus the controls.Conclusion.As a whole, the utilization of aligned collagen nanofibers has demonstrated significant advantages in promoting cell growth and improving cell function. These findings have important implications for the field of regenerative medicine and suggest that the approach may hold promise for the future therapeutic applications.
Collapse
Affiliation(s)
- Yugang Liu
- Department of Spinal Surgery, The Third Hospital of Hebei Medical University, 139 Ziqiang Road, Shijiazhuang, 050051, People's Republic of China
- Department of Orthopedic Surgery, Affiliated Hospital of Hebei University of Engineering, 81 Congtai Road, Handan, 056002, People's Republic of China
| | - Yansong Wang
- Department of Spine Surgery, Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Harbin, 150001, People's Republic of China
| | - Ying Wang
- Department of Orthopedic Surgery, Affiliated Hospital of Hebei University of Engineering, 81 Congtai Road, Handan, 056002, People's Republic of China
| | - Jihui Zhou
- Department of Spine Surgery, Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Harbin, 150001, People's Republic of China
| | - Wenyuan Ding
- Department of Spinal Surgery, The Third Hospital of Hebei Medical University, 139 Ziqiang Road, Shijiazhuang, 050051, People's Republic of China
| |
Collapse
|
12
|
Zuo Y, Ye J, Cai W, Guo B, Chen X, Lin L, Jin S, Zheng H, Fang A, Qian X, Abdelrahman Z, Wang Z, Zhang Z, Chen Z, Yu B, Gu X, Wang X. Controlled delivery of a neurotransmitter-agonist conjugate for functional recovery after severe spinal cord injury. NATURE NANOTECHNOLOGY 2023; 18:1230-1240. [PMID: 37308588 DOI: 10.1038/s41565-023-01416-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 05/04/2023] [Indexed: 06/14/2023]
Abstract
Despite considerable unmet medical needs, effective pharmacological treatments that promote functional recovery after spinal cord injury remain limited. Although multiple pathological events are implicated in spinal cord injuries, the development of a microinvasive pharmacological approach that simultaneously targets the different mechanisms involved in spinal cord injury remains a formidable challenge. Here we report the development of a microinvasive nanodrug delivery system that consists of amphiphilic copolymers responsive to reactive oxygen species and an encapsulated neurotransmitter-conjugated KCC2 agonist. Upon intravenous administration, the nanodrugs enter the injured spinal cord due to a disruption in the blood-spinal cord barrier and disassembly due to damage-triggered reactive oxygen species. The nanodrugs exhibit dual functions in the injured spinal cord: scavenging accumulated reactive oxygen species in the lesion, thereby protecting spared tissues, and facilitating the integration of spared circuits into the host spinal cord through targeted modulation of inhibitory neurons. This microinvasive treatment leads to notable functional recovery in rats with contusive spinal cord injury.
Collapse
Affiliation(s)
- Yanming Zuo
- Department of Neurobiology and Department of Rehabilitation Medicine, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, P. R. China
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| | - Jingjia Ye
- Department of Neurobiology and Department of Rehabilitation Medicine, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, P. R. China
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| | - Wanxiong Cai
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| | - Binjie Guo
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| | - Xiangfeng Chen
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| | - Lingmin Lin
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| | - Shuang Jin
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| | - Hanyu Zheng
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| | - Ao Fang
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| | - Xingran Qian
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| | - Zeinab Abdelrahman
- Department of Neurobiology and Department of Rehabilitation Medicine, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, P. R. China
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| | - Zhiping Wang
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| | - Zhipeng Zhang
- School of Chemistry and Molecular Engineering, Frontiers Science Center for Materiobiology and Dynamic Chemistry, East China University of Science & Technology, Shanghai, P. R. China
| | - Zuobin Chen
- Department of Neurobiology and Department of Rehabilitation Medicine, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, P. R. China
| | - Bin Yu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Nantong University, Nantong, P. R. China
| | - Xiaosong Gu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Nantong University, Nantong, P. R. China
| | - Xuhua Wang
- Department of Neurobiology and Department of Rehabilitation Medicine, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, P. R. China.
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, China.
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China.
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong, P. R. China.
| |
Collapse
|
13
|
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
|
14
|
Furube E, Ohgidani M, Yoshida S. Systemic Inflammation Leads to Changes in the Intracellular Localization of KLK6 in Oligodendrocytes in Spinal Cord White Matter. Neurochem Res 2023; 48:2645-2659. [PMID: 37067738 DOI: 10.1007/s11064-023-03929-5] [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: 11/21/2022] [Revised: 02/24/2023] [Accepted: 03/31/2023] [Indexed: 04/18/2023]
Abstract
Axonal injury and demyelination occur in demyelinating diseases, such as multiple sclerosis, and the detachment of myelin from axons precedes its degradation. Paranodes are the areas at which each layer of the myelin sheath adheres tightly to axons. The destruction of nodal and paranodal structures during inflammation is an important pathophysiology of various neurological disorders. However, the underlying pathological changes in these structures remain unclear. Kallikrein 6 (KLK6), a serine protease produced by oligodendrocytes, is involved in demyelinating diseases. In the present study, we intraperitoneally injected mice with LPS for several days and examined changes in the localization of KLK6. Transient changes in the intracellular localization of KLK6 to paranodes in the spinal cord were observed during LPS-induced systemic inflammation. However, these changes were not detected in the upper part of brain white matter. LPS-induced changes were suppressed by minocycline, suggesting the involvement of microglia. Moreover, nodal lengths were elongated in LPS-treated wild-type mice, but not in LPS-treated KLK6-KO mice. These results demonstrate the potential involvement of KLK6 in the process of demyelination.
Collapse
Affiliation(s)
- Eriko Furube
- Department of Functional Anatomy and Neuroscience, Asahikawa Medical University, Asahikawa, Hokkaido, 078-8510, Japan.
| | - Masahiro Ohgidani
- Department of Functional Anatomy and Neuroscience, Asahikawa Medical University, Asahikawa, Hokkaido, 078-8510, Japan
| | - Shigetaka Yoshida
- Department of Functional Anatomy and Neuroscience, Asahikawa Medical University, Asahikawa, Hokkaido, 078-8510, Japan
| |
Collapse
|
15
|
Gál L, Bellák T, Marton A, Fekécs Z, Weissman D, Török D, Biju R, Vizler C, Kristóf R, Beattie MB, Lin PJ, Pardi N, Nógrádi A, Pajer K. Restoration of Motor Function through Delayed Intraspinal Delivery of Human IL-10-Encoding Nucleoside-Modified mRNA after Spinal Cord Injury. RESEARCH (WASHINGTON, D.C.) 2023; 6:0056. [PMID: 36930811 PMCID: PMC10013810 DOI: 10.34133/research.0056] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 01/05/2023] [Indexed: 01/13/2023]
Abstract
Efficient in vivo delivery of anti-inflammatory proteins to modulate the microenvironment of an injured spinal cord and promote neuroprotection and functional recovery is a great challenge. Nucleoside-modified messenger RNA (mRNA) has become a promising new modality that can be utilized for the safe and efficient delivery of therapeutic proteins. Here, we used lipid nanoparticle (LNP)-encapsulated human interleukin-10 (hIL-10)-encoding nucleoside-modified mRNA to induce neuroprotection and functional recovery following rat spinal cord contusion injury. Intralesional administration of hIL-10 mRNA-LNP to rats led to a remarkable reduction of the microglia/macrophage reaction in the injured spinal segment and induced significant functional recovery compared to controls. Furthermore, hIL-10 mRNA treatment induced increased expression in tissue inhibitor of matrix metalloproteinase 1 and ciliary neurotrophic factor levels in the affected spinal segment indicating a time-delayed secondary effect of IL-10 5 d after injection. Our results suggest that treatment with nucleoside-modified mRNAs encoding neuroprotective factors is an effective strategy for spinal cord injury repair.
Collapse
Affiliation(s)
- László Gál
- Department of Anatomy, Histology and Embryology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Tamás Bellák
- Department of Anatomy, Histology and Embryology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Annamária Marton
- National Biotechnology Laboratory, Institute of Genetics, Biological Research Centre, Szeged, Hungary
- Institute of Biochemistry, Biological Research Centre, Szeged, Hungary
| | - Zoltán Fekécs
- Department of Anatomy, Histology and Embryology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Drew Weissman
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Dénes Török
- Department of Anatomy, Histology and Embryology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Rachana Biju
- Department of Anatomy, Histology and Embryology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Csaba Vizler
- National Biotechnology Laboratory, Institute of Genetics, Biological Research Centre, Szeged, Hungary
- Institute of Biochemistry, Biological Research Centre, Szeged, Hungary
| | - Rebeka Kristóf
- Department of Anatomy, Histology and Embryology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | | | | | - Norbert Pardi
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Antal Nógrádi
- Department of Anatomy, Histology and Embryology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Krisztián Pajer
- Department of Anatomy, Histology and Embryology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| |
Collapse
|
16
|
Guan B, Fan Y, Zheng R, Fu R, Yao L, Wang W, Li G, Chen L, Zhou H, Feng S. A critical appraisal of clinical practice guidelines on pharmacological treatments for spinal cord injury. Spine J 2023; 23:392-402. [PMID: 36182069 DOI: 10.1016/j.spinee.2022.09.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 09/16/2022] [Accepted: 09/20/2022] [Indexed: 02/03/2023]
Abstract
BACKGROUND CONTEXT Spinal cord injury brings devastating consequences and huge economic burden. Different authoritative organizations have developed different guidelines for pharmacological treatments of spinal cord injury, but there is a lack of a critical appraisal of them. PURPOSE To systematically review and appraise guidelines regarding their recommendations for pharmacological treatments for spinal cord injury. STUDY DESIGN Systematic review. METHODS We searched Medline, Embase, Cochrane, and Web of Science from January 2000 to January 2022 as well as guideline-specific databases (eg, Congress of Neurological Surgeons) and Google Scholar. We included the most updated guideline containing evidence-based recommendations or consensus-based recommendations developed by specific authoritative organizations if multiple versions were available. We appraised guidelines through the Appraisal of Guidelines for Research and Evaluation, 2nd edition instrument consisting of six domains (eg, applicability). With supporting evidence, recommendations were classified as: for, against, neither for nor against. We utilized an evidence assessment system to categorize the quality of supporting evidence as poor, fair, or good. RESULTS Eight guidelines developed from 2008 to 2020 were included, but all of them scored lowest in the domain of applicability among all six domains. Twelve pharmacological agents (eg, methylprednisolone) were studied. For methylprednisolone, three guidelines (3/8=37.5%) recommended for (one evidence-based and two consensus-based), three (3/8=37.5%) recommended against (all evidence-based), and two (2/8=25%) recommended neither for nor against. For monosialotetrahexosylganglioside (GM-1), one guideline (1/4=25%) recommended for (consensus-based), one (1/4=25%) recommended against (evidence-based), and two (2/4=50%) recommended neither for nor against. For other agents (eg, minocycline), most guidelines (3/5=60%) recommended neither for nor against, one (1/5=20%) recommended against naloxone (evidence-based) and nimodipine (evidence-based), and one (1/5=20%) recommended for neural growth factor (consensus-based). The quality of most of the supporting evidence was poor, and the rest was fair. CONCLUSIONS There were inconsistencies among recommendations for methylprednisolone and GM-1. Evidence-based recommendations tended to recommend against, whereas consensus-based recommendations tended to recommend for.
Collapse
Affiliation(s)
- Bin Guan
- Department of Orthopedics, Qilu Hospital of Shandong University, Shandong University Centre for Orthopedics, Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, P.R. China
| | - Yuxuan Fan
- Department of Orthopedics, Tianjin Medical University General Hospital, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord, Tianjin, 300052, P.R. China
| | - Ruiyuan Zheng
- Department of Orthopedics, Qilu Hospital of Shandong University, Shandong University Centre for Orthopedics, Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, P.R. China
| | - Runhan Fu
- Department of Orthopedics, Qilu Hospital of Shandong University, Shandong University Centre for Orthopedics, Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, P.R. China
| | - Liang Yao
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Canada
| | - Wei Wang
- Department of Orthopedics, Qilu Hospital of Shandong University, Shandong University Centre for Orthopedics, Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, P.R. China
| | - Guoyu Li
- Department of Orthopedics, Qilu Hospital of Shandong University, Shandong University Centre for Orthopedics, Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, P.R. China
| | - Lingxiao Chen
- Department of Orthopedics, Qilu Hospital of Shandong University, Shandong University Centre for Orthopedics, Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, P.R. China; Faculty of Medicine and Health, The Back Pain Research Team, Sydney Musculoskeletal Health, The Kolling Institute, University of Sydney, Sydney, Australia.
| | - Hengxing Zhou
- Department of Orthopedics, Qilu Hospital of Shandong University, Shandong University Centre for Orthopedics, Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, P.R. China; Department of Orthopedics, Tianjin Medical University General Hospital, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord, Tianjin, 300052, P.R. China.
| | - Shiqing Feng
- Department of Orthopedics, Qilu Hospital of Shandong University, Shandong University Centre for Orthopedics, Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, P.R. China; Department of Orthopedics, Tianjin Medical University General Hospital, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord, Tianjin, 300052, P.R. China.
| |
Collapse
|
17
|
Kim HN, McCrea MR, Li S. Advances in molecular therapies for targeting pathophysiology in spinal cord injury. Expert Opin Ther Targets 2023; 27:171-187. [PMID: 37017093 PMCID: PMC10148912 DOI: 10.1080/14728222.2023.2194532] [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: 01/23/2023] [Revised: 03/20/2023] [Accepted: 03/20/2023] [Indexed: 04/06/2023]
Abstract
INTRODUCTION Spinal cord injury (SCI) affects 25,000-50,000 people around the world each year and there is no cure for SCI patients currently. The primary injury damages spinal cord tissues and secondary injury mechanisms, including ischemia, apoptosis, inflammation, and astrogliosis, further exacerbate the lesions to the spinal cord. Recently, researchers have designed various therapeutic approaches for SCI by targeting its major cellular or molecular pathophysiology. AREAS COVERED Some strategies have shown promise in repairing injured spinal cord for functional recoveries, such as administering neuroprotective reagents, targeting specific genes to promote robust axon regeneration of disconnected spinal fiber tracts, targeting epigenetic factors to enhance cell survival and neural repair, and facilitating neuronal relay pathways and neuroplasticity for restoration of function after SCI. This review focuses on the major advances in preclinical molecular therapies for SCI reported in recent years. EXPERT OPINION Recent progress in developing novel and effective repairing strategies for SCI is encouraging, but many challenges remain for future design of effective treatments, including developing highly effective neuroprotectants for early interventions, stimulating robust neuronal regeneration with functional synaptic reconnections among disconnected neurons, maximizing the recovery of lost neural functions with combination strategies, and translating the most promising therapies into human use.
Collapse
Affiliation(s)
- Ha Neui Kim
- Shriners Hospitals Pediatric Research Center, Department of Neural Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Madeline R. McCrea
- Shriners Hospitals Pediatric Research Center, Department of Neural Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Shuxin Li
- Shriners Hospitals Pediatric Research Center, Department of Neural Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| |
Collapse
|
18
|
Overexpression of the X-Linked Inhibitor of Apoptosis Protein (XIAP) in Neurons Improves Cell Survival and the Functional Outcome after Traumatic Spinal Cord Injury. Int J Mol Sci 2023; 24:ijms24032791. [PMID: 36769152 PMCID: PMC9917926 DOI: 10.3390/ijms24032791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/24/2023] [Accepted: 01/26/2023] [Indexed: 02/05/2023] Open
Abstract
Mechanical trauma to the spinal cord causes extensive neuronal death, contributing to the loss of sensory-motor and autonomic functions below the injury location. Apoptosis affects neurons after spinal cord injury (SCI) and is associated with increased caspase activity. Cleavage of X-linked inhibitor of apoptosis protein (XIAP) after SCI may contribute to this rise in caspase activity. Accordingly, we have shown that the elevation of XIAP resulted in increased neuronal survival after SCI and improved functional recovery. Therefore, we hypothesise that neuronal overexpression of XIAP can be neuroprotective after SCI with improved functional recovery. In line with this, studies of a transgenic mice with overexpression of XIAP in neurons revealed that higher levels of XIAP after spinal cord trauma favours neuronal survival, tissue preservation, and motor recovery after the spinal cord trauma. Using human SH-SY5Y cells overexpressing XIAP, we further showed that XIAP reduced caspase activity and apoptotic cell death after pro-apoptotic stimuli. In conclusion, this study shows that the levels of XIAP expression are an important factor for the outcome of spinal cord trauma and identifies XIAP as an important therapeutic target for alleviating the deleterious effects of SCI.
Collapse
|
19
|
The Proteostasis Network: A Global Therapeutic Target for Neuroprotection after Spinal Cord Injury. Cells 2022; 11:cells11213339. [PMID: 36359735 PMCID: PMC9658791 DOI: 10.3390/cells11213339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/14/2022] [Accepted: 10/20/2022] [Indexed: 01/18/2023] Open
Abstract
Proteostasis (protein homeostasis) is critical for cellular as well as organismal survival. It is strictly regulated by multiple conserved pathways including the ubiquitin-proteasome system, autophagy, the heat shock response, the integrated stress response, and the unfolded protein response. These overlapping proteostasis maintenance modules respond to various forms of cellular stress as well as organismal injury. While proteostasis restoration and ultimately organism survival is the main evolutionary driver of such a regulation, unresolved disruption of proteostasis may engage pro-apoptotic mediators of those pathways to eliminate defective cells. In this review, we discuss proteostasis contributions to the pathogenesis of traumatic spinal cord injury (SCI). Most published reports focused on the role of proteostasis networks in acute/sub-acute tissue damage post-SCI. Those reports reveal a complex picture with cell type- and/or proteostasis mediator-specific effects on loss of neurons and/or glia that often translate into the corresponding modulation of functional recovery. Effects of proteostasis networks on such phenomena as neuro-repair, post-injury plasticity, as well as systemic manifestations of SCI including dysregulation of the immune system, metabolism or cardiovascular function are currently understudied. However, as potential interventions that target the proteostasis networks are expected to impact many cell types across multiple organ systems that are compromised after SCI, such therapies could produce beneficial effects across the wide spectrum of highly variable human SCI.
Collapse
|
20
|
Camargo C, Abode-Iyamah K, Shah JS, Bechtle PS, Freeman WD. Comprehensive Perioperative Approach to Complex Spine Deformity Management. Clin Spine Surg 2022; 35:310-318. [PMID: 34334699 DOI: 10.1097/bsd.0000000000001240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 06/27/2021] [Indexed: 10/20/2022]
Abstract
OBJECTIVE Study perioperative strategies for optimizing neuroprotection in complex spine deformity correction surgery. METHODS We report the case of a patient with severe lumbar dextroscoliosis, thoracolumbar junction hyperkyphosis with a 40-degree Cobb angle levoconvex scoliosis who underwent spinal deformity correction with loss of neuromonitoring during surgery. We performed a literature review on perioperative management of complex spine deformity. RESULTS A 50-year-old man presented with lumbar pain and right L4 radiculopathy. Surgical intervention for deformity correction and decompression was indicated with T4-L4 posterior instrumentation L2/L3 and L3/L4 transforaminal lumbar interbody fusion. Surgery was aborted due to the loss of neuromonitoring. Postsurgery, the patient had left sensory deficit and the neurocritical care team clinically suspected and deduced the anatomic location of the spinal cord compression. Magnetic resonance imaging confirmed a T10-T11 hyperintensity suggestive of cord ischemia due to osteophyte compressing the spinal cord. The patient underwent a second corrective surgery with no intraoperative events and has no long-term neurological sequela. CONCLUSIONS This case illustrates that a comprehensive perioperative approach and individualized risk factor assessment is useful in complex spine deformity surgery. Further research is needed to determine how this individualized comprehensive approach can lead to intraoperative and postoperative countermeasures that improved spine surgery outcomes. LEVEL OF EVIDENCE Level V.
Collapse
Affiliation(s)
| | | | | | | | - William D Freeman
- Departments of Neurologic Surgery
- Neurology
- Critical Care Medicine, Mayo Clinic, Jacksonville, FL
| |
Collapse
|
21
|
Sefiani A, Rusyn I, Geoffroy CG. Novel adult cortical neuron processing and screening method illustrates sex- and age-dependent effects of pharmaceutical compounds. Sci Rep 2022; 12:13125. [PMID: 35908049 PMCID: PMC9338961 DOI: 10.1038/s41598-022-17389-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 07/25/2022] [Indexed: 11/22/2022] Open
Abstract
Neurodegenerative diseases and neurotraumatic injuries are typically age-associated disorders that can reduce neuron survival, neurite outgrowth, and synaptic plasticity leading to loss of cognitive capacity, executive function, and motor control. In pursuit of reducing the loss of said neurological functions, novel compounds are sought that promote neuron viability, neuritogenesis, and/or synaptic plasticity. Current high content in vitro screenings typically use cells that are iPSC-derived, embryonic, or originate from post-natal tissues; however, most patients suffering from neurodegenerative diseases and neurotrauma are of middle-age and older. The chasm in maturity between the neurons used in drug screens and those in a target population is a barrier for translational success of in vitro results. It has been historically challenging to culture adult neurons let alone conduct screenings; therefore, age-appropriate drug screenings have previously not been plausible. We have modified Miltenyi's protocol to increase neuronal yield, neuron purity, and neural viability at a reduced cost to expand our capacity to screen compounds directly in primary adult neurons. To our knowledge, we developed the first morphology-based screening system using adult cortical neurons and the first to incorporate age and sex as biological variables in a screen using adult cortical neurons. By using primary adult cortical neurons from mice that were 4 to 48 weeks old for screening pharmaceutical agents, we have demonstrated age- and sex-dependent effects on neuritogenesis and neuron survival in vitro. Utilizing age- and sex-appropriate in vitro models to find novel compounds increasing neuron survival and neurite outgrowth, made possible by our modified adult neuron processing method, will greatly increase the relevance of in vitro screening for finding neuroprotective compounds.
Collapse
Affiliation(s)
- Arthur Sefiani
- Department of Neuroscience and Experimental Therapeutics, School of Medicine, Texas A&M University, Bryan, TX, 77807, USA
| | - Ivan Rusyn
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, TX, 77843, USA
| | - Cédric G Geoffroy
- Department of Neuroscience and Experimental Therapeutics, School of Medicine, Texas A&M University, Bryan, TX, 77807, USA.
| |
Collapse
|
22
|
Mothe AJ, Jacobson PB, Caprelli M, Ulndreaj A, Rahemipour R, Huang L, Monnier PP, Fehlings MG, Tator CH. Delayed administration of elezanumab, a human anti-RGMa neutralizing monoclonal antibody, promotes recovery following cervical spinal cord injury. Neurobiol Dis 2022; 172:105812. [PMID: 35810963 DOI: 10.1016/j.nbd.2022.105812] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 07/03/2022] [Accepted: 07/04/2022] [Indexed: 11/19/2022] Open
Abstract
Spinal cord injury (SCI) elicits a cascade of degenerative events including cell death, axonal degeneration, and the upregulation of inhibitory molecules which limit repair. Repulsive guidance molecule A (RGMa) is an axon growth inhibitor which is also involved in neuronal cell death and differentiation. SCI causes upregulation of RGMa in the injured rodent, non-human primate, and human spinal cord. Recently, we showed that delayed administration of elezanumab, a high affinity human RGMa-specific monoclonal antibody, promoted neuroprotective and regenerative effects following thoracic SCI. Since most human traumatic SCI is at the cervical level, and level-dependent anatomical and molecular differences may influence pathophysiological responses to injury and treatment, we examined the efficacy of elezanumab and its therapeutic time window of administration in a clinically relevant rat model of cervical impact-compression SCI. Pharmacokinetic analysis of plasma and spinal cord tissue lysate showed comparable levels of RGMa antibodies with delayed administration following cervical SCI. At 12w after SCI, elezanumab promoted long term benefits including perilesional sparing of motoneurons and increased neuroplasticity of key descending pathways involved in locomotion and fine motor function. Elezanumab also promoted growth of corticospinal axons into spinal cord gray matter and enhanced serotonergic innervation of the ventral horn to form synaptic connections caudal to the cervical lesion. Significant recovery in grip and trunk/core strength, locomotion and gait, and spontaneous voiding ability was found in rats treated with elezanumab either immediately post-injury or at 3 h post-SCI, and improvements in specific gait parameters were found when elezanumab was delayed to 24 h post-injury. We also developed a new locomotor score, the Cervical Locomotor Score, a simple and sensitive measure of trunk/core and limb strength and stability during dynamic locomotion.
Collapse
Affiliation(s)
- Andrea J Mothe
- Division of Experimental and Translational Neuroscience, Krembil Brain Institute & University Health Network, Toronto, M5T 0S8, ON, Canada.
| | - Peer B Jacobson
- Department of Translational Sciences, AbbVie Inc., North Chicago, IL 60064, USA
| | - Mitchell Caprelli
- Division of Experimental and Translational Neuroscience, Krembil Brain Institute & University Health Network, Toronto, M5T 0S8, ON, Canada
| | - Antigona Ulndreaj
- Division of Experimental and Translational Neuroscience, Krembil Brain Institute & University Health Network, Toronto, M5T 0S8, ON, Canada
| | - Radmehr Rahemipour
- Division of Experimental and Translational Neuroscience, Krembil Brain Institute & University Health Network, Toronto, M5T 0S8, ON, Canada
| | - Lili Huang
- AbbVie Biologics, AbbVie Bioresearch Center, Worcester, MA 01605, USA
| | - Philippe P Monnier
- Division of Experimental and Translational Neuroscience, Krembil Brain Institute & University Health Network, Toronto, M5T 0S8, ON, Canada; Department of Ophthalmology and Vision Science, University of Toronto, Toronto, M5S 3H6, ON, Canada
| | - Michael G Fehlings
- Division of Experimental and Translational Neuroscience, Krembil Brain Institute & University Health Network, Toronto, M5T 0S8, ON, Canada; Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, M5T 2S8, ON, Canada
| | - Charles H Tator
- Division of Experimental and Translational Neuroscience, Krembil Brain Institute & University Health Network, Toronto, M5T 0S8, ON, Canada; Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, M5T 2S8, ON, Canada.
| |
Collapse
|
23
|
Capuz A, Karnoub MA, Osien S, Rose M, Mériaux C, Fournier I, Devos D, Vanden Abeele F, Rodet F, Cizkova D, Salzet M. The Antibody Dependant Neurite Outgrowth Modulation Response Involvement in Spinal Cord Injury. Front Immunol 2022; 13:882830. [PMID: 35784350 PMCID: PMC9245426 DOI: 10.3389/fimmu.2022.882830] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 05/02/2022] [Indexed: 12/25/2022] Open
Abstract
Spinal cord injury (SCI) represents a major medical challenge. At present, there is still no cure to treat it efficiently and enable functional recovery below the injury site. Previously, we demonstrated that inflammation determines the fate of the physiopathology. To decipher the molecular mechanisms involved in this process, we performed a meta-analysis of our spatio-temporal proteomic studies in the time course of SCI. This highlighted the presence of IgG isotypes in both spinal cord explants and their secretomes. These IgGs were detected in the spinal cord even if no SCI occurred. However, during the time course following SCI, abundance of IgG1 and IgG2 subclasses (a, b, c) varied according to the spatial repartition. IgG1 was clearly mostly abundant at 12 h, and a switch to IgG2a was observed after 24 h. This IgG stayed predominant 3, 7, and 10 days after SCI. A protein related to IgM as well as a variable heavy chain were only detected 12 h after lesion. Interestingly, treatment with RhoA inhibitor influenced the abundance of the various IgG isotypes and a preferential switch to IgG2c was observed. By data reuse of rat dorsal root ganglion (DRG) neurons RNAseq datasets and RT-PCR experiments performed on cDNA from DRG sensory neurons ND7/23 and N27 dopaminergic neural cell lines, we confirmed expression of immunoglobulin heavy and light chains (constant and variable) encoding genes in neurons. We then identified CD16 and CD32b as their specific receptors in sensory neuron cell line ND7/23 and their activation regulated neurites outgrowth. These results suggest that during SCI, neuronal IgG isotypes are released to modulate neurites outgrowth. Therefore, we propose a new view of the SCI response involving an antibody dependent neurite outgrowth modulation (ADNM) which could be a precursor to the neuroinflammatory response in pathological conditions.
Collapse
Affiliation(s)
- Alice Capuz
- Université de Lille, Inserm U1192, Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse (PRISM), Lille, France
| | - Mélodie-Anne Karnoub
- Université de Lille, Inserm U1192, Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse (PRISM), Lille, France
| | - Sylvain Osien
- Université de Lille, Inserm U1192, Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse (PRISM), Lille, France
| | - Mélanie Rose
- Université de Lille, Inserm U1192, Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse (PRISM), Lille, France
| | - Céline Mériaux
- Université de Lille, Inserm U1192, Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse (PRISM), Lille, France
| | - Isabelle Fournier
- Université de Lille, Inserm U1192, Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse (PRISM), Lille, France
- Institut Universitaire de France, Paris, France
| | - David Devos
- Université de Lille, Inserm U1172, CHU-Lille, Lille Neuroscience Cognition Research Centre, Lille, France
| | - Fabien Vanden Abeele
- Université de Lille, Inserm U1003, Laboratory of Cell Physiology, Villeneuve d’Ascq, France
| | - Franck Rodet
- Université de Lille, Inserm U1192, Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse (PRISM), Lille, France
| | - Dasa Cizkova
- Université de Lille, Inserm U1192, Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse (PRISM), Lille, France
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
- Centre for Experimental and Clinical Regenerative Medicine, University of Veterinary Medicine and Pharmacy in Kosice, Kosice, Slovakia
- *Correspondence: Michel Salzet, ; Dasa Cizkova,
| | - Michel Salzet
- Université de Lille, Inserm U1192, Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse (PRISM), Lille, France
- Institut Universitaire de France, Paris, France
- *Correspondence: Michel Salzet, ; Dasa Cizkova,
| |
Collapse
|
24
|
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
|
25
|
Estrada V, Oldenburg E, Popa O, Muller HW. Mapping the long rocky road to effective spinal cord injury therapy - A meta-review of pre-clinical and clinical research. J Neurotrauma 2022; 39:591-612. [PMID: 35196894 DOI: 10.1089/neu.2021.0298] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Spinal cord injury (SCI) is a rare condition, which even after decades of research, to date still presents an incurable condition with a complex symptomatology. SCI can result in paralysis, pain, loss of sensation, bladder and sexual dysfunction, and muscle degeneration to name but a few. The large number of publications makes it difficult to keep track of current progress in the field and of the many treatment options, which have been suggested and are being proposed with increasing frequency. Scientific databases with user-oriented search options will offer possible solutions, but they are still mostly in the development phase. In this meta-analysis, we summarize and narrow down SCI therapeutic approaches applied in pre-clinical and clinical research. Statistical analyses of treatment clusters - assorted after counting annual publication numbers in PubMed and ClinicalTrials.gov databases - were performed to allow the comparison of research foci and of their translation efficacy into clinical therapy. Using the example of SCI research, our findings demonstrate the challenges that come with the accelerating research progress - an issue, which many research fields are faced with today. The analyses point out similarities and differences in the prioritization of SCI research in pre-clinical versus clinical therapy strategies. Moreover, the results demonstrate the rapidly growing importance of modern (bio-)engineering technologies.
Collapse
Affiliation(s)
- Veronica Estrada
- Heinrich Heine University Düsseldorf, 9170, Neurology, Molecular Neurobiology Laboratory, Düsseldorf, Germany;
| | - Ellen Oldenburg
- Heinrich Heine University Düsseldorf, 9170, Institute of Quantitative and Theoretical Biology, Düsseldorf, Germany;
| | - Ovidiu Popa
- Heinrich Heine University Düsseldorf, 9170, Institute of Quantitative and Theoretical Biology, Düsseldorf, Germany;
| | - Hans W Muller
- Heinrich Heine University Düsseldorf, 9170, Neurology, Düsseldorf, Germany;
| |
Collapse
|
26
|
Masterman E, Ahmed Z. Experimental Treatments for Oedema in Spinal Cord Injury: A Systematic Review and Meta-Analysis. Cells 2021; 10:cells10102682. [PMID: 34685662 PMCID: PMC8534777 DOI: 10.3390/cells10102682] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 09/21/2021] [Accepted: 10/05/2021] [Indexed: 12/09/2022] Open
Abstract
The incidence of spinal cord injury (SCI) is ever-growing, resulting in life-changing neurological deficits which can have devastating long-term impacts on a person’s quality of life. There is an unmet clinical need for a treatment which will prevent progression of the injury, allowing improved axonal regeneration and functional recovery to occur. The initial mechanical insult, followed by a cascade of secondary mechanisms, leads to the exacerbation and remodelling of the lesion site, thus inhibiting neurological recovery. Oedema rapidly accumulates following SCI and contributes to the detrimental pathophysiology and worsens functional outcomes. This study systematically reviewed the current experimental treatments being explored in the field of SCI, which specifically target oedema. Abiding by PRISMA guidelines and strict inclusion criteria, 14 studies were identified and analysed from three online databases (PubMed, Web of Science and EMBASE). As a result, we identified three key modalities which attenuate oedema: selective inhibition of the main water channel protein, aquaporin 4 (AQP4), modulation of inflammation and surgical interventions. Collectively, however, they all result in the downregulation of AQP4, which crucially leads to a reduction in oedema and improved functional outcomes. We concluded that trifluoperazine (TFP), a calmodulin kinase inhibitor which prevents the cell-surface localisation of AQP4, was the most efficacious treatment, significantly eliminating oedema within 7 days of administration. To date, this study is the most concise analysis of current experimental treatments for oedema, exposing its molecular mechanisms and assessing potential therapeutic pathways for future research.
Collapse
Affiliation(s)
- Emma Masterman
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK;
| | - Zubair Ahmed
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK;
- Centre for Trauma Sciences Research, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
- Surgical Reconstruction and Microbiology Research Centre, National Institute for Health Research, Queen Elizabeth Hospital, Birmingham B15 2TH, UK
- Correspondence:
| |
Collapse
|
27
|
Campos J, Silva NA, Salgado AJ. Nutritional interventions for spinal cord injury: preclinical efficacy and molecular mechanisms. Nutr Rev 2021; 80:1206-1221. [PMID: 34472615 DOI: 10.1093/nutrit/nuab068] [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] [Indexed: 12/17/2022] Open
Abstract
Spinal cord injury (SCI) is a debilitating condition that leads to motor, sensory, and autonomic impairments. Its intrinsic pathophysiological complexity has hindered the establishment of effective treatments for decades. Nutritional interventions (NIs) for SCI have been proposed as a route to circumvent some of the problems associated with this condition. Results obtained in animal models point to a more holistic effect, rather than to specific modulation, of several relevant SCI pathophysiological processes. Indeed, published data have shown NI improves energetic imbalance, oxidative damage, and inflammation, which are promoters of improved proteostasis and neurotrophic signaling, leading ultimately to neuroprotection and neuroplasticity. This review focuses on the most well-documented Nis. The mechanistic implications and their translational potential for SCI are discussed.
Collapse
Affiliation(s)
- Jonas Campos
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Nuno A Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - António J Salgado
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| |
Collapse
|
28
|
Gilmour AD, Reshamwala R, Wright AA, Ekberg JAK, St John JA. Optimizing Olfactory Ensheathing Cell Transplantation for Spinal Cord Injury Repair. J Neurotrauma 2021; 37:817-829. [PMID: 32056492 DOI: 10.1089/neu.2019.6939] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Cell transplantation constitutes an important avenue for development of new treatments for spinal cord injury (SCI). These therapies are aimed at supporting neural repair and/or replacing lost cells at the injury site. To date, various cell types have been trialed, with most studies focusing on different types of stem cells or glial cells. Here, we review commonly used cell transplantation approaches for spinal cord injury (SCI) repair, with focus on transplantation of olfactory ensheathing cells (OECs), the glial cells of the primary olfactory nervous system. OECs are promising candidates for promotion of neural repair given that they support continuous regeneration of the olfactory nerve that occurs throughout life. Further, OECs can be accessed from the nasal mucosa (olfactory neuroepithelium) at the roof of the nasal cavity and can be autologously transplanted. OEC transplantation has been trialed in many animal models of SCI, as well as in human clinical trials. While several studies have been promising, outcomes are variable and the method needs improvement to enhance aspects such as cell survival, integration, and migration. As a case study, we include the approaches used by our team (the Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Nathan, QLD, Australia) to address the current problems with OEC transplantation and discuss how the therapeutic potential of OEC transplantation can be improved. Our approach includes discovery research to improve our knowledge of OEC biology, identifying natural and synthetic compounds to stimulate the neural repair properties of OECs, and designing three-dimensional cell constructs to create stable and transplantable cell structures.
Collapse
Affiliation(s)
- Aaron D Gilmour
- Clem Jones Centre for Neurobiology and Stem Cell Research and Griffith University, Nathan, Queensland, Australia.,Menzies Health Institute Queensland, Griffith University, Southport, Queensland, Australia
| | - Ronak Reshamwala
- Clem Jones Centre for Neurobiology and Stem Cell Research and Griffith University, Nathan, Queensland, Australia.,Menzies Health Institute Queensland, Griffith University, Southport, Queensland, Australia.,Griffith Institute for Drug Discovery, Griffith University, Nathan, Queensland, Australia
| | - Alison A Wright
- Clem Jones Centre for Neurobiology and Stem Cell Research and Griffith University, Nathan, Queensland, Australia.,Menzies Health Institute Queensland, Griffith University, Southport, Queensland, Australia
| | - Jenny A K Ekberg
- Clem Jones Centre for Neurobiology and Stem Cell Research and Griffith University, Nathan, Queensland, Australia.,Menzies Health Institute Queensland, Griffith University, Southport, Queensland, Australia.,Griffith Institute for Drug Discovery, Griffith University, Nathan, Queensland, Australia
| | - James A St John
- Clem Jones Centre for Neurobiology and Stem Cell Research and Griffith University, Nathan, Queensland, Australia.,Menzies Health Institute Queensland, Griffith University, Southport, Queensland, Australia.,Griffith Institute for Drug Discovery, Griffith University, Nathan, Queensland, Australia
| |
Collapse
|
29
|
Liu X, Zhang Y, Wang Y, Qian T. Inflammatory Response to Spinal Cord Injury and Its Treatment. World Neurosurg 2021; 155:19-31. [PMID: 34375779 DOI: 10.1016/j.wneu.2021.07.148] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/30/2021] [Accepted: 07/31/2021] [Indexed: 01/14/2023]
Abstract
Spinal cord injury (SCI), as one of the intractable diseases in clinical medicine, affects thousands of human beings, and the pathologic changes after injury have been a hot spot for exploration in clinical medicine. With the development of new treatments, the survival of patients has shown an increasing trend; however, the inflammatory response after injury has not yet been effectively controlled. SCI is divided into primary injury and secondary injury according to the time of injury and pathophysiologic changes. Primary injury occurs immediately and the damage to the injury site is irreversible; however, secondary injury occurs after primary injury and involves pathologic changes at the cellular and molecular levels, which are reversible. Thus, the inflammatory response from secondary injuries has become the main direction of research. In recent years, a complex pathophysiologic mechanism has gradually been unveiled, which has been followed by an upgrade of treatment methods. This article describes the mechanisms of the inflammatory response after SCI and the mainstream treatment modalities. Also, neuroprotective agents and nerve regeneration agent agents are commonly used in the treatment of SCI; the therapeutic mechanism and classification of these agents are reviewed.
Collapse
Affiliation(s)
- Xiangyu Liu
- Department of Orthopedics, First Affiliated Hospital of Bengbu Medical College, Bengbu, P.R. China
| | - Yiwen Zhang
- Department of Orthopedics, First Affiliated Hospital of Bengbu Medical College, Bengbu, P.R. China
| | - Yitong Wang
- Department of Orthopedics, First Affiliated Hospital of Bengbu Medical College, Bengbu, P.R. China
| | - Taibao Qian
- Department of Orthopedics, First Affiliated Hospital of Bengbu Medical College, Bengbu, P.R. China.
| |
Collapse
|
30
|
Brakel K, Aceves M, Garza A, Yoo C, Escobedo G, Panchani N, Shapiro L, Hook M. Inflammation increases the development of depression behaviors in male rats after spinal cord injury. Brain Behav Immun Health 2021; 14:100258. [PMID: 34589764 PMCID: PMC8474513 DOI: 10.1016/j.bbih.2021.100258] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 04/01/2021] [Accepted: 04/08/2021] [Indexed: 02/08/2023] Open
Abstract
Following spinal cord injury, 18-26% of patients are diagnosed with depressive disorders, compared to 8-12% in the general population. As increased inflammation strongly correlates with depression in both animal and human studies, we hypothesized that the immune activation inherent to SCI could increase depression-like behavior. Thus, we proposed that reducing immune activation with minocycline, a microglial inhibitor, would decrease depression-like behavior following injury. Male Sprague-Dawley rats were given minocycline in their drinking water for 14 days following a moderate, mid-thoracic (T12) spinal contusion. An array of depression-like behaviors (social activity, sucrose preference, forced swim, open field activity) were examined prior to injury as well as on days 9-10, 19-20, and 29-30 post-injury. Peripheral cytokine levels were analyzed in serum collected prior to injury and 10 days post-injury. Hierarchical cluster analysis divided subjects into two groups based on behavior: depressed and not-depressed. Depressed subjects displayed lower levels of open field activity and social interaction relative to their not-depressed counterparts. Depressed subjects also showed significantly greater expression of pro-inflammatory cytokines both before and after injury and displayed lower levels of hippocampal neurogenesis than not-depressed subjects. Intriguingly, subjects who later showed depressive behaviors had higher baseline levels of the pro-inflammatory cytokine IL-6, which persisted throughout the duration of the experiment. Minocycline, however, did not affect serum cytokine levels and did not block the development of depression; equal numbers of minocycline versus vehicle-treated subjects appeared in both phenotypic groups. Despite this, these data overall suggest that molecular correlates of inflammation prior to injury could predict the development of depression after a physical stressor.
Collapse
Affiliation(s)
- Kiralyn Brakel
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University, Medical Research and Education Building, Ste. 1005 8447 Riverside Pkwy, Bryan, TX, 77807, United States
- Texas A&M Institute of Neuroscience, Texas A&M University, Interdisciplinary Life Sciences Building, Rm 3148, 3474, TAMU, College Station, TX, United States
| | - Miriam Aceves
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University, Medical Research and Education Building, Ste. 1005 8447 Riverside Pkwy, Bryan, TX, 77807, United States
- Texas A&M Institute of Neuroscience, Texas A&M University, Interdisciplinary Life Sciences Building, Rm 3148, 3474, TAMU, College Station, TX, United States
- Department of Biology, Texas A&M University, Interdisciplinary Life Sciences Building, College Station, TX, United States
| | - Aryana Garza
- Texas A&M Institute of Neuroscience, Texas A&M University, Interdisciplinary Life Sciences Building, Rm 3148, 3474, TAMU, College Station, TX, United States
| | - Chaeyoung Yoo
- Texas A&M Institute of Neuroscience, Texas A&M University, Interdisciplinary Life Sciences Building, Rm 3148, 3474, TAMU, College Station, TX, United States
| | - Gabriel Escobedo
- Texas A&M Institute of Neuroscience, Texas A&M University, Interdisciplinary Life Sciences Building, Rm 3148, 3474, TAMU, College Station, TX, United States
| | - Nishah Panchani
- Texas A&M Institute of Neuroscience, Texas A&M University, Interdisciplinary Life Sciences Building, Rm 3148, 3474, TAMU, College Station, TX, United States
| | - Lee Shapiro
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University, Medical Research and Education Building, Ste. 1005 8447 Riverside Pkwy, Bryan, TX, 77807, United States
- Texas A&M Institute of Neuroscience, Texas A&M University, Interdisciplinary Life Sciences Building, Rm 3148, 3474, TAMU, College Station, TX, United States
| | - Michelle Hook
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University, Medical Research and Education Building, Ste. 1005 8447 Riverside Pkwy, Bryan, TX, 77807, United States
- Texas A&M Institute of Neuroscience, Texas A&M University, Interdisciplinary Life Sciences Building, Rm 3148, 3474, TAMU, College Station, TX, United States
| |
Collapse
|
31
|
Sangari S, Kirshblum S, Guest JD, Oudega M, Perez MA. Distinct patterns of spasticity and corticospinal connectivity following complete spinal cord injury. J Physiol 2021; 599:4441-4454. [PMID: 34107068 DOI: 10.1113/jp281862] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 06/01/2021] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS Damage to corticospinal axons have implications for the development of spasticity following spinal cord injury (SCI). Here, we examined to which extent residual corticospinal connections and spasticity are present in muscles below the injury (quadriceps femoris and soleus) in humans with motor complete thoracic SCI. We found three distinct sub-groups of people: participants with spasticity and corticospinal responses in the quadriceps femoris and soleus, participants with spasticity and corticospinal responses in the quadriceps femoris only, and participants with no spasticity or corticospinal responses in either muscle. Spasticity and corticospinal responses were present in the quadriceps but never only in the soleus muscle, suggesting a proximal to distal gradient of symptoms of hyperreflexia. These results suggest that concomitant patterns of residual corticospinal connectivity and spasticity exist in humans with motor complete SCI and that a clinical exam of spasticity might be a good predictor of residual corticospinal connectivity. ABSTRACT The loss of corticospinal axons has implications for the development of spasticity following spinal cord injury (SCI). However, the extent to which residual corticospinal connections and spasticity are present across muscles below the injury remains unknown. To address this question, we tested spasticity using the Modified Ashworth Scale and transmission in the corticospinal pathway by examining motor evoked potentials elicited by transcranial magnetic stimulation over the leg motor cortex (cortical MEPs) and by direct activation of corticospinal axons by electrical stimulation over the thoracic spine (thoracic MEPs), in the quadriceps femoris and soleus muscles, in 30 individuals with motor complete thoracic SCI. Cortical MEPs were also conditioned by thoracic electrical stimulation at intervals allowing their summation or collision. We found three distinct sub-groups of participants: 47% showed spasticity in the quadriceps femoris and soleus muscle, 30% showed spasticity in the quadriceps femoris muscle only, and 23% showed no spasticity in either muscle. While cortical MEPs were present only in the quadriceps in participants with spasticity, thoracic MEPs were present in both muscles when spasticity was present. Thoracic electrical stimulation facilitated and suppressed cortical MEPs, showing that both forms of stimulation activated similar corticospinal axons. Cortical and thoracic MEPs correlated with the degree of spasticity in both muscles. These results provide the first evidence that related patterns of residual corticospinal connectivity and spasticity exist in muscles below the injury after motor complete thoracic SCI and highlight that a clinical exam of spasticity can predict residual corticospinal connectivity after severe paralysis. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Sina Sangari
- Shirley Ryan AbilityLab, Chicago, Illinois, 60611.,Department of Physical Medicine and Rehabilitation, Northwestern University, Chicago, Illinois, 60611
| | - Steven Kirshblum
- Kessler Institute for Rehabilitation, Department of Physical Medicine and Rehabilitation, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - James D Guest
- The Miami Project to Cure Paralysis, University of Miami, Miami, 33136
| | - Martin Oudega
- Shirley Ryan AbilityLab, Chicago, Illinois, 60611.,Department of Physical Therapy and Human Movement Sciences, Northwestern University, Chicago, Illinois, 60611.,Edward Hines Jr. VA Hospital, Hines, Illinois, 60141
| | - Monica A Perez
- Shirley Ryan AbilityLab, Chicago, Illinois, 60611.,Department of Physical Medicine and Rehabilitation, Northwestern University, Chicago, Illinois, 60611.,Department of Physical Therapy and Human Movement Sciences, Northwestern University, Chicago, Illinois, 60611.,Edward Hines Jr. VA Hospital, Hines, Illinois, 60141
| |
Collapse
|
32
|
Sharma P, Shah PK. In vivo electrophysiological mechanisms underlying cervical epidural stimulation in adult rats. J Physiol 2021; 599:3121-3150. [PMID: 33894695 DOI: 10.1113/jp281146] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 03/15/2021] [Indexed: 12/11/2022] Open
Abstract
KEY POINTS To electrophysiologically determine the predominant neural structures activated with cervical epidural stimulation (ES), well-established electrophysiological protocols (single-pulse, paired-pulse and multiple frequency stimulation) were delivered at rest, during motor activity and under anaesthesia in adult rats. Cervical ES resulted in spinal evoked motor responses with three different waveforms - early response (ER), middle response (MR) and late response (LR). ERs remained unmodulated by repeated stimulation protocols. In contrast, MRs and LRs were modulated by repeated stimulation protocols and volitional motor activity. ERs are consequential to the direct activation of motor efferents; MRs are secondary to type-I sensory afferent activation and LRs result from the engagement of wider spinal interneuronal circuitry with potential influence from supraspinal pathways. Evidence from this work is fundamental in enhancing our understanding of cervical ES, and critical in refining the design of neuromodulation-based rehabilitative strategies and in the construction of neuroprosthetics. ABSTRACT Epidural stimulation (ES) of the lumbar spinal cord has demonstrated significant improvements in various physiological functions after a traumatic spinal cord injury in humans. Electrophysiological evidence from rodent, human and computational studies collectively suggest that the functional recovery following lumbar ES is mediated via direct activation of sensory afferent fibres. However, the mechanisms underlying cervical ES have not been comprehensively studied, which greatly limits our understanding of its effectiveness in restoring upper limb function. In this work, we determined the predominant neural structures that are activated with cervical ES using in vivo cervical spinal evoked motor responses (SEMRs). Standard electrophysiological protocols (single-pulse, paired-pulse and multiple frequency stimulation) were implemented in 11 awake and anaesthetized rats in four experimental stages. Three distinct types of cervical SEMRs were identified based on latency of their appearance: early response (ER), middle response (MR) and late response (LR). ERs remained unmodulated by repeated stimulation protocols. MRs and LRs were modulated by repeated stimulation protocols and volitional motor activity. Except for LRs being completely abolished under urethane, ketamine or urethane anaesthesia did not affect the appearance of cervical SEMRs. Our data, backed by literature, suggest that ERs are secondary to the direct activation of motor efferents, MRs are elicited by activation of type-I sensory afferents and LRs result from the engagement of interneuronal circuitry with potential influence from supraspinal pathways. The gathered information paves the way to designing motor rehabilitation strategies that can utilize cervical ES to recover upper limb function following neurological deficits.
Collapse
Affiliation(s)
- Pawan Sharma
- Division of Rehabilitation Sciences, Department of Physical Therapy, School of Health Technology and Management, Stony Brook University, Stony Brook, NY, 11727, USA
| | - Prithvi K Shah
- Division of Rehabilitation Sciences, Department of Physical Therapy, School of Health Technology and Management, Stony Brook University, Stony Brook, NY, 11727, USA
| |
Collapse
|
33
|
Jacobson PB, Goody R, Lawrence M, Mueller BK, Zhang X, Hooker BA, Pfleeger K, Ziemann A, Locke C, Barraud Q, Droescher M, Bernhard J, Popp A, Boeser P, Huang L, Mollon J, Mordashova Y, Cui YF, Savaryn JP, Grinnell C, Dreher I, Gold M, Courtine G, Mothe A, Tator CH, Guest JD. Elezanumab, a human anti-RGMa monoclonal antibody, promotes neuroprotection, neuroplasticity, and neurorecovery following a thoracic hemicompression spinal cord injury in non-human primates. Neurobiol Dis 2021; 155:105385. [PMID: 33991647 DOI: 10.1016/j.nbd.2021.105385] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 03/10/2021] [Accepted: 04/30/2021] [Indexed: 01/21/2023] Open
Abstract
Spinal cord injury (SCI) is a devastating condition characterized by loss of function, secondary to damaged spinal neurons, disrupted axonal connections, and myelin loss. Spontaneous recovery is limited, and there are no approved pharmaceutical treatments to reduce ongoing damage or promote repair. Repulsive guidance molecule A (RGMa) is upregulated following injury to the central nervous system (CNS), where it is believed to induce neuronal apoptosis and inhibit axonal growth and remyelination. We evaluated elezanumab, a human anti-RGMa monoclonal antibody, in a novel, newly characterized non-human primate (NHP) hemicompression model of thoracic SCI. Systemic intravenous (IV) administration of elezanumab over 6 months was well tolerated and associated with significant improvements in locomotor function. Treatment of animals for 16 weeks with a continuous intrathecal infusion of elezanumab below the lesion was not efficacious. IV elezanumab improved microstructural integrity of extralesional tissue as reflected by higher fractional anisotropy and magnetization transfer ratios in treated vs. untreated animals. IV elezanumab also reduced SCI-induced increases in soluble RGMa in cerebrospinal fluid, and membrane bound RGMa rostral and caudal to the lesion. Anterograde tracing of the corticospinal tract (CST) from the contralesional motor cortex following 20 weeks of IV elezanumab revealed a significant increase in the density of CST fibers emerging from the ipsilesional CST into the medial/ventral gray matter. There was a significant sprouting of serotonergic (5-HT) fibers rostral to the injury and in the ventral horn of lower thoracic regions. These data demonstrate that 6 months of intermittent IV administration of elezanumab, beginning within 24 h after a thoracic SCI, promotes neuroprotection and neuroplasticity of key descending pathways involved in locomotion. These findings emphasize the mechanisms leading to improved recovery of neuromotor functions with elezanumab in acute SCI in NHPs.
Collapse
Affiliation(s)
- Peer B Jacobson
- Department of Translational Sciences, Imaging Research, AbbVie Inc., 1 North Waukegan Rd., North Chicago, IL 60064, United States of America.
| | - Robin Goody
- Virscio, New Haven, CT, United States of America
| | | | - Bernhard K Mueller
- Discovery Biology, AbbVie Deutschland GmbH & Co. KG, Neuroscience Research, Knollstrasse, 67061 Ludwigshafen, Germany
| | - Xiaomeng Zhang
- Department of Translational Sciences, Imaging Research, AbbVie Inc., 1 North Waukegan Rd., North Chicago, IL 60064, United States of America
| | - Bradley A Hooker
- Department of Translational Sciences, Imaging Research, AbbVie Inc., 1 North Waukegan Rd., North Chicago, IL 60064, United States of America
| | - Kimberly Pfleeger
- Department of Neuroscience Development, AbbVie Inc., 1 North Waukegan Rd., North Chicago, IL 60064, United States of America
| | - Adam Ziemann
- Department of Neuroscience Development, AbbVie Inc., 1 North Waukegan Rd., North Chicago, IL 60064, United States of America
| | - Charles Locke
- Department of Biometrics, AbbVie Inc., 1 North Waukegan Rd., North Chicago, IL 60064, United States of America
| | - Quentin Barraud
- Center for Neuroprosthetics and Brain Mind Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Geneva, Switzerland; Department of Clinical Neuroscience, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland; Defitech Center for Interventional Neurotherapies, (NeuroRestore), CHUV/UNIL/EPFL, Lausanne, Switzerland
| | - Mathias Droescher
- Discovery Biology, AbbVie Deutschland GmbH & Co. KG, Neuroscience Research, Knollstrasse, 67061 Ludwigshafen, Germany
| | - Joerg Bernhard
- Discovery Biology, AbbVie Deutschland GmbH & Co. KG, Neuroscience Research, Knollstrasse, 67061 Ludwigshafen, Germany
| | - Andreas Popp
- Department of Preclinical Safety, AbbVie Deutschland GmbH & Co. KG, Neuroscience Research, Knollstrasse, 67061 Ludwigshafen, Germany
| | - Preethne Boeser
- Department of Preclinical Safety, AbbVie Deutschland GmbH & Co. KG, Neuroscience Research, Knollstrasse, 67061 Ludwigshafen, Germany
| | - Lili Huang
- AbbVie Biologics, AbbVie Bioresearch Center, 381 Plantation St., Worcester, MA 01605, United States of America
| | - Jennifer Mollon
- Data and Statistical Sciences, AbbVie Deutschland GmbH & Co. KG, Neuroscience Research, Knollstrasse, 67061 Ludwigshafen, Germany
| | - Yulia Mordashova
- Data and Statistical Sciences, AbbVie Deutschland GmbH & Co. KG, Neuroscience Research, Knollstrasse, 67061 Ludwigshafen, Germany
| | - Yi-Fang Cui
- Discovery Biology, AbbVie Deutschland GmbH & Co. KG, Neuroscience Research, Knollstrasse, 67061 Ludwigshafen, Germany
| | - John P Savaryn
- Department of Drug Metabolism and Pharmacokinetics, AbbVie Inc., 1 North Waukegan Rd., North Chicago, IL 60064, United States of America
| | - Christine Grinnell
- Department of Drug Metabolism and Pharmacokinetics, AbbVie Inc., 1 North Waukegan Rd., North Chicago, IL 60064, United States of America
| | - Ingeborg Dreher
- Department of Bioanalytics, AbbVie Deutschland GmbH & Co. KG, Neuroscience Research, Knollstrasse, 67061 Ludwigshafen, Germany
| | - Michael Gold
- Department of Neuroscience Development, AbbVie Inc., 1 North Waukegan Rd., North Chicago, IL 60064, United States of America
| | - Grégoire Courtine
- Center for Neuroprosthetics and Brain Mind Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Geneva, Switzerland; Department of Clinical Neuroscience, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland; Defitech Center for Interventional Neurotherapies, (NeuroRestore), CHUV/UNIL/EPFL, Lausanne, Switzerland
| | - Andrea Mothe
- Krembil Brain Institute, Toronto Western Hospital, University Health Network, Toronto, Canada
| | - Charles H Tator
- Division of Neurosurgery, Toronto Western Hospital, and University of Toronto, Toronto, Canada
| | - James D Guest
- Department of Neurosurgery and The Miami Project to Cure Paralysis, The Miller School of Medicine, University of Miami, Miami, FL, United States of America
| |
Collapse
|
34
|
Cui J, Fan J, Li H, Zhang J, Tong J. Neuroprotective potential of fisetin in an experimental model of spinal cord injury: via modulation of NF-κB/IκBα pathway. Neuroreport 2021; 32:296-305. [PMID: 33470764 PMCID: PMC7886366 DOI: 10.1097/wnr.0000000000001596] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Accepted: 12/08/2020] [Indexed: 01/19/2023]
Abstract
AIM To evaluate neuroprotective efficacy of fisetin against the experimental model of spinal cord injury (SCI). MATERIALS AND METHODS SCI was induced in male Sprague-Dawley rats by placing an aneurysm clip extradurally. Rats were treated either with vehicle or fisetin for 28 days after SCI. RESULTS Treatment with fisetin significantly attenuated SCI-induced alternations in mechano-tactile and thermal allodynia, hyperalgesia and nerve conduction velocities. SCI-induced upregulated tumor necrosis factor-alpha, interleukins, inducible nitric oxide synthase, cyclooxygenase-II, Bcl-2-associated X protein and caspase-3 mRNA expressions in the spinal cord and these were markedly reduced by fisetin. Spinal nuclear factor kappa B and nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor-alpha protein levels were also significantly downregulated by fisetin. Hematoxylin and eosin staining of spinal cord suggested that fisetin significantly ameliorated histological aberrations such as neuronal degeneration, necrosis and inflammatory infiltration induced in it. CONCLUSION Fisetin exerts neuroprotection via modulation of nuclear factor kappa B/nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor-alpha pathway by inhibiting release of inflammatory mediators (inducible nitric oxide synthase and cyclooxygenase-II), proinflammatory cytokines (tumor necrosis factor-alpha and interleukins), apoptotic mediators (Bcl-2-associated X protein and caspase-3).
Collapse
Affiliation(s)
| | - Jingshi Fan
- Department of Pathology, Baoding First Central Hospital, Baoding, Hebei Province, China
| | | | - Jinku Zhang
- Department of Pathology, Baoding First Central Hospital, Baoding, Hebei Province, China
| | | |
Collapse
|
35
|
Alishahi M, Anbiyaiee A, Farzaneh M, Khoshnam SE. Human Mesenchymal Stem Cells for Spinal Cord Injury. Curr Stem Cell Res Ther 2021; 15:340-348. [PMID: 32178619 DOI: 10.2174/1574888x15666200316164051] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Revised: 08/03/2019] [Accepted: 09/17/2019] [Indexed: 12/13/2022]
Abstract
Spinal Cord Injury (SCI), as a devastating and life-altering neurological disorder, is one of the most serious health issues. Currently, the management of acute SCI includes pharmacotherapy and surgical decompression. Both the approaches have been observed to have adverse physiological effects on SCI patients. Therefore, novel therapeutic targets for the management of SCI are urgently required for developing cell-based therapies. Multipotent stem cells, as a novel strategy for the treatment of tissue injury, may provide an effective therapeutic option against many neurological disorders. Mesenchymal stem cells (MSCs) or multipotent stromal cells can typically self-renew and generate various cell types. These cells are often isolated from bone marrow (BM-MSCs), adipose tissues (AD-MSCs), umbilical cord blood (UCB-MSCs), and placenta (PMSCs). MSCs have remarkable potential for the development of regenerative therapies in animal models and humans with SCI. Herein, we summarize the therapeutic potential of human MSCs in the treatment of SCI.
Collapse
Affiliation(s)
- Masoumeh Alishahi
- Department of Biology, Tehran North Branch, Islamic Azad University, Tehran, Iran
| | - Amir Anbiyaiee
- Department of Surgery, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz 61357-15794, Iran
| | - Maryam Farzaneh
- Physiology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Seyed E Khoshnam
- Physiology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
36
|
Kwiecien JM. The Pathogenesis of Neurotrauma Indicates Targets for Neuroprotective Therapies. Curr Neuropharmacol 2021; 19:1191-1201. [PMID: 33550977 PMCID: PMC8719295 DOI: 10.2174/1570159x19666210125153308] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 11/06/2020] [Accepted: 01/24/2021] [Indexed: 11/22/2022] Open
Abstract
The spinal cord injury (SCI) initiates an extraordinarily protracted disease with 3 phases; acute, inflammatory, and resolution that are restricted to the cavity of injury (COI) or arachnoiditis by a unique CNS reaction against the severity of destructive inflammation. While the severity of inflammation involving the white matter is fueled by a potently immunogenic activity of damaged myelin, its sequestration in the COI and its continuity with the cerebrospinal fluid of the subdural space allow anti-inflammatory therapeutics infused subdurally to inhibit phagocytic macrophage infiltration and thus provide neuroprotection. The role of astrogliosis in containing and ultimately in eliminating severe destructive inflammation post-trauma appears obvious but is not yet sufficiently understood to use in therapeutic neuroprotective and neuroregenerative strategies. An apparent antiinflammatory activity of reactive astrocytes is paralleled by their active role in removing excess edema fluid in blood-brain barrier damaged by inflammation. Recently elucidated pathogenesis of neurotrauma, including SCI, traumatic brain injury (TBI), and stroke, calls for the following principal therapeutic steps in its treatment leading to the recovery of neurologic function: (1) inhibition and elimination of destructive inflammation from the COI with accompanying reduction of vasogenic edema, (2) insertion into the COI of a functional bridge supporting the crossing of regenerating axons, (3) enabling regeneration of axons to their original synaptic targets by temporary safe removal of myelin in targeted areas of white matter, (4) in vivo, systematic monitoring of the consecutive therapeutic steps. The focus of this paper is on therapeutic step 1.
Collapse
Affiliation(s)
- Jacek M. Kwiecien
- Department of Pathology and Molecular Medicine, Faculty of Health Sciences, McMaster University, Room HSC 1U22D, 1280 Main Street West, Hamilton, ON, L4S 4K1, Canada
| |
Collapse
|
37
|
Takami T, Shimokawa N, Parthiban J, Zileli M, Ali S. Pharmacologic and Regenerative Cell Therapy for Spinal Cord Injury: WFNS Spine Committee Recommendations. Neurospine 2020; 17:785-796. [PMID: 33401856 PMCID: PMC7788403 DOI: 10.14245/ns.2040408.204] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 09/15/2020] [Indexed: 12/17/2022] Open
Abstract
This is a review article examining the pharmacologic and regenerative cell therapy for spinal cord injury. A literature search during last 10 years were conducted using key words. Case reports, experimental (nonhuman) studies, papers other than English language were excluded. Up-to-date information on the pharmacologic and regenerative cell therapy for spinal cord injury was reviewed and statements were produced to reach a consensus in 2 separate consensus meeting of WFNS Spine Committee. The statements were voted and reached a consensus using Delphi method. Pharmacologic and regenerative cell therapy for spinal cord injury have long been an interest of many experimental and clinical researches. Clinical studies with methylpredinisolone have not shown clear cut benefit. Other drugs such as Rho inhibitor, minocycline, riluzole, granulocyte colony-stimulating factor have also been tried without significant benefits. Regenerative cell therapy using different types of stem cells, different inoculation techniques, and scaffolds have undergone many trials highlighting the efficacies of cells and their limitations. This review article summarizes the current knowledge on pharmacologic and regenerative cell therapy for spinal cord injury. Unfortunately, there is a need for further experimental and human trials to recommend effective pharmacologic and regenerative cell therapy.
Collapse
Affiliation(s)
- Toshihiro Takami
- Department of Neurosurgery, Osaka City University Graduate School of Medicine, Osaka, Japan
| | | | - Jutty Parthiban
- Department of Neurosurgery, Kovai Medical Center and Hospital Coimbatore, Tamilnadu, India
| | - Mehmet Zileli
- Department of Neurosurgery, Ege University, Izmir, Turkey
| | - Sheena Ali
- Department of Neurosurgery, Kovai Medical Center and Hospital Coimbatore, Tamilnadu, India
| |
Collapse
|
38
|
Farzaneh M, Anbiyaiee A, Khoshnam SE. Human Pluripotent Stem Cells for Spinal Cord Injury. Curr Stem Cell Res Ther 2020; 15:135-143. [PMID: 31656156 DOI: 10.2174/1574362414666191018121658] [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: 06/08/2019] [Revised: 07/16/2019] [Accepted: 09/17/2019] [Indexed: 12/27/2022]
Abstract
Spinal cord injury (SCI) as a serious public health issue and neurological insult is one of the most severe cause of long-term disability. To date, a variety of techniques have been widely developed to treat central nervous system injury. Currently, clinical treatments are limited to surgical decompression and pharmacotherapy. Because of their negative effects and inefficiency, novel therapeutic approaches are required in the management of SCI. Improvement and innovation of stem cell-based therapies have a huge potential for biological and future clinical applications. Human pluripotent stem cells (hPSCs) including embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) are defined by their abilities to divide asymmetrically, self-renew and ultimately differentiate into various cell lineages. There are considerable research efforts to use various types of stem cells, such as ESCs, neural stem cells (NSCs), and mesenchymal stem cells (MSCs) in the treatment of patients with SCI. Moreover, the use of patient-specific iPSCs holds great potential as an unlimited cell source for generating in vivo models of SCI. In this review, we focused on the potential of hPSCs in treating SCI.
Collapse
Affiliation(s)
- Maryam Farzaneh
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Amir Anbiyaiee
- Department of Obstetrics and Gynecology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz 61357-15794, Iran
| | - Seyed Esmaeil Khoshnam
- Physiology Research Center, Department of Physiology, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
39
|
Abbas WA, Ibrahim ME, El-Naggar M, Abass WA, Abdullah IH, Awad BI, Allam NK. Recent Advances in the Regenerative Approaches for Traumatic Spinal Cord Injury: Materials Perspective. ACS Biomater Sci Eng 2020; 6:6490-6509. [PMID: 33320628 DOI: 10.1021/acsbiomaterials.0c01074] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Spinal cord injury (SCI) is a devastating health condition that may lead to permanent disabilities and death. Understanding the pathophysiological perspectives of traumatic SCI is essential to define mechanisms that can help in designing recovery strategies. Since central nervous system tissues are notorious for their deficient ability to heal, efforts have been made to identify solutions to aid in restoration of the spinal cord tissues and thus its function. The two main approaches proposed to address this issue are neuroprotection and neuro-regeneration. Neuroprotection involves administering drugs to restore the injured microenvironment to normal after SCI. As for the neuro-regeneration approach, it focuses on axonal sprouting for functional recovery of the injured neural tissues and damaged axons. Despite the progress made in the field, neural regeneration treatment after SCI is still unsatisfactory owing to the disorganized way of axonal growth and extension. Nanomedicine and tissue engineering are considered promising therapeutic approaches that enhance axonal growth and directionality through implanting or injecting of the biomaterial scaffolds. One of these recent approaches is nanofibrous scaffolds that are used to provide physical support to maintain directional axonal growth in the lesion site. Furthermore, these preferable tissue-engineered substrates can afford axonal regeneration by mimicking the extracellular matrix of the neural tissues in terms of biological, chemical, and architectural characteristics. In this review, we discuss the regenerative approach using nanofibrous scaffolds with a focus on their fabrication methods and their properties that define their functionality performed to heal the neural tissue efficiently.
Collapse
Affiliation(s)
- Walaa A Abbas
- Energy Materials Laboratory, School of Sciences and Engineering, The American University in Cairo, New Cairo 11835, Egypt
| | - Maha E Ibrahim
- Department of Physical Medicine, Rheumatology and Rehabilitation, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Manar El-Naggar
- Energy Materials Laboratory, School of Sciences and Engineering, The American University in Cairo, New Cairo 11835, Egypt
| | - Wessam A Abass
- Center of Sustainable Development, School of Sciences and Engineering, The American University in Cairo, New Cairo 11835, Egypt
| | - Ibrahim H Abdullah
- Energy Materials Laboratory, School of Sciences and Engineering, The American University in Cairo, New Cairo 11835, Egypt
| | - Basem I Awad
- Mansoura Experimental Research Center (MERC), Department of Neurological Surgery, School of Medicine, Mansoura University, Mansoura, Egypt
| | - Nageh K Allam
- Energy Materials Laboratory, School of Sciences and Engineering, The American University in Cairo, New Cairo 11835, Egypt
| |
Collapse
|
40
|
Gonzalez-Ruiz C, Cordero-Anguiano P, Morales-Guadarrama A, Mondragón-Lozano R, Sánchez-Torres S, Salgado-Ceballos H, Villarreal F, Meaney E, Ceballos G, Nájera N. (-)-Epicatechin reduces muscle waste after complete spinal cord transection in a murine model: role of ubiquitin-proteasome system. Mol Biol Rep 2020; 47:8975-8985. [PMID: 33151476 DOI: 10.1007/s11033-020-05954-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 10/28/2020] [Indexed: 12/17/2022]
Abstract
The skeletal muscle mass reduces 30-60% after spinal cord injury, this is mostly due to protein degradation through ubiquitin-proteasome system. In this work, we propose that the flavanol (-)-epicatechin, due its widespread biological effects on muscle health, can prevent muscle mass decrease after spinal cord injury. Thirty-six female Long Evans rats were randomized into 5 groups: (1) Spinal cord injury 7 days, (2) Spinal cord injury + (-)-epicatechin 7 days, (3) Spinal cord injury 30 days, (4) Spinal cord injury + (-)-epicatechin 30 days and (5) Sham (Only laminectomy). Hind limb perimeter, muscle cross section area, fiber cross section area and ubiquitin-proteasome system protein expression together with total protein ubiquitination were assessed. At 30 days Spinal cord injury group lost 49.52 ± 2.023% of muscle cross section area (-)-epicatechin treated group lost only 24.28 ± 15.45% being a significant difference. Ubiquitin-proteasome markers showed significant changes. FOXO1a increased in spinal cord injury group vs Sham (-)-epicatechin reduced this increase. In spinal cord injury group MAFbx increased significantly vs Sham but decrease in (-)-epicatechin treatment group at 30 days. At 7 and 30 days MuRF1 increased in the spinal cord injury and decreased in the (-)-epicatechin group. The global protein ubiquitination increases after spinal cord injury, epicatechin treatment induce a significant decrease in protein ubiquitination. These results suggest that (-)-epicatechin reduces the muscle waste after spinal cord injury through down regulation of the ubiquitin-proteasome system.
Collapse
Affiliation(s)
- Cristian Gonzalez-Ruiz
- Laboratorio de Investigación Integral Cardiometabólica, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico, Mexico
| | - Paola Cordero-Anguiano
- Laboratorio de Investigación Integral Cardiometabólica, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico, Mexico
| | - Axayacatl Morales-Guadarrama
- Centro Nacional de Investigación en Imagenología e Instrumentación Médica, Departamento de Ingeniería Eléctrica, Universidad Autónoma Metropolitana Iztapalapa, Mexico, Mexico
| | - Rodrigo Mondragón-Lozano
- Consejo Nacional de Ciencia y Tecnología, Unidad de Investigación Médica en Enfermedades Neurológicas, Hospital de Especialidades Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico, Mexico
| | - Stephanie Sánchez-Torres
- División de Ciencias Biológicas y de la Salud, Posgrado en Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana, Unidad Iztapalapa, Mexico, Mexico
| | - Hermelinda Salgado-Ceballos
- Unidad de Investigación Médica en Enfermedades Neurológicas, Hospital de Especialidades Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico, Mexico
| | | | - Eduardo Meaney
- Laboratorio de Investigación Integral Cardiometabólica, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico, Mexico
| | - Guillermo Ceballos
- Laboratorio de Investigación Integral Cardiometabólica, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico, Mexico.
| | - Nayelli Nájera
- Laboratorio de Investigación Integral Cardiometabólica, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico, Mexico.
| |
Collapse
|
41
|
Vawda R, Badner A, Hong J, Mikhail M, Dragas R, Xhima K, Jose A, Fehlings MG. Harnessing the Secretome of Mesenchymal Stromal Cells for Traumatic Spinal Cord Injury: Multicell Comparison and Assessment of In Vivo Efficacy. Stem Cells Dev 2020; 29:1429-1443. [PMID: 32962528 PMCID: PMC7703247 DOI: 10.1089/scd.2020.0079] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Cell therapy offers significant promise for traumatic spinal cord injury (SCI), which despite many medical advances, has limited treatment strategies. Able to address the multifactorial and dynamic pathophysiology of SCI, cells present various advantages over standard pharmacological approaches. However, the use of live cells is also severely hampered by logistical and practical considerations. These include specialized equipment and expertise, standardization of cell stocks, sustained cell viability post-thawing, and cryopreservation-induced delayed-onset cell death. For this reason, we suggest a novel and clinically translatable alternative to live-cell systemic infusion, which retains the efficacy of the latter while overcoming many of its limitations. This strategy involves the administration of concentrated cell secretome and exploits the trophic mechanism by which stromal cells function. In this study, we compare the efficacy of intravenously delivered concentrated conditioned media (CM) from human umbilical cord matrix cells (HUCMCs), bone marrow mesenchymal stromal cells, as well as newborn and adult fibroblasts in a rat model of moderately severe cervical clip compression/contusion injury (C7--T1, 35 g). This is further paired with a thorough profile of the CM cytokines, chemokines, and angiogenic factors. The HUCMC-derived CM was most effective at limiting acute (48 h post-SCI) vascular pathology, specifically lesion volume, and functional vascularity. Principle component analysis (PCA), hierarchical clustering, and interaction analysis of proteins highly expressed in the HUCMC secretome suggest involvement of the MAPK/ERK, JAK/STAT, and immune cell migratory pathways. This "secretotherapeutic" strategy represents a novel and minimally invasive method to target multiple organ systems and several pathologies shortly after traumatic SCI.
Collapse
Affiliation(s)
- Reaz Vawda
- Department of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Canada
| | - Anna Badner
- Department of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Canada.,Institute of Medical Science, School of Medicine, University of Toronto, Toronto, Canada
| | - James Hong
- Department of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Canada.,Institute of Medical Science, School of Medicine, University of Toronto, Toronto, Canada
| | - Mirriam Mikhail
- Department of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Canada
| | - Rachel Dragas
- Department of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Canada.,Institute of Medical Science, School of Medicine, University of Toronto, Toronto, Canada
| | - Kristiana Xhima
- Department of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Canada
| | - Alejandro Jose
- Department of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Canada
| | - Michael G Fehlings
- Department of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Canada.,Institute of Medical Science, School of Medicine, University of Toronto, Toronto, Canada.,Spinal Program, Toronto Western Hospital, University Health Network, Toronto, Canada
| |
Collapse
|
42
|
Afshari K, Momeni Roudsari N, Lashgari NA, Haddadi NS, Haj-Mirzaian A, Hassan Nejad M, Shafaroodi H, Ghasemi M, Dehpour AR, Abdolghaffari AH. Antibiotics with therapeutic effects on spinal cord injury: a review. Fundam Clin Pharmacol 2020; 35:277-304. [PMID: 33464681 DOI: 10.1111/fcp.12605] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 08/06/2020] [Accepted: 09/08/2020] [Indexed: 12/17/2022]
Abstract
Accumulating evidence indicates that a considerable number of antibiotics exert anti-inflammatory and neuroprotective effects in different central and peripheral nervous system diseases including spinal cord injury (SCI). Both clinical and preclinical studies on SCI have found therapeutic effects of antibiotics from different families on SCI. These include macrolides, minocycline, β-lactams, and dapsone, all of which have been found to improve SCI sequels and complications. These antibiotics may target similar signaling pathways such as reducing inflammatory microglial activity, promoting autophagy, inhibiting neuronal apoptosis, and modulating the SCI-related mitochondrial dysfunction. In this review paper, we will discuss the mechanisms underlying therapeutic effects of these antibiotics on SCI, which not only could supply vital information for investigators but also guide clinicians to consider administering these antibiotics as part of a multimodal therapeutic approach for management of SCI and its complications.
Collapse
Affiliation(s)
- Khashayar Afshari
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, 1419733141, Iran.,Experimental Medicine Research Center, Tehran University of Medical Sciences, P.O. Box 13145-784, Tehran, Iran.,Department of Dermatology, University of Massachusetts Medical School, Worcester, MA, 01655, USA
| | - Nazanin Momeni Roudsari
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, No. 99, Yakhchal, Gholhak, Shariati St., Tehran, P. O. Box: 19419-33111, Iran
| | - Naser-Aldin Lashgari
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, No. 99, Yakhchal, Gholhak, Shariati St., Tehran, P. O. Box: 19419-33111, Iran
| | - Nazgol-Sadat Haddadi
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, 1419733141, Iran.,Experimental Medicine Research Center, Tehran University of Medical Sciences, P.O. Box 13145-784, Tehran, Iran.,Department of Dermatology, University of Massachusetts Medical School, Worcester, MA, 01655, USA
| | - Arvin Haj-Mirzaian
- Experimental Medicine Research Center, Tehran University of Medical Sciences, P.O. Box 13145-784, Tehran, Iran
| | - Malihe Hassan Nejad
- Department of Infectious Diseases, Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran, 1419733141, Iran
| | - Hamed Shafaroodi
- Experimental Medicine Research Center, Tehran University of Medical Sciences, P.O. Box 13145-784, Tehran, Iran
| | - Mehdi Ghasemi
- Department of Neurology, University of Massachusetts School of Medicine, Worcester, MA, 01655, USA
| | - Ahmad Reza Dehpour
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, 1419733141, Iran.,Experimental Medicine Research Center, Tehran University of Medical Sciences, P.O. Box 13145-784, Tehran, Iran
| | - Amir Hossein Abdolghaffari
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, No. 99, Yakhchal, Gholhak, Shariati St., Tehran, P. O. Box: 19419-33111, Iran.,Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Karaj, 31375-1369, Iran.,Gastrointestinal Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, 1419733151, Iran
| |
Collapse
|
43
|
Kwiecien JM, Dabrowski W, Kwiecien-Delaney BJ, Kwiecien-Delaney CJ, Siwicka-Gieroba D, Yaron JR, Zhang L, Delaney KH, Lucas AR. Neuroprotective Effect of Subdural Infusion of Serp-1 in Spinal Cord Trauma. Biomedicines 2020; 8:E372. [PMID: 32977430 PMCID: PMC7598159 DOI: 10.3390/biomedicines8100372] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 09/09/2020] [Accepted: 09/21/2020] [Indexed: 12/16/2022] Open
Abstract
Spinal cord injury (SCI) initiates a severe, destructive inflammation with pro-inflammatory, CD68+/CD163-, phagocytic macrophages infiltrating the area of necrosis and hemorrhage by day 3 and persisting for the next 16 weeks. Inhibition of macrophage infiltration of the site of necrosis that is converted into a cavity of injury (COI) during the first week post-SCI, should limit inflammatory damage, shorten its duration and result in neuroprotection. By sustained subdural infusion we administered Serp-1, a Myxoma virus-derived immunomodulatory protein previously shown to improve neurologic deficits and inhibit macrophage infiltration in the COI in rats with the balloon crush SCI. Firstly, in a 7 day long study, we determined that the optimal dose for macrophage inhibition was 0.2 mg/week. Then, we demonstrated that a continuous subdural infusion of Serp-1 for 8 weeks resulted in consistently accelerated lowering of pro-inflammatory macrophages in the COI and in their almost complete elimination similar to that previously observed at 16 weeks in untreated SCI rats. The macrophage count in the COI is a quantitative test directly related to the severity of destructive inflammation initiated by the SCI. This test has consistently demonstrated anti-inflammatory effect of Serp-1 interpreted as neuroprotection, the first and necessary step in a therapeutic strategy in neurotrauma.
Collapse
Affiliation(s)
- Jacek M. Kwiecien
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada;
| | - Wojciech Dabrowski
- Department of Anaesthesiology and Intensive Therapy, Medical University of Lublin, 20-090 Lublin, Poland; (W.D.); (D.S.-G.)
| | | | | | - Dorota Siwicka-Gieroba
- Department of Anaesthesiology and Intensive Therapy, Medical University of Lublin, 20-090 Lublin, Poland; (W.D.); (D.S.-G.)
| | - Jordan R. Yaron
- Center for Personalized Diagnostics and Center for Immunotherapy, Vaccines and Virotherapy, Biodesign Institute, Arizona State University, Tempe, AZ 85287, USA; (J.R.Y.); (L.Z.); (A.R.L.)
| | - Liqiang Zhang
- Center for Personalized Diagnostics and Center for Immunotherapy, Vaccines and Virotherapy, Biodesign Institute, Arizona State University, Tempe, AZ 85287, USA; (J.R.Y.); (L.Z.); (A.R.L.)
| | - Kathleen H. Delaney
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada;
| | - Alexandra R. Lucas
- Center for Personalized Diagnostics and Center for Immunotherapy, Vaccines and Virotherapy, Biodesign Institute, Arizona State University, Tempe, AZ 85287, USA; (J.R.Y.); (L.Z.); (A.R.L.)
| |
Collapse
|
44
|
Li X, Yu Z, Zong W, Chen P, Li J, Wang M, Ding F, Xie M, Wang W, Luo X. Deficiency of the microglial Hv1 proton channel attenuates neuronal pyroptosis and inhibits inflammatory reaction after spinal cord injury. J Neuroinflammation 2020; 17:263. [PMID: 32891159 PMCID: PMC7487532 DOI: 10.1186/s12974-020-01942-x] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Accepted: 08/25/2020] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND Spinal cord injury (SCI) causes neurological dysfunction with devastating consequences. SCI pathogenesis is accompanied by inflammasome activation and neuronal damage. But the spatial pattern and the time course of neuronal pyroptosis and apoptosis after SCI should be further elucidated. The microglial voltage-gated proton channel (Hv1) is implicated in reactive oxygen species (ROS)-induced neuronal damage following ischemic stroke. However, there is a lack of quantification on the neuronal pyroptosis and apoptosis associated with microglial Hv1 after SCI. METHODS We analyzed spatial and temporal characteristics of neuronal pyroptosis and apoptosis following SCI and investigated the effects of Hv1 deficiency on neuronal pyroptosis and the nod-like receptor 3 (NLRP3) inflammasome pathway by using a mouse model of SCI. We tested the effects of Hv1-deficient microglia on ROS production in vivo and examined the relationship between ROS and neuronal pyroptosis in vitro. RESULTS We observed that apoptosis was detected closer to the injury core than pyroptosis. The incidence of neuronal apoptosis peaked on day 1 after SCI and occurred before pyroptosis. Hv1 deficiency reduced neuronal apoptosis and NLRP3-inflammasome-mediated pyroptosis, improved axonal regeneration, and reduced motor deficits. SCI led to elevated ROS levels, whereas Hv1 deficiency downregulated microglial ROS generation. In vitro, ROS upregulated neuronal pyroptosis and activated the NLRP3 inflammasome pathway, both of which were reversed by addition of a ROS scavenger. Our results suggested that microglial Hv1 regulated neuronal apoptosis and NLRP3-induced neuronal pyroptosis after SCI by mediating ROS production. CONCLUSION Following SCI, neuronal pyroptosis lasted longer and occurred farther away from the injury core compared with that of neuronal apoptosis. Microglial Hv1 deficiency downregulated microglial ROS generation and reduced apoptosis and NLRP3-induced neuronal pyroptosis. Our findings may provide novel insights into Hv1-associated mechanisms underlying neuronal damage after SCI.
Collapse
Affiliation(s)
- Xuefei Li
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zhiyuan Yu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Weifeng Zong
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Peng Chen
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jia Li
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Minghuan Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Fengfei Ding
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Minjie Xie
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Wei Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiang Luo
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
45
|
Yu Z, Li H, Xia P, Kong W, Chang Y, Fu C, Wang K, Yang X, Qi Z. Application of fibrin-based hydrogels for nerve protection and regeneration after spinal cord injury. J Biol Eng 2020; 14:22. [PMID: 32774454 PMCID: PMC7397605 DOI: 10.1186/s13036-020-00244-3] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 07/27/2020] [Indexed: 12/13/2022] Open
Abstract
Traffic accidents, falls, and many other events may cause traumatic spinal cord injuries (SCIs), resulting in nerve cells and extracellular matrix loss in the spinal cord, along with blood loss, inflammation, oxidative stress (OS), and others. The continuous development of neural tissue engineering has attracted increasing attention on the application of fibrin hydrogels in repairing SCIs. Except for excellent biocompatibility, flexibility, and plasticity, fibrin, a component of extracellular matrix (ECM), can be equipped with cells, ECM protein, and various growth factors to promote damage repair. This review will focus on the advantages and disadvantages of fibrin hydrogels from different sources, as well as the various modifications for internal topographical guidance during the polymerization. From the perspective of further improvement of cell function before and after the delivery of stem cell, cytokine, and drug, this review will also evaluate the application of fibrin hydrogels as a carrier to the therapy of nerve repair and regeneration, to mirror the recent development tendency and challenge.
Collapse
Affiliation(s)
- Ziyuan Yu
- Department of Orthopedic Surgery, The Second Hospital of Jilin University, Ziqiang Street No. 218, Changchun, TX 130041 PR China
| | - Hongru Li
- Department of Orthopedic Surgery, The Second Hospital of Jilin University, Ziqiang Street No. 218, Changchun, TX 130041 PR China
| | - Peng Xia
- Department of Orthopedic Surgery, The Second Hospital of Jilin University, Ziqiang Street No. 218, Changchun, TX 130041 PR China
| | - Weijian Kong
- Department of Orthopedic Surgery, The Second Hospital of Jilin University, Ziqiang Street No. 218, Changchun, TX 130041 PR China
| | - Yuxin Chang
- Department of Orthopedic Surgery, The Second Hospital of Jilin University, Ziqiang Street No. 218, Changchun, TX 130041 PR China
| | - Chuan Fu
- Department of Orthopedic Surgery, The Second Hospital of Jilin University, Ziqiang Street No. 218, Changchun, TX 130041 PR China
| | - Kai Wang
- Department of Orthopedic Surgery, The Second Hospital of Jilin University, Ziqiang Street No. 218, Changchun, TX 130041 PR China
| | - Xiaoyu Yang
- Department of Orthopedic Surgery, The Second Hospital of Jilin University, Ziqiang Street No. 218, Changchun, TX 130041 PR China
| | - Zhiping Qi
- Department of Orthopedic Surgery, The Second Hospital of Jilin University, Ziqiang Street No. 218, Changchun, TX 130041 PR China
| |
Collapse
|
46
|
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: 146] [Impact Index Per Article: 36.5] [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
|
47
|
The effects of mouse strain and age on a model of unilateral cervical contusion spinal cord injury. PLoS One 2020; 15:e0234245. [PMID: 32542053 PMCID: PMC7295191 DOI: 10.1371/journal.pone.0234245] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 05/21/2020] [Indexed: 12/30/2022] Open
Abstract
There are approximately 1.2 million people currently living with spinal cord injury (SCI), with a majority of cases at the cervical level and half involving incomplete injuries. Yet, as most preclinical research has been focused on bilateral thoracic models, there remains a disconnect between bench and bedside that limits translational success. Here, we profile a clinically relevant model of unilateral cervical contusion injury in the mouse (30kD with 0, 2, 5, or 10 second dwell time). We demonstrate sustained behavioral deficits in performance on grip strength, cylinder reaching, horizontal ladderbeam and CatWalk automated gait analysis tasks. Beyond highlighting reliable parameters for injury assessment, we also explored the effect of mouse strain and age on injury outcome, including evaluation of constitutively immunodeficient mice relevant for neurotransplantation and cellular therapy testing. Comparison of C57Bl/6 and immunodeficient Rag2gamma(c)-/- as well as Agouti SCIDxRag2Gamma(c)-/- hybrid mouse strains revealed fine differences in post-injury ipsilateral grip strength as well as total number of rearings on the cylinder task. Differences in post-SCI contralateral forepaw duty cycle and regularity index as measured by CatWalk gait analysis between the two immunodeficient strains were also observed. Further, assessment of young (3–4 months old) and aging (16–17 months old) Rag2gamma(c)-/- mice identified age-related pre-injury differences in strength and rearing that were largely masked following cervical contusion injury; observations that may help interpret previous results in aged rodents as well as human clinical trials. Collectively, the work provides useful insight for experimental design and analysis of future pre-clinical studies in a translational unilateral cervical contusion injury model.
Collapse
|
48
|
CORM-2-Solid Lipid Nanoparticles Maintain Integrity of Blood-Spinal Cord Barrier After Spinal Cord Injury in Rats. Mol Neurobiol 2020; 57:2671-2689. [DOI: 10.1007/s12035-020-01914-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Accepted: 03/30/2020] [Indexed: 12/21/2022]
|
49
|
Neuroprotective Agents as an Adjuvant Treatment in Patients With Acute Spinal Cord Injuries: A Qualitative Systematic Review of Randomized Trials. Clin Spine Surg 2020; 33:65-75. [PMID: 31404015 DOI: 10.1097/bsd.0000000000000861] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
STUDY DESIGN This was a systematic literature review. OBJECTIVE The objective of this study was to evaluate randomized clinical trials that address potential neuroprotective agents used to improve neurological outcome in patients with spinal cord injury (SCI). SUMMARY OF BACKGROUND DATA Clinical treatment of acute SCI has evolved significantly, but neurological recovery of severely injured patients remains modest. Neuroprotective agents may act to limit secondary damage in the sequence of pathophysiologic insults that occur after primary SCI. METHODS We performed a systematic review in accordance with Preferred Reporting Items for Systematic Reviews and Meta-analysis (PRISMA) guidelines of all clinical randomized trials that evaluated potential neuroprotective agents (drugs, stem cells, and any type of medicative interventions) in neurological outcome of acute SCI. All the studies were graded according to their level of evidence in accordance with the Oxford Level of Evidence-based Medicine. RESULTS A total of 16 randomized clinical trials were included and fully analyzed in our review. The following 12 substances/drugs were analyzed: methylprednisolone (MP), naloxone, tirilizad, nimodipine, Sygen, autologous incubated macrophages, autologous bone marrow cells, minocycline, erythropoietin, ganglioside, vitamin D, and progesterone. Modest benefits were attributed to minocycline and Sygen (without statistical significance), and some benefits were obtained with erythropoietin and progesterone plus vitamin D in neurological outcome. For MP, the benefits are also controversial and may be attributed to statistical artifacts and with a high risk of adverse effects. The other substances did not change the final outcome. All studies were considered as grade B of recommendation (100%) and levels of evidences as B2 (81.25%) and B3 (18.75%). CONCLUSIONS Our review reported some potential substances that may improve neurological outcome in acute SCI: MP, vitamin D associated with progesterone, and erythropoietin. Their potential benefits were modest in the evaluated studies, requiring further randomized clinical trials with large samples of patients, without statistical artifacts, for routine clinical use. Furthermore, potential adverse effects must be considered with the use of neuroprotective agents in SCI. Until then, the use of these substances may be experimental or restricted to specific clinical situations.
Collapse
|
50
|
Lindsay SL, McCanney GA, Willison AG, Barnett SC. Multi-target approaches to CNS repair: olfactory mucosa-derived cells and heparan sulfates. Nat Rev Neurol 2020; 16:229-240. [PMID: 32099190 DOI: 10.1038/s41582-020-0311-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/13/2020] [Indexed: 02/06/2023]
Abstract
Spinal cord injury (SCI) remains one of the biggest challenges in the development of neuroregenerative therapeutics. Cell transplantation is one of numerous experimental strategies that have been identified and tested for efficacy at both preclinical and clinical levels in recent years. In this Review, we briefly discuss the state of human olfactory cell transplantation as a therapy, considering both its current clinical status and its limitations. Furthermore, we introduce a mesenchymal stromal cell derived from human olfactory tissue, which has the potential to induce multifaceted reparative effects in the environment within and surrounding the lesion. We argue that no single therapy will be sufficient to treat SCI effectively and that a combination of cell-based, rehabilitation and pharmaceutical interventions is the most promising approach to aid repair. For this reason, we also introduce a novel pharmaceutical strategy based on modifying the activity of heparan sulfate, an important regulator of a wide range of biological cell functions. The multi-target approach that is exemplified by these types of strategies will probably be necessary to optimize SCI treatment.
Collapse
Affiliation(s)
- Susan L Lindsay
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - George A McCanney
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Alice G Willison
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Susan C Barnett
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK.
| |
Collapse
|