1
|
Zipser CM, Curt A. Disease-specific interventions using cell therapies for spinal cord disease/injury. HANDBOOK OF CLINICAL NEUROLOGY 2024; 205:263-282. [PMID: 39341658 DOI: 10.1016/b978-0-323-90120-8.00007-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Traumatic spinal cord injury (SCI) may occur across the lifespan and is of global relevance. Damage of the spinal cord results in para- or tetraplegia and is associated with neuropathic pain, spasticity, respiratory, and autonomic dysfunction (i.e., control of bladder-bowel function). While the acute surgical treatment aims at stabilizing the spine and decompressing the damaged spinal cord, SCI patients require neurorehabilitation to restore neural function and to compensate for any impairments including motor disability, pain treatment, and bladder/bowel management. However, the spinal cord has a limited capacity to regenerate and much of the disability may persist, depending on the initial lesion severity and level of injury. For this reason, and the lack of effective drug treatments, there is an emerging interest and urgent need in promoting axonal regeneration and remyelination after SCI through cell- and stem-cell based therapies. This review briefly summarizes the state-of the art management of acute SCI and its neurorehabilitation to critically appraise phase I/II trials from the last two decades that have investigated cell-based therapies (i.e., Schwann cells, macrophages, and olfactory ensheathing cells) and stem cell-based therapies (i.e., neural stem cells, mesenchymal, and hematopoietic stem cells). Recently, two large multicenter trials provided evidence for the safety and feasibility of neural stem cell transplantation into the injured cord, whilst two monocenter trials also showed this to be the case for the transplantation of Schwann cells into the posttraumatic cord cavity. These are milestone studies that will facilitate further interventional trials. However, the clinical adoption of such approaches remains unproven, as there is only limited encouraging data, often in single patients, and no proven trial evidence to support regulatory approval.
Collapse
Affiliation(s)
- Carl Moritz Zipser
- Spinal Cord Injury Center, Balgrist University Hospital, Zurich, Switzerland
| | - Armin Curt
- Spinal Cord Injury Center, Balgrist University Hospital, Zurich, Switzerland.
| |
Collapse
|
2
|
Ji R, Hao Z, Wang H, Li X, Duan L, Guan F, Ma S. Application of Injectable Hydrogels as Delivery Systems in Spinal Cord Injury. Gels 2023; 9:907. [PMID: 37998998 PMCID: PMC10670785 DOI: 10.3390/gels9110907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 11/11/2023] [Accepted: 11/13/2023] [Indexed: 11/25/2023] Open
Abstract
Spinal cord injury (SCI) is a severe neurological injury caused by traffic accidents, trauma, or falls, which leads to significant loss of sensory, motor, and autonomous functions and seriously affects the patient's life quality. Although considerable progress has been made in mitigating secondary injury and promoting the regeneration/repair of SCI, the therapeutic effects need to be improved due to drug availability. Given their good biocompatibility, biodegradability, and low immunogenicity, injectable hydrogels can be used as delivery systems to achieve controlled release of drugs and other substances (cells and proteins, etc.), offering new hope for SCI repair. In this article, we summarized the types of injectable hydrogels, analyzed their application as delivery systems in SCI, and further discussed the mechanisms of hydrogels in the treatment of SCI, such as anti-inflammatory, antioxidant, anti-apoptosis, and pro-neurogenesis. Moreover, we highlighted the potential benefits of hydrogels in the treatment of SCI in combination with therapies, including the recent advances and achievements of these promising tools. Our review may offer new strategies for the development of SCI treatments based on injectable hydrogels as delivery systems.
Collapse
Affiliation(s)
| | | | | | | | | | - Fangxia Guan
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China; (R.J.); (Z.H.); (H.W.); (X.L.); (L.D.)
| | - Shanshan Ma
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China; (R.J.); (Z.H.); (H.W.); (X.L.); (L.D.)
| |
Collapse
|
3
|
Dong W, Peng Q, Liu Z, Xie Z, Guo X, Li Y, Chen C. Estrogen plays an important role by influencing the NLRP3 inflammasome. Biomed Pharmacother 2023; 167:115554. [PMID: 37738797 DOI: 10.1016/j.biopha.2023.115554] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 09/16/2023] [Accepted: 09/18/2023] [Indexed: 09/24/2023] Open
Abstract
The nucleotide-binding oligomerization domain-like receptor protein 3 (NLRP3) inflammasome is an important part of the natural immune system that plays an important role in many diseases. Estrogen is a sex hormone that plays an important role in controlling reproduction and regulates many physiological and pathological processes. Recent studies have indicated that estrogen is associated with disease progression. Estrogen can ameliorate some diseases (e. g, sepsis, mood disturbances, cerebral ischemia, some hepatopathy, Parkinson's disease, amyotrophic lateral sclerosis, inflammatory bowel disease, spinal cord injury, multiple sclerosis, myocardial ischemia/reperfusion injury, osteoarthritis, and renal fibrosis) by inhibiting the NLRP3 inflammasome. Estrogen can also promote the development of diseases (e.g., ovarian endometriosis, dry eye disease, and systemic lupus erythematosus) by upregulating the NLRP3 inflammasome. In addition, estrogen has a dual effect on the development of cancers and asthma. However, the mechanism of these effects is not summarized. This article reviewed the progress in understanding the effects of estrogen on the NLRP3 inflammasome and its mechanisms in recent years to provide a theoretical basis for an in-depth study.
Collapse
Affiliation(s)
- Wanglin Dong
- Institute of Nursing and Health, College of Nursing and Health, Henan University, Kaifeng, Henan, China
| | - Qianwen Peng
- Institute of Nursing and Health, College of Nursing and Health, Henan University, Kaifeng, Henan, China
| | - Zhuoxin Liu
- Clinical College of Medicine, Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Zhenxing Xie
- School of Basic Medical Science, Henan University, Jinming Avenue, Kaifeng, Henan 475004, China.
| | - Xiajun Guo
- Institute of Nursing and Health, College of Nursing and Health, Henan University, Kaifeng, Henan, China
| | - Yuanyuan Li
- Institute of Nursing and Health, College of Nursing and Health, Henan University, Kaifeng, Henan, China
| | - Chaoran Chen
- Institute of Nursing and Health, College of Nursing and Health, Henan University, Kaifeng, Henan, China.
| |
Collapse
|
4
|
Scholpa NE. Role of DNA methylation during recovery from spinal cord injury with and without β 2-adrenergic receptor agonism. Exp Neurol 2023; 368:114494. [PMID: 37488045 DOI: 10.1016/j.expneurol.2023.114494] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 07/14/2023] [Accepted: 07/20/2023] [Indexed: 07/26/2023]
Abstract
Daily treatment with the FDA-approved β2-adrenergic receptor agonist formoterol beginning 8 h after severe spinal cord injury (SCI) induces mitochondrial biogenesis and improves recovery in mice. We observed decreased DNA methyltransferase (DNMT) expression, global DNA methylation and methylation of the mitochondrial genes PGC-1α and NDUFS1 in the injury site of formoterol-treated mice 1 DPI, but this effect was lost by 7 DPI. To investigate the role of DNA methylation on recovery post-SCI, injured mice were treated daily with formoterol or vehicle, plus the DNMT inhibitor decitabine (DAC) on days 7-9. While DAC had no apparent effect on formoterol-induced recovery, mice treated with vehicle plus DAC exhibited increased BMS scores compared to vehicle alone beginning 15 DPI, reaching a degree of functional recovery similar to that of formoterol-treated mice by 21 DPI. Furthermore, DAC treatment increased injury site mitochondrial protein expression in vehicle-treated mice to levels comparable to that of formoterol-treated mice. The effect of DNMT inhibition on pain response with and without formoterol was assessed following moderate SCI. While all injured mice not treated with DAC displayed thermal hyperalgesia by 21 DPI, mice treated with formoterol exhibited decreased thermal hyperalgesia compared to vehicle-treated mice by 35 DPI. Injured mice treated with DAC, regardless of formoterol treatment, did not demonstrate thermal hyperalgesia at any time point assessed. Although these data do not suggest enhanced formoterol-induced recovery with DNMT inhibition, our findings indicate the importance of DNA methylation post-SCI and support both DNMT inhibition and formoterol as potential therapeutic avenues.
Collapse
Affiliation(s)
- Natalie E Scholpa
- Southern Arizona VA Health Care System, Tucson, AZ, United States of America; Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, United States of America.
| |
Collapse
|
5
|
Hatch MN, Etingen B, Raad J, Siddiqui S, Stroupe KT, Smith BM. Dual utilization of Medicare and VA outpatient care among Veterans with spinal cord injuries and disorders. J Spinal Cord Med 2023; 46:716-724. [PMID: 35108176 PMCID: PMC10446768 DOI: 10.1080/10790268.2022.2027321] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
OBJECTIVE Veterans with spinal cord injuries and disorders (SCI/D) utilizing Veterans Affairs healthcare facilities are also Medicare eligible. Use of multiple health care systems potentially duplicates or fragments care in this population; yet little is known about those using multiple systems. This study describes dual use of services paid for by VA and Medicare among Veterans with SCI/D. DESIGN Retrospective, cross-sectional, observational study. PARTICIPANTS Veterans with SCI/D (n = 13,902) who received healthcare services within the VA SCI System of Care and were eligible for or enrolled in Medicare in 2011. INTERVENTIONS N/A. OUTCOME MEASURES Patient characteristics, average number of visits and patient level frequencies of reasons for visits were determined for individuals within healthcare utilization (VA only, Medicare only, or dual VA/Medicare) groups. Multinomial logistic regression analyses were used to investigate associations of patient variables on dual use. RESULTS 65.3% of Veterans with SCI/D were VA only users for outpatient encounters, 4.4% had encounters paid for by Medicare only, and 30.3% were dual users. Veterans were less likely to be VA only users if they were older than 69 and if they had been injured for greater than ten years. African American Veterans with SCI (compared to white) were more likely to be VA only users. CONCLUSION A substantial number (∼30%) of Veterans with SCI/D are dual users. These numbers highlight the importance of improved strategies to coordinate care and increase health information sharing across systems.
Collapse
Affiliation(s)
- Maya N Hatch
- Spinal Cord Injury/Disorder Center, Long Beach Veterans Affairs (VA) Medical Center, Long Beach, California, USA
- Physical Medicine & Rehabilitation Department, UC Irvine School of Medicine, Irvine, California, USA
| | - Bella Etingen
- Center of Innovation for Complex Chronic Healthcare, Edward Hines Jr. Department of VA Hospital, Chicago, Illinois, USA
| | - Jason Raad
- Econometrica, Inc, Bethesda, Massachusetts, USA
| | - Sameer Siddiqui
- Spinal Cord Injury/Disorder Center, Louis Stokes Cleveland (VA) Medical Center, Cleveland, Ohio, USA
- Department of Physical Medicine & Rehabilitation, Case Western Reserve University, Cleveland, Ohio, USA
| | - Kevin T Stroupe
- Center of Innovation for Complex Chronic Healthcare, Edward Hines Jr. Department of VA Hospital, Chicago, Illinois, USA
| | - Bridget M Smith
- Center of Innovation for Complex Chronic Healthcare, Edward Hines Jr. Department of VA Hospital, Chicago, Illinois, USA
- Institute for Healthcare Studies, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
6
|
McCoy HM, Polcyn R, Banik NL, Haque A. Regulation of enolase activation to promote neural protection and regeneration in spinal cord injury. Neural Regen Res 2023; 18:1457-1462. [PMID: 36571342 PMCID: PMC10075133 DOI: 10.4103/1673-5374.361539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 10/10/2022] [Accepted: 10/26/2022] [Indexed: 11/19/2022] Open
Abstract
Spinal cord injury (SCI) is a debilitating condition characterized by damage to the spinal cord resulting in loss of function, mobility, and sensation with no U.S. Food and Drug Administration-approved cure. Enolase, a multifunctional glycolytic enzyme upregulated after SCI, promotes pro- and anti-inflammatory events and regulates functional recovery in SCI. Enolase is normally expressed in the cytosol, but the expression is upregulated at the cell surface following cellular injury, promoting glial cell activation and signal transduction pathway activation. SCI-induced microglia activation triggers pro-inflammatory mediators at the injury site, activating other immune cells and metabolic events, i.e., Rho-associated kinase, contributing to the neuroinflammation found in SCI. Enolase surface expression also activates cathepsin X, resulting in cleavage of the C-terminal end of neuron-specific enolase (NSE) and non-neuronal enolase (NNE). Fully functional enolase is necessary as NSE/NNE C-terminal proteins activate many neurotrophic processes, i.e., the plasminogen activation system, phosphatidylinositol-4,5-bisphosphate 3-kinase/protein kinase B, and mitogen-activated protein kinase/extracellular signal-regulated kinase. Studies here suggest an enolase inhibitor, ENOblock, attenuates the activation of Rho-associated kinase, which may decrease glial cell activation and promote functional recovery following SCI. Also, ENOblock inhibits cathepsin X, which may help prevent the cleavage of the neurotrophic C-terminal protein allowing full plasminogen activation and phosphatidylinositol-4,5-bisphosphate 3-kinase/mitogen-activated protein kinase activity. The combined NSE/cathepsin X inhibition may serve as a potential therapeutic strategy for preventing neuroinflammation/degeneration and promoting neural cell regeneration and recovery following SCI. The role of cell membrane-expressed enolase and associated metabolic events should be investigated to determine if the same strategies can be applied to other neurodegenerative diseases. Hence, this review discusses the importance of enolase activation and inhibition as a potential therapeutic target following SCI to promote neuronal survival and regeneration.
Collapse
Affiliation(s)
- Hannah M. McCoy
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
| | - Rachel Polcyn
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
| | - Naren L. Banik
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
- Department of Neurosurgery, Medical University of South Carolina, Charleston, SC, USA
- Ralph H. Johnson Veterans Administration Medical Center, Charleston, SC, USA
| | - Azizul Haque
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
- Department of Neurosurgery, Medical University of South Carolina, Charleston, SC, USA
- Ralph H. Johnson Veterans Administration Medical Center, Charleston, SC, USA
| |
Collapse
|
7
|
Lv Z, Dong C, Zhang T, Zhang S. Hydrogels in Spinal Cord Injury Repair: A Review. Front Bioeng Biotechnol 2022; 10:931800. [PMID: 35800332 PMCID: PMC9253563 DOI: 10.3389/fbioe.2022.931800] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 05/26/2022] [Indexed: 12/18/2022] Open
Abstract
Traffic accidents and falling objects are responsible for most spinal cord injuries (SCIs). SCI is characterized by high disability and tends to occur among the young, seriously affecting patients' lives and quality of life. The key aims of repairing SCI include preventing secondary nerve injury, inhibiting glial scarring and inflammatory response, and promoting nerve regeneration. Hydrogels have good biocompatibility and degradability, low immunogenicity, and easy-to-adjust mechanical properties. While providing structural scaffolds for tissues, hydrogels can also be used as slow-release carriers in neural tissue engineering to promote cell proliferation, migration, and differentiation, as well as accelerate the repair of damaged tissue. This review discusses the characteristics of hydrogels and their advantages as delivery vehicles, as well as expounds on the progress made in hydrogel therapy (alone or combined with cells and molecules) to repair SCI. In addition, we discuss the prospects of hydrogels in clinical research and provide new ideas for the treatment of SCI.
Collapse
Affiliation(s)
- Zhenshan Lv
- The Department of Spinal Surgery, 1st Hospital, Jilin University, Jilin Engineering Research Center for Spine and Spine Cord Injury, Changchun, China
| | - Chao Dong
- Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Tianjiao Zhang
- Medical Insurance Management Department, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Shaokun Zhang
- The Department of Spinal Surgery, 1st Hospital, Jilin University, Jilin Engineering Research Center for Spine and Spine Cord Injury, Changchun, China
| |
Collapse
|
8
|
Islam A, Tom VJ. The use of viral vectors to promote repair after spinal cord injury. Exp Neurol 2022; 354:114102. [PMID: 35513025 DOI: 10.1016/j.expneurol.2022.114102] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 04/21/2022] [Accepted: 04/27/2022] [Indexed: 11/16/2022]
Abstract
Spinal cord injury (SCI) is a devastating event that can permanently disrupt multiple modalities. Unfortunately, the combination of the inhibitory environment at a central nervous system (CNS) injury site and the diminished intrinsic capacity of adult axons for growth results in the failure for robust axonal regeneration, limiting the ability for repair. Delivering genetic material that can either positively or negatively modulate gene expression has the potential to counter the obstacles that hinder axon growth within the spinal cord after injury. A popular gene therapy method is to deliver the genetic material using viral vectors. There are considerations when deciding on a viral vector approach for a particular application, including the type of vector, as well as serotypes, and promoters. In this review, we will discuss some of the aspects to consider when utilizing a viral vector approach to as a therapy for SCI. Additionally, we will discuss some recent applications of gene therapy to target extrinsic and/or intrinsic barriers to promote axon regeneration after SCI in preclinical models. While still in early stages, this approach has potential to treat those living with SCI.
Collapse
Affiliation(s)
- Ashraful Islam
- Drexel University College of Medicine, Department of Neurobiology and Anatomy, Marion Murray Spinal Cord Research Center, Philadelphia, PA, USA
| | - Veronica J Tom
- Drexel University College of Medicine, Department of Neurobiology and Anatomy, Marion Murray Spinal Cord Research Center, Philadelphia, PA, USA.
| |
Collapse
|
9
|
Zabarsky ZK, Luo TD, Ma X, Dean GM, Smith TL. Pharmacologic Recruitment of Endogenous Neural Stem/Progenitor Cells for the Treatment of Spinal Cord Injury. Spine (Phila Pa 1976) 2022; 47:505-513. [PMID: 34669674 DOI: 10.1097/brs.0000000000004264] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
STUDY DESIGN Laboratory study using a rat T9 contusion model of spinal cord injury. OBJECTIVE This study aims to examine whether a combinatory treatment of Pioglitazone (PGZ) and granulocyte colony-stimulating factor (GCSF) can support neural stem/progenitor cells (NSPCs) directly and provide a sustainable microenvironment through immunomodulatory mechanisms. SUMMARY OF BACKGROUND DATA Neuroinflammation plays a crucial role in the progression of spinal cord injury (SCI) and hinders NSPC-mediated repair and regeneration. Broad acting drugs that mitigate inflammation and support NSPC proliferation have not been tested together in SCI research models. METHODS Isolated NSPCs were treated with vehicle control, PGZ, GCSF, or both PGZ and GSCF for 24 hours and stained with proliferation marker Ki67. Adult female Sprague-Dawley rats sustained moderate-to-severe contusion-based SCI at T9 and were administered either vehicle control, PGZ, GCSF, or both PGZ and GCSF treatments. RESULTS Immunocytochemistry revealed that cultured NSPCs treated with both drugs produced higher numbers of actively proliferating cells and total cell numbers. ELISA on spinal cord tissue lysates at 1, 3, and 7 days post-injury (DPI) demonstrated that animals treated with PGZ, GCSF, or combination therapy showed significantly higher doublecortin levels at 7 DPI compared to control animals (P < 0.05). Immunohistochemistry of injured tissue at 3, 7, and 14 DPI revealed no difference of ependymal NSPC proliferation between groups, but showed a significant decrease in lesion size with combination therapy compared to controls. Functional recovery was assessed by the Basso, Beattie, Bresnahan locomotor rating scale. Animals treated with both drugs had significantly higher levels of function at 1 (P < 0.001), 3 (P < 0.001), 7 (P < 0.05), and 14 (P < 0.05) DPI compared to controls. CONCLUSION These results indicate that PGZ and GCSF treatment synergistically enhance NSPCs numbers and improve functional recovery after SCI. Our findings support an immunomodulatory strategy to recruit native NSPCs as a potential acute care intervention for SCI.Level of Evidence: N/A.
Collapse
Affiliation(s)
- Zachary K Zabarsky
- Wake Forest School of Medicine, Department of Orthopaedic Surgery, Winston-Salem, NC
| | | | | | | | | |
Collapse
|
10
|
Khan H, Sharma K, Kumar A, Kaur A, Singh TG. Therapeutic implications of cyclooxygenase (COX) inhibitors in ischemic injury. Inflamm Res 2022; 71:277-292. [PMID: 35175358 DOI: 10.1007/s00011-022-01546-6] [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: 10/28/2021] [Revised: 01/19/2022] [Accepted: 01/22/2022] [Indexed: 12/15/2022] Open
Abstract
INTRODUCTION Ischemia-reperfusion injury (IRI) is the inexplicable aggravation of cellular dysfunction that results in blood flow restoration to previously ischemic tissues. COX mediates the oxidative conversion of AA to various prostaglandins and thromboxanes, which are involved in various physiological and pathological processes. In the pathophysiology of I/R injuries, COX has been found to play an important role. I/R injuries affect most vital organs and are characterized by inflammation, oxidative stress, cell death, and apoptosis, leading to morbidity and mortality. MATERIALS AND METHODS A systematic literature review of Bentham, Scopus, PubMed, Medline, and EMBASE (Elsevier) databases was carried out to understand the Nature and mechanistic interventions of the Cyclooxygenase modulations in ischemic injury. Here, we have discussed the COX Physiology and downstream signalling pathways modulated by COX, e.g., Camp Pathway, Peroxisome Proliferator-Activated Receptor Activity, NF-kB Signalling, PI3K/Akt Signalling in ischemic injury. CONCLUSION This review will discuss the various COX types, specifically COX-1 and COX-2, which are involved in developing I/R injury in organs such as the brain, spinal cord, heart, kidney, liver, and intestine.
Collapse
Affiliation(s)
- Heena Khan
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
| | - Kunal Sharma
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
| | - Amit Kumar
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
| | - Amarjot Kaur
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
| | - Thakur Gurjeet Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India.
| |
Collapse
|
11
|
New Therapy for Spinal Cord Injury: Autologous Genetically-Enriched Leucoconcentrate Integrated with Epidural Electrical Stimulation. Cells 2022; 11:cells11010144. [PMID: 35011706 PMCID: PMC8750549 DOI: 10.3390/cells11010144] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 12/07/2021] [Accepted: 12/29/2021] [Indexed: 12/17/2022] Open
Abstract
The contemporary strategy for spinal cord injury (SCI) therapy aims to combine multiple approaches to control pathogenic mechanisms of neurodegeneration and stimulate neuroregeneration. In this study, a novel regenerative approach using an autologous leucoconcentrate enriched with transgenes encoding vascular endothelial growth factor (VEGF), glial cell line-derived neurotrophic factor (GDNF), and neural cell adhesion molecule (NCAM) combined with supra- and sub-lesional epidural electrical stimulation (EES) was tested on mini-pigs similar in morpho-physiological scale to humans. The complex analysis of the spinal cord recovery after a moderate contusion injury in treated mini-pigs compared to control animals revealed: better performance in behavioural and joint kinematics, restoration of electromyography characteristics, and improvement in selected immunohistology features related to cell survivability, synaptic protein expression, and glial reorganization above and below the injury. These results for the first time demonstrate the positive effect of intravenous infusion of autologous genetically-enriched leucoconcentrate producing recombinant molecules stimulating neuroregeneration combined with neuromodulation by translesional multisite EES on the restoration of the post-traumatic spinal cord in mini-pigs and suggest the high translational potential of this novel regenerative therapy for SCI patients.
Collapse
|
12
|
Walsh CM, Wychowaniec JK, Brougham DF, Dooley D. Functional hydrogels as therapeutic tools for spinal cord injury: New perspectives on immunopharmacological interventions. Pharmacol Ther 2021; 234:108043. [PMID: 34813862 DOI: 10.1016/j.pharmthera.2021.108043] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 11/10/2021] [Accepted: 11/11/2021] [Indexed: 02/06/2023]
Abstract
Spinal cord injury (SCI) is a complex medical and psychological challenge for which there is no curative therapy currently available. Despite major progress in pharmacological and surgical approaches, clinical trials for SCI patients have been uniformly disappointing thus far as there are many practical and biological issues yet to be resolved. Neuroinflammation is a critical event of the secondary injury phase after SCI, and recent research strategies have focused on modulating the immune response after injury to provide a more favorable recovery environment. Biomaterials can serve this purpose by providing physical and trophic support to the injured spinal cord after SCI. Of all potential biomaterials, functional hydrogels are emerging as a key component in novel treatment strategies for SCI, including controlled and localized delivery of immunomodulatory therapies to drive polarization of immune cells towards a pro-regenerative phenotype. Here, we extensively review recent developments in the use of functional hydrogels as immunomodulatory therapies for SCI. We briefly describe physicochemical properties of hydrogels and demonstrate how advanced fabrication methods lead to the required heterogeneity and hierarchical arrangements that increasingly mimic complex spinal cord tissue. We then summarize potential SCI therapeutic modalities including: (i) hydrogels alone; (ii) hydrogels as cellular or (iii) bioactive molecule delivery vehicles, and; (iv) combinatorial approaches. By linking the structural properties of hydrogels to their functions in treatment with particular focus on immunopharmacological stimuli, this may accelerate further development of functional hydrogels for SCI, and indeed next-generation central nervous system regenerative therapies.
Collapse
Affiliation(s)
- Ciara M Walsh
- School of Medicine, Health Sciences Centre, University College Dublin, Belfield, Dublin 4, Ireland; UCD Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| | - Jacek K Wychowaniec
- School of Chemistry, University College Dublin, Belfield, Dublin 4, Ireland; AO Research Institute Davos, Clavadelerstrasse 8, 7270 Davos, Switzerland
| | - Dermot F Brougham
- School of Chemistry, University College Dublin, Belfield, Dublin 4, Ireland
| | - Dearbhaile Dooley
- School of Medicine, Health Sciences Centre, University College Dublin, Belfield, Dublin 4, Ireland; UCD Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland.
| |
Collapse
|
13
|
Shams R, Banik NL, Haque A. Implications of enolase in the RANKL-mediated osteoclast activity following spinal cord injury. ACTA ACUST UNITED AC 2021; 45:1453-1457. [PMID: 34539043 PMCID: PMC8445338 DOI: 10.32604/biocell.2021.017659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Spinal Cord Injury (SCI) is a debilitating condition characterized by damage to the spinal cord, resulting in loss of function, mobility, and sensation. Although increasingly prevalent in the US, no FDA-approved therapy exists due to the unfortunate complexity of the condition, and the difficulties of SCI may be furthered by the development of SCI-related complications, such as osteoporosis. SCI demonstrates two crucial stages for consideration: the primary stage and the secondary stage. While the primary stage is suggested to be immediate and irreversible, the secondary stage is proposed as a promising window of opportunity for therapeutic intervention. Enolase, a metabolic enzyme upregulated after SCI, performs non-glycolytic functions, promoting inflammatory events via extracellular degradative actions and increased production of inflammatory cytokines and chemokines. Neuron-specific enolase (NSE) serves as a biomarker of functional damage to neurons following SCI, and the inhibition of NSE has been demonstrated to reduce signs of secondary injury of SCI and to ameliorate dysfunction. This Viewpoint article involves enolase activation in the regulation of RANK-RANKL pathway and summarizes succinctly the mechanisms influencing osteoclast-mediated resorption of bone in SCI. Our laboratory proposes that inhibition of enolase activation may reduce SCI-induced inflammatory response and decrease osteoclast activity, limiting the chances of skeletal tissue loss in SCI.
Collapse
Affiliation(s)
- Ramsha Shams
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC 29425, USA.,Department of Neurosurgery, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Naren L Banik
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC 29425, USA.,Department of Neurosurgery, Medical University of South Carolina, Charleston, SC 29425, USA.,Ralph H. Johnson Veterans Administration Medical Center, Charleston, SC 29401, USA
| | - Azizul Haque
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
14
|
Feng Z, Min L, Liang L, Chen B, Chen H, Zhou Y, Deng W, Liu H, Hou J. Neutrophil Extracellular Traps Exacerbate Secondary Injury via Promoting Neuroinflammation and Blood-Spinal Cord Barrier Disruption in Spinal Cord Injury. Front Immunol 2021; 12:698249. [PMID: 34456910 PMCID: PMC8385494 DOI: 10.3389/fimmu.2021.698249] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 07/19/2021] [Indexed: 12/12/2022] Open
Abstract
As the first inflammatory cell recruited to the site of spinal cord injury (SCI), neutrophils were reported to be detrimental to SCI. However, the precise mechanisms as to how neutrophils exacerbate SCI remain largely obscure. In the present study, we demonstrated that infiltrated neutrophils produce neutrophil extracellular traps (NETs), which subsequently promote neuroinflammation and blood–spinal cord barrier disruption to aggravate spinal cord edema and neuronal apoptosis following SCI in rats. Both inhibition of NETs formation by peptidylarginine deiminase 4 (PAD4) inhibitor and disruption of NETs by DNase 1 alleviate secondary damage, thus restraining scar formation and promoting functional recovery after SCI. Furthermore, we found that NETs exacerbate SCI partly via elevating transient receptor potential vanilloid type 4 (TRPV4) level in the injured spinal cord. Therefore, our results indicate that NETs might be a promising therapeutic target for SCI.
Collapse
Affiliation(s)
- Zhou Feng
- Department of Rehabilitation, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Lingxia Min
- Department of Rehabilitation, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Liang Liang
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Beike Chen
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Hui Chen
- Department of Rehabilitation, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yi Zhou
- Department of Rehabilitation, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Weiwei Deng
- Department of Rehabilitation, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Hongliang Liu
- Department of Rehabilitation, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Jingming Hou
- Department of Rehabilitation, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| |
Collapse
|
15
|
Shams R, Drasites KP, Zaman V, Matzelle D, Shields DC, Garner DP, Sole CJ, Haque A, Banik NL. The Pathophysiology of Osteoporosis after Spinal Cord Injury. Int J Mol Sci 2021; 22:3057. [PMID: 33802713 PMCID: PMC8002377 DOI: 10.3390/ijms22063057] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 03/15/2021] [Accepted: 03/15/2021] [Indexed: 12/12/2022] Open
Abstract
Spinal cord injury (SCI) affects approximately 300,000 people in the United States. Most individuals who sustain severe SCI also develop subsequent osteoporosis. However, beyond immobilization-related lack of long bone loading, multiple mechanisms of SCI-related bone density loss are incompletely understood. Recent findings suggest neuronal impairment and disability may lead to an upregulation of receptor activator of nuclear factor-κB ligand (RANKL), which promotes bone resorption. Disruption of Wnt signaling and dysregulation of RANKL may also contribute to the pathogenesis of SCI-related osteoporosis. Estrogenic effects may protect bones from resorption by decreasing the upregulation of RANKL. This review will discuss the current proposed physiological and cellular mechanisms explaining osteoporosis associated with SCI. In addition, we will discuss emerging pharmacological and physiological treatment strategies, including the promising effects of estrogen on cellular protection.
Collapse
Affiliation(s)
- Ramsha Shams
- Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas St., Charleston, SC 29425, USA; (R.S.); (K.P.D.); (V.Z.); (D.M.); (D.C.S.)
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA
- Department of Health and Human Performance, The Citadel, 171 Moultrie St., Charleston, SC 29409, USA; (D.P.G.); (C.J.S.)
| | - Kelsey P. Drasites
- Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas St., Charleston, SC 29425, USA; (R.S.); (K.P.D.); (V.Z.); (D.M.); (D.C.S.)
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA
- Department of Health and Human Performance, The Citadel, 171 Moultrie St., Charleston, SC 29409, USA; (D.P.G.); (C.J.S.)
| | - Vandana Zaman
- Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas St., Charleston, SC 29425, USA; (R.S.); (K.P.D.); (V.Z.); (D.M.); (D.C.S.)
- Ralph H. Johnson Veterans Administration Medical Center, 109 Bee St., Charleston, SC 29401, USA
| | - Denise Matzelle
- Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas St., Charleston, SC 29425, USA; (R.S.); (K.P.D.); (V.Z.); (D.M.); (D.C.S.)
- Ralph H. Johnson Veterans Administration Medical Center, 109 Bee St., Charleston, SC 29401, USA
| | - Donald C. Shields
- Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas St., Charleston, SC 29425, USA; (R.S.); (K.P.D.); (V.Z.); (D.M.); (D.C.S.)
| | - Dena P. Garner
- Department of Health and Human Performance, The Citadel, 171 Moultrie St., Charleston, SC 29409, USA; (D.P.G.); (C.J.S.)
| | - Christopher J. Sole
- Department of Health and Human Performance, The Citadel, 171 Moultrie St., Charleston, SC 29409, USA; (D.P.G.); (C.J.S.)
| | - Azizul Haque
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA
| | - Narendra L. Banik
- Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas St., Charleston, SC 29425, USA; (R.S.); (K.P.D.); (V.Z.); (D.M.); (D.C.S.)
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA
- Ralph H. Johnson Veterans Administration Medical Center, 109 Bee St., Charleston, SC 29401, USA
| |
Collapse
|
16
|
Cox A, Capone M, Matzelle D, Vertegel A, Bredikhin M, Varma A, Haque A, Shields DC, Banik NL. Nanoparticle-Based Estrogen Delivery to Spinal Cord Injury Site Reduces Local Parenchymal Destruction and Improves Functional Recovery. J Neurotrauma 2020; 38:342-352. [PMID: 32680442 DOI: 10.1089/neu.2020.7047] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Spinal cord injury (SCI) patients sustain significant functional impairments; this is causally related to restricted neuronal regeneration after injury. The ensuing reactive gliosis, inflammatory cascade, and glial scar formation impede axonal regrowth. Although systemic anti-inflammatory agents (steroids) have been previously administered to counteract this, no current therapeutic is approved for post-injury neuronal regeneration, in part because of related side effects. Likewise, therapeutic systemic estrogen levels exhibit neuroprotective properties, but dose-dependent side effects are prohibitive. The current study thus uses low-dose estrogen delivery to the spinal cord injury (SCI) site using an agarose gel patch embedded with estrogen-loaded nanoparticles. Compared to controls, spinal cords from rodents treated with nanoparticle site-directed estrogen demonstrated significantly decreased post-injury lesion size, reactive gliosis, and glial scar formation. However, axonal regeneration, vascular endothelial growth factor production, and glial-cell-derived neurotrophic factor levels were increased with estrogen administration. Concomitantly improved locomotor and bladder functional recovery were observed with estrogen administration after injury. Therefore, low-dose site-directed estrogen may provide a future approach for enhanced neuronal repair and functional recovery in SCI patients.
Collapse
Affiliation(s)
- April Cox
- Department of Neurosurgery, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Mollie Capone
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Denise Matzelle
- Department of Neurosurgery, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Alexey Vertegel
- Department of Bioengineering, Clemson University, Clemson, South Carolina, USA
| | - Mikhail Bredikhin
- Department of Bioengineering, Clemson University, Clemson, South Carolina, USA
| | - Abhay Varma
- Department of Neurosurgery, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Azizul Haque
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Donald C Shields
- Department of Neurosurgery, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Naren L Banik
- Department of Neurosurgery, Medical University of South Carolina, Charleston, South Carolina, USA.,Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina, USA.,Ralph H. Johnson Veterans Administration Medical Center, Charleston, South Carolina, USA
| |
Collapse
|
17
|
Polcyn R, Capone M, Matzelle D, Hossain A, Chandran R, Banik NL, Haque A. Enolase inhibition alters metabolic hormones and inflammatory factors to promote neuroprotection in spinal cord injury. Neurochem Int 2020; 139:104788. [PMID: 32650031 DOI: 10.1016/j.neuint.2020.104788] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 06/10/2020] [Accepted: 06/20/2020] [Indexed: 12/17/2022]
Abstract
Enolase inhibition is a potential therapeutic strategy currently being investigated for treatment of spinal cord injury (SCI) as it reduces pro-inflammatory cytokines and chemokines, alters metabolic factors, and reduces gliosis in acute SCI. Herein, the role of enolase in SCI has been examined to better understand the effects of this enzyme on inflammation, metabolic hormones, glial cell activation, and neuroprotection under these shorter injury conditions. Immunohistochemical analyses of inflammatory markers vimentin, Cox-2, and caspase-1 indicated that enolase inhibition attenuated the elevated levels of inflammation seen following SCI. Iba1, GFAP, NFP, and CSPG staining indicated that enolase inhibition with prolonged administration of ENOblock reduced microglia/astrocyte activation and lead to enhanced neuroprotection in SCI. An analysis of metabolic hormones revealed that ENOblock treatment significantly upregulated plasma concentrations of peptide YY, glucagon-like peptide 1, glucose-dependent insulinotropic peptide, glucagon, and insulin hormones as compared to vehicle-treated controls (Mann-Whitney, p ≤ 0.05). ENOblock did not have a significant effect on plasma concentrations of pancreatic polypeptide. Interestingly, ENOblock treatment inhibited chondroitin sulfate proteoglycan (CSPG), which is produced by activated glia and serves to block regrowth of axons across the lesion site following injury. An increased level of NeuN and MBP with reduced caspase-1 was detected in SCI tissues after ENOblock treatment, suggesting preservation of myelin and induction of neuroprotection. ENOblock also induced improved motor function in SCI rats, indicating a role for enolase in modulating inflammatory and metabolic factors in SCI with important implications for clinical consideration.
Collapse
Affiliation(s)
- Rachel Polcyn
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA
| | - Mollie Capone
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA; Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas Street Charleston, SC 29425, USA
| | - Denise Matzelle
- Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas Street Charleston, SC 29425, USA; Ralph H. Johnson Veterans Administration Medical Center, 109 Bee St, Charleston, SC 29401, USA
| | - Azim Hossain
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA
| | - Raghavendar Chandran
- Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas Street Charleston, SC 29425, USA
| | - Naren L Banik
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA; Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas Street Charleston, SC 29425, USA; Ralph H. Johnson Veterans Administration Medical Center, 109 Bee St, Charleston, SC 29401, USA
| | - Azizul Haque
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA.
| |
Collapse
|
18
|
Shah J, Smith RH, Al-Gharaibeh A. A Novel Electromagnetic-Neurobiologic Interface for Functional Animation of Dormant Motor Nerve Roots in Spinal Cord Injury via Neuromodulation. Front Surg 2020; 6:73. [PMID: 31998742 PMCID: PMC6970190 DOI: 10.3389/fsurg.2019.00073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 12/06/2019] [Indexed: 11/19/2022] Open
Abstract
Complete spinal cord injury is a devastating occurrence afflicting millions of people worldwide with no available treatment for functional motor recovery. In this report, we describe a procedure in which we used parts of a device available for chronic pain treatment to provide a neuromodulation of motor nerve roots in a case with complete motor and sensory paraplegia. By using a retrograde trans-foraminal approach to implant electrodes close to L2-S1 motor nerve roots bilaterally, we were able to stimulate those nerves and induce precise movements at the joints of lower extremity in a T5 complete spinal cord injury case. We believe that our approach shows potential of the device as a rehabilitation system with the possibility of a parallel electric circuitry that can bridge a damaged spinal cord.
Collapse
Affiliation(s)
- Jawad Shah
- Department of Neurosurgery, Insight Institute of Neurosurgery & Neuroscience, Flint, MI, United States.,Department of Research, Insight Institute of Neurosurgery & Neuroscience, Flint, MI, United States.,Center for Cognition and Neuroethics, University of Michigan-Flint - IINN, Flint, MI, United States.,Department of Medicine, College of Human Medicine, Michigan State University, East Lansing, MI, United States
| | - Richard H Smith
- Department of Research, Insight Institute of Neurosurgery & Neuroscience, Flint, MI, United States
| | - Abeer Al-Gharaibeh
- Department of Research, Insight Institute of Neurosurgery & Neuroscience, Flint, MI, United States.,Center for Cognition and Neuroethics, University of Michigan-Flint - IINN, Flint, MI, United States
| |
Collapse
|
19
|
Zhou Y, Su P, Pan Z, Liu D, Niu Y, Zhu W, Yao P, Song Y, Sun Y. Combination Therapy With Hyperbaric Oxygen and Erythropoietin Inhibits Neuronal Apoptosis and Improves Recovery in Rats With Spinal Cord Injury. Phys Ther 2019; 99:1679-1689. [PMID: 31504911 DOI: 10.1093/ptj/pzz125] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 01/03/2019] [Accepted: 04/20/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND Apoptosis plays an important role in various diseases, including spinal cord injury (SCI). Hyperbaric oxygen (HBO) and erythropoietin (EPO) promote the recovery from SCI, but the relationship between apoptosis and the combination therapeutic effect is not completely clear. OBJECTIVE The purpose of this study was to investigate the effects of HBO and EPO on SCI and the mechanisms that underlie their therapeutic benefits. DESIGN The study was designed to explore the effects of HBO and EPO on SCI through a randomized controlled trial. METHODS Sixty young developing female Sprague-Dawley rats were randomly divided into groups of 12 rats receiving sham, SCI, HBO, EPO, or HBO plus EPO. The SCI model was modified with the Allen method to better control consistency. HBO was performed for 1 hour per day for a total of 21 days, and EPO was given once per week for a total of 3 weeks. Both methods were performed 2 hours after SCI. Locomotor function was evaluated with the 21-point Basso-Beattie-Bresnahan Locomotor Rating Scale, an inclined-plane test, and a footprint analysis. All genes were detected by Western blotting and immunohistochemistry. The level of cell apoptosis was determined by Hoechst staining. RESULTS The results showed that HBO and EPO promoted the recovery of locomotor function in the hind limbs of rats by inhibiting the apoptosis of neurons. During this period, the expression of B-cell lymphoma/leukemia 2 protein (Bcl-2) increased significantly, whereas the expression of Bcl-2-associated X protein (Bax) and cleaved caspase 3 decreased significantly, indicating the inhibition of apoptosis. Meanwhile, the expression of G protein-coupled receptor 17 decreased, and that of myelin basic protein increased, suggesting that there may be a potential connection between demyelination and neuronal apoptosis. LIMITATIONS The limitations of the study include deviations in the preparation of SCI models; lack of reverse validation of molecular mechanisms; absence of in vitro cell experiments; and only one time point after SCI was studied. CONCLUSIONS HBO and EPO treatments are beneficial for SCI, especially when the 2 therapies are combined.
Collapse
Affiliation(s)
- Yue Zhou
- Department of Orthopedics, Shaoxing People's Hospital, Shaoxing, China; Department of Orthopedics and The Experimental Center, The Second Affiliated Hospital of Soochow University, Suzhou, China.,Department of Orthopedics, The Second Affiliated Hospital of Soochow University
| | - Peng Su
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University
| | - Zhenzhen Pan
- Department of Radiology, People's Hospital of Changshan, Quzhou, China
| | - Dong Liu
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University
| | - Yanping Niu
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University
| | - Weiqing Zhu
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University
| | - Pengfei Yao
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University
| | - Yue Song
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University
| | - Yongming Sun
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, 1055 Sanxiang Road, Suzhou 215004, China
| |
Collapse
|
20
|
Zhao L, Zhai M, Yang X, Guo H, Cao Y, Wang D, Li P, Liu C. Dexmedetomidine attenuates neuronal injury after spinal cord ischaemia-reperfusion injury by targeting the CNPY2-endoplasmic reticulum stress signalling. J Cell Mol Med 2019; 23:8173-8183. [PMID: 31625681 PMCID: PMC6850922 DOI: 10.1111/jcmm.14688] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 06/13/2019] [Accepted: 08/14/2019] [Indexed: 02/06/2023] Open
Abstract
Dexmedetomidine (Dex) has been proven to exert protective effects on multiple organs in response to ischaemia-reperfusion injury, but the specific mechanism by which this occurs has not been fully elucidated. The purpose of this study was to investigate whether Dex attenuates spinal cord ischaemia-reperfusion injury (SCIRI) by inhibiting endoplasmic reticulum stress (ERS). Our team established a model of SCIRI and utilized the endoplasmic reticulum agonist thapsigargin. Dex (25 g/kg) was intraperitoneally injected 30 minutes before spinal cord ischaemia. After 45 minutes of ischaemia, the spinal cord was reperfused for 24 hours. To evaluate the neuroprotective effect of Dex on SCIRI, neurological function scores were assessed in rats and apoptosis of spinal cord cells was determined by TUNEL staining. To determine whether the endoplasmic reticulum apoptosis pathway CNPY2-PERK was involved in the neuroprotective mechanism of Dex, the expression levels of related proteins (CNPY2, GRP78, PERK, CHOP, caspase-12, caspase-9 and caspase-3) were detected by western blot analysis and RT-PCR. We observed that Dex significantly increased the neurological function scores after SCIRI and decreased apoptosis of spinal cord cells. The expression of ERS-related apoptosis proteins was significantly increased by SCIRI but was significantly decreased in response to Dex administration. Taken together, the results of this study indicate that Dex may attenuate SCIRI by inhibiting the CNPY2-ERS apoptotic pathway.
Collapse
Affiliation(s)
- Lina Zhao
- Department of Anaesthesiology, Tianjin Hospital, Tianjin, China
| | - Meili Zhai
- Department of Anesthesiology, Tianjin Central Hospital of Gynecology Obstetrics, Gynecology Obstetrics Hospital of Nankai University, Tianjin Key Laboratory of human development and reproductive regulation, Tianjin, China
| | - Xu Yang
- Department of medicine, Tianjin Medical College, Tianjin, China
| | - Hongjie Guo
- Department of Critical Care Medicine, Tianjin 4th Centre Hospital, Tianjin, China
| | - Ying Cao
- Department of Critical Care Medicine, Tianjin 4th Centre Hospital, Tianjin, China
| | - Donghui Wang
- Department of Medical Imaging, Shanxi Medical University, Taiyuan City, China
| | - Ping Li
- Department of Anaesthesiology, Tianjin Hospital, Tianjin, China
| | - Chong Liu
- Department of Anaesthesiology, Central Laboratory, Tianjin 4th Centre Hospital, The Fourth Central Hospital Affiliated to Nankai University, The Fourth Center Clinical College of Tianjin Medical University, Tianjin, China
| |
Collapse
|
21
|
Ren Z, Wang X, Xu M, Frank JA, Luo J. Minocycline attenuates ethanol-induced cell death and microglial activation in the developing spinal cord. Alcohol 2019; 79:25-35. [PMID: 30529756 DOI: 10.1016/j.alcohol.2018.12.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 12/03/2018] [Accepted: 12/04/2018] [Indexed: 12/12/2022]
Abstract
Developmental exposure to ethanol may cause fetal alcohol spectrum disorders (FASD), and the immature central nervous system (CNS) is particularly vulnerable to ethanol. In addition to vulnerability in the developing brain, we previously showed that ethanol also caused neuroapoptosis, microglial activation, and neuroinflammation in the spinal cord. Minocycline is an antibiotic that inhibits microglial activation and alleviates neuroinflammation. We sought to determine whether minocycline could protect spinal cord neurons against ethanol-induced damage. In this study, we showed that minocycline significantly inhibited ethanol-induced caspase-3 activation, microglial activation, and the expression of pro-inflammatory cytokines in the developing spinal cord. Moreover, minocycline blocked ethanol-induced activation of glycogen synthase kinase 3 beta (GSK3β), a key regulator of microglial activation. Meanwhile, minocycline significantly restored ethanol-induced inhibition of protein kinase B (AKT), mammalian target of the rapamycin (mTOR), and ERK1/2 signaling pathways, which were important pro-survival signaling pathways for neurons. Together, minocycline may attenuate ethanol-induced damage to the developing spinal cord by inhibiting microglial activation/neuroinflammation and by restoring the pro-survival signaling.
Collapse
|
22
|
3D Bioprinting Human Induced Pluripotent Stem Cell-Derived Neural Tissues Using a Novel Lab-on-a-Printer Technology. APPLIED SCIENCES-BASEL 2018. [DOI: 10.3390/app8122414] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Most neurological diseases and disorders lack true cures, including spinal cord injury (SCI). Accordingly, current treatments only alleviate the symptoms of these neurological diseases and disorders. Engineered neural tissues derived from human induced pluripotent stem cells (hiPSCs) can serve as powerful tools to identify drug targets for treating such diseases and disorders. In this work, we demonstrate how hiPSC-derived neural progenitor cells (NPCs) can be bioprinted into defined structures using Aspect Biosystems’ novel RX1 bioprinter in combination with our unique fibrin-based bioink in rapid fashion as it takes under 5 min to print four tissues. This printing process preserves high levels of cell viability (>81%) and their differentiation capacity in comparison to less sophisticated bioprinting methods. These bioprinted neural tissues expressed the neuronal marker, βT-III (45 ± 20.9%), after 15 days of culture and markers associated with spinal cord (SC) motor neurons (MNs), such as Olig2 (68.8 ± 6.9%), and HB9 (99.6 ± 0.4%) as indicated by flow cytometry. The bioprinted neural tissues expressed the mature MN marker, ChaT, after 30 days of culture as indicated by immunocytochemistry. In conclusion, we have presented a novel method for high throughput production of mature hiPSC-derived neural tissues with defined structures that resemble those found in the SC.
Collapse
|
23
|
Doyle C, Cristofaro V, Sullivan MP, Adam RM. Inosine - a Multifunctional Treatment for Complications of Neurologic Injury. Cell Physiol Biochem 2018; 49:2293-2303. [PMID: 30261493 DOI: 10.1159/000493831] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 09/18/2018] [Indexed: 01/31/2023] Open
Abstract
Spinal cord injury (SCI) caused by trauma or disease leads to motor and sensory abnormalities that depend on the level, severity and duration of the lesion. The most obvious consequence of SCI is paralysis affecting lower and upper limbs. SCI also leads to loss of bladder and bowel control, both of which have a deleterious, life-long impact on the social, psychological, functional, medical and economic well being of affected individuals. Currently, there is neither a cure for SCI nor is there adequate management of its consequences. Although medications provide symptomatic relief for the complications of SCI including muscle spasms, lower urinary tract dysfunction and hyperreflexic bowel, strategies for repair of spinal injuries and recovery of normal limb and organ function are still to be realized. In this review, we discuss experimental evidence supporting the use of the naturally occurring purine nucleoside inosine to improve the devastating sequelae of SCI. Evidence suggests inosine is a safe, novel agent with multifunctional properties that is effective in treating complications of SCI and other neuropathies.
Collapse
Affiliation(s)
- Claire Doyle
- Urological Diseases Research Center, Boston Children's Hospital, Boston, Massachusetts, USA.,Department of Surgery, Harvard Medical School, Boston, Massachusetts, USA
| | - Vivian Cristofaro
- Department of Surgery, Harvard Medical School, Boston, Massachusetts, USA.,Division of Urology, VA Boston Healthcare System, Boston, Massachusetts, USA.,Department of Surgery, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Maryrose P Sullivan
- Department of Surgery, Harvard Medical School, Boston, Massachusetts, USA.,Division of Urology, VA Boston Healthcare System, Boston, Massachusetts, USA.,Department of Surgery, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Rosalyn M Adam
- Urological Diseases Research Center, Boston Children's Hospital, Boston, Massachusetts, USA.,Department of Surgery, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
24
|
Chen XG, Chen LH, Xu RX, Zhang HT. Effect evaluation of methylprednisolone plus mitochondrial division inhibitor-1 on spinal cord injury rats. Childs Nerv Syst 2018; 34:1479-1487. [PMID: 29682689 DOI: 10.1007/s00381-018-3792-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Accepted: 03/28/2018] [Indexed: 12/30/2022]
Abstract
PURPOSE To investigate the combination effect of methylprednisolone (MP) and mitochondrial division inhibitor-1 (Mdivi-1) on the neurological function recovery of rat spinal cord injury (SCI) model. METHODS The weight-drop method was used to establish the rat SCI model; then, rats were randomized into sham group, SCI group, MP group, Mdivi-1 group and MP+Mdivi-1 group. Motor function scores were quantified to evaluate locomotor ability; HE staining was used to assess spinal cord histopathology; tissue water content, oxidative stress, tissue mitochondrial function, neurons apoptosis, and apoptosis-related protein expression were detected. RESULTS From the third day after SCI, BBB score of the MP+Mdivi-1 group was obviously higher than the other experimental groups (p < 0.05). Compared with the SCI group, tissue water content of the Mdivi-1 group and MP+Mdivi-1 group reduced obviously (p < 0.05), mitochondrial membrane potential (MMP) level and ATP content in the Mdivi-1 group and MP+Mdivi-1 group were both higher (p < 0.05). Meanwhile, three kinds of treatment all reduced apoptosis significantly, while MP plus Mdivi-1 exhibited the best inhibition effect on apoptosis (p < 0.05). The expression levels of Drp1, cytochrome c, and caspase-3 were all upregulated obviously; Mdivi-1 could inhibit Drp1 upregulation induced by SCI; for the upregulation of cytochrome c and caspase-3, the inhibition effect of Mdivi-1 approached MP. When MP combined with Mdivi-1, there was the best inhibition effect. CONCLUSIONS MP combined with Mdivi-1 may produce better neurological function recovery, through improving functional status of mitochondria and inhibiting lipid peroxidation in damaged tissue of SCI rats, and thus alleviating apoptosis.
Collapse
Affiliation(s)
- Xu-Gui Chen
- The Affiliated Bayi Brain Hospital, the Army General Hospital PLA, No. 5, Nanmencang, Dongcheng District, Beijing, 100700, China
| | - Li-Hua Chen
- The Affiliated Bayi Brain Hospital, the Army General Hospital PLA, No. 5, Nanmencang, Dongcheng District, Beijing, 100700, China
| | - Ru-Xiang Xu
- The Affiliated Bayi Brain Hospital, the Army General Hospital PLA, No. 5, Nanmencang, Dongcheng District, Beijing, 100700, China
| | - Hong-Tian Zhang
- The Affiliated Bayi Brain Hospital, the Army General Hospital PLA, No. 5, Nanmencang, Dongcheng District, Beijing, 100700, China.
| |
Collapse
|
25
|
Study on the effect of nano-hydroxyapatite on bone repair of athletes. Wideochir Inne Tech Maloinwazyjne 2018; 13:221-226. [PMID: 30002755 PMCID: PMC6041591 DOI: 10.5114/wiitm.2018.73973] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 01/21/2018] [Indexed: 12/05/2022] Open
Abstract
Introduction Minimally invasive surgery is a widely used method for bone repair. Nano-hydroxyapatite (n-HA) is a non-toxic and harmless bone repair material. Aim To study the repair effect of n-HA/PA66 on the lumbar vertebrae injury of basketball players. Material and methods Firstly, the n-HA/PA66 material was prepared and its properties were studied. Then, the n-HA/PA66 material was used in a rabbit vertebral replacement experiment to study its bone coverage, fiber coverage and bone density. Afterwards, the material was used in vertebral injury treatment of basketball players to study its bone repairing effect. Results It was found that the fiber coverage of artificial vertebral bodies of n-HA/PA66 material tended to decrease with the increase of replacement time but no significant difference was found in the fiber coverage at different times (p > 0.05); the longer the replacement time, the greater the bone coverage, with statistically significant differences (p < 0.05); as the replacement time increased, the density and osteogenic volume increased; the Japanese Orthopaedic Association (JOA) scoring both 3 months and 6 months after surgery was greater than that immediately after surgery, with a significant difference (p < 0.05); the difference of angle of Cobb showed a decreasing trend with the increase of replacement time, and there was a significant difference (p < 0.05); follow-up results showed that the subjects had no rejection reaction. Conclusions N-HA material played an important role in basketball players’ bone repair treatment and is worth being promoted for application.
Collapse
|
26
|
Mechanism of Neuroprotection Against Experimental Spinal Cord Injury by Riluzole or Methylprednisolone. Neurochem Res 2017; 44:200-213. [PMID: 29290040 DOI: 10.1007/s11064-017-2459-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 12/12/2017] [Accepted: 12/26/2017] [Indexed: 12/29/2022]
Abstract
Any spinal cord injury carries the potential for persistent disability affecting motor, sensory and autonomic functions. To prevent this outcome, it is highly desirable to block a chain of deleterious reactions developing in the spinal areas immediately around the primary lesion. Thus, early timing of pharmacological neuroprotection should be one major strategy whose impact may be first studied with preclinical models. Using a simple in vitro model of the rat spinal cord it is possible to mimic pathological processes like excitotoxicity that damages neurons because of excessive glutamate receptor activation due to injury, or hypoxic/dysmetabolic insult that preferentially affects glia following vascular dysfunction. While ongoing research is exploring the various components of pathways leading to cell death, current treatment principally relies on the off-label use of riluzole (RLZ) or methylprednisolone sodium succinate (MPSS). The mechanism of action of these drugs is diverse as RLZ targets mainly neurons and MPSS targets glia. Even when applied after a transient excitotoxic stimulus, RLZ can provide effective prevention of secondary excitotoxic damage to premotoneurons, although not to motoneurons that remain very vulnerable. This observation indicates persistent inability to express locomotor activity despite pharmacological treatment conferring some histological protection. MPSS can protect glia from dysmetabolic insult, yet it remains poorly effective to prevent neuronal death. In summary, it appears that these pharmacological agents can produce delayed protection for certain cell types only, and that their combined administration does not provide additional benefit. The search should continue for better, mechanism-based neuroprotective agents.
Collapse
|
27
|
Polcyn R, Capone M, Hossain A, Matzelle D, Banik NL, Haque A. Enolase and Acute Spinal Cord Injury. JOURNAL OF CLINICAL & CELLULAR IMMUNOLOGY 2017; 8:536. [PMID: 29423333 PMCID: PMC5800513 DOI: 10.4172/2155-9899.1000536] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Rachel Polcyn
- Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, USA
| | - Mollie Capone
- Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, USA
- Department of Neurosurgery, Medical University of South Carolina, Charleston, USA
| | - Azim Hossain
- Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, USA
| | - Denise Matzelle
- Department of Neurosurgery, Medical University of South Carolina, Charleston, USA
- Department of Immunology, Ralph H. Johnson Veterans Administration Medical Center, Charleston, SC, USA
| | - Naren L. Banik
- Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, USA
- Department of Neurosurgery, Medical University of South Carolina, Charleston, USA
- Department of Immunology, Ralph H. Johnson Veterans Administration Medical Center, Charleston, SC, USA
| | - Azizul Haque
- Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, USA
| |
Collapse
|
28
|
Polcyn R, Capone M, Hossain A, Matzelle D, Banik NL, Haque A. Neuron specific enolase is a potential target for regulating neuronal cell survival and death: implications in neurodegeneration and regeneration. NEUROIMMUNOLOGY AND NEUROINFLAMMATION 2017; 4:254-257. [PMID: 29423430 PMCID: PMC5800407 DOI: 10.20517/2347-8659.2017.59] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Rachel Polcyn
- Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Mollie Capone
- Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
- Department of Neurosurgery, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Azim Hossain
- Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Denise Matzelle
- Department of Neurosurgery, Medical University of South Carolina, Charleston, SC 29425, USA
- Ralph H. Johnson Veterans Administration Medical Center, Charleston, SC 29401, USA
| | - Naren L. Banik
- Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
- Department of Neurosurgery, Medical University of South Carolina, Charleston, SC 29425, USA
- Ralph H. Johnson Veterans Administration Medical Center, Charleston, SC 29401, USA
| | - Azizul Haque
- Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
29
|
Spatio-temporal expression of Hexokinase-3 in the injured female rat spinal cords. Neurochem Int 2017; 113:23-33. [PMID: 29196144 DOI: 10.1016/j.neuint.2017.11.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 11/22/2017] [Accepted: 11/27/2017] [Indexed: 11/20/2022]
Abstract
Hexokinase-3 (HK3) is a member of hexokinase family, which can catalyze the first step of glucose metabolism. It can increase ATP levels, reduce the production of reactive oxygen species, increase mitochondrial biogenesis, protect mitochondrial membrane potential and play an antioxidant role. However, the change of its expression in spinal cord after injury is still unknown. In this study, we investigated the spatio-temporal expression of HK3 in the spinal cords by using a spinal cord injury (SCI) model in adult female Sprague-Dawley rats. Quantitative reverse transcription-PCR and western blot analysis revealed that HK3 could be detected in sham-opened spinal cords. After SCI, it gradually increased, reached a peak at 7 days post-injury (dpi), and then gradually decreased with the prolonging of injury time, but still maintained at a higher level for up to 28 dpi (the longest time evaluated in this study). Immunofluorescence staining showed that HK3 was found in GFAP+, β-tubulin III+ and IBA-1+ cells in sham-opened spinal cords. After SCI, in addition to the above-mentioned cells, it could also be found in CD45+ and CD68+ cells. These results demonstrate that HK3 is mainly expressed in astrocytes, neurons and microglia in normal spinal cords, and could rapidly increase in infiltrated leukocytes, activated microglia/macrophages and astrocytes after SCI. These data suggest that HK3 may be involved in the pathologic process of SCI by promoting glucose metabolism.
Collapse
|
30
|
Minocycline protects developing brain against ethanol-induced damage. Neuropharmacology 2017; 129:84-99. [PMID: 29146504 DOI: 10.1016/j.neuropharm.2017.11.019] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 10/20/2017] [Accepted: 11/10/2017] [Indexed: 12/24/2022]
Abstract
Fetal alcohol spectrum disorders (FASD) are caused by ethanol exposure during the pregnancy and is the leading cause of mental retardation. Ethanol exposure during the development results in the loss of neurons in the developing brain, which may underlie many neurobehavioral deficits associated with FASD. It is important to understand the mechanisms underlying ethanol-induced neuronal loss and develop appropriate therapeutic strategies. One of the potential mechanisms involves neuroimmune activation. Using a third trimester equivalent mouse model of ethanol exposure, we demonstrated that ethanol induced a wide-spread neuroapoptosis, microglial activation, and neuroinflammation in C57BL/6 mice. Minocycline is an antibiotic that inhibits microglial activation and alleviates neuroinflammation. We tested the hypothesis that minocycline may protect neurons ethanol-induced neuron death by inhibiting microglial activation and neuroinflammation. We showed that minocycline significantly inhibited ethanol-induced caspase-3 activation, microglial activation, and the expression of pro-inflammatory cytokines. In contrast, minocycline reversed ethanol inhibition of anti-inflammatory cytokines. Minocycline blocked ethanol-induced activation of GSK3β, a key mediator of neuroinflammation and microglial activation in the developing brain. Consistent with the in vivo observations, minocycline inhibited ethanol-induced the expression of pro-inflammatory cytokines and activation of GSK3β in a microglia cell line (SIM-9). GSK3β inhibitor eliminated ethanol activation of pro-inflammatory cytokines in SIM-9 cells. Co-cultures of cortical neurons and SIM-9 microglia cells sensitized neurons to alcohol-induced neuronal death. Minocycline protected neurons against ethanol-induced neuronal death in neurons/microglia co-cultures. Together, these results suggest that minocycline may ameliorate ethanol neurotoxicity in the developing by alleviating GSK3β-mediated neuroinflammation.
Collapse
|
31
|
Zendedel A, Mönnink F, Hassanzadeh G, Zaminy A, Ansar MM, Habib P, Slowik A, Kipp M, Beyer C. Estrogen Attenuates Local Inflammasome Expression and Activation after Spinal Cord Injury. Mol Neurobiol 2017; 55:1364-1375. [PMID: 28127698 DOI: 10.1007/s12035-017-0400-2] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 01/11/2017] [Indexed: 10/20/2022]
Abstract
17-estradiol (E2) is a neuroprotective hormone with a high anti-inflammatory potential in different neurological disorders. The inflammatory response initiated by spinal cord injury (SCI) involves the processing of interleukin-1beta (IL-1b) and IL-18 mediated by caspase-1 which is under the control of an intracellular multiprotein complex called inflammasome. We recently described in a SCI model that between 24 and 72 h post-injury, most of inflammasome components including IL-18, IL-1b, NLRP3, ASC, and caspase-1 are upregulated. In this study, we investigated the influence of E2 treatment after spinal cord contusion on inflammasome regulation. After contusion of T9 spinal segment, 12-week-old male Wistar rats were treated subcutaneously with E2 immediately after injury and every 12 h for the next 3 days. Behavioral scores were significantly improved in E2-treated animals compared to vehicle-treated groups. Functional improvement in E2-treated animals was paralleled by the attenuated expression of certain inflammasome components such as ASC, NLRP1b, and NLRP3 together with IL1b, IL-18, and caspase-1. On the histopathological level, microgliosis and oligodendrocyte injury was ameliorated. These findings support and extend the knowledge of the E2-mediated neuroprotective function during SCI. The control of the inflammasome machinery by E2 might be a missing piece of the puzzle to understand the anti-inflammatory potency of E2.
Collapse
Affiliation(s)
- Adib Zendedel
- Institute of Neuroanatomy, Faculty of Medicine, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany.,Giulan Neuroscience Research Center, Department of Anatomical Sciences, Faculty of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Fabian Mönnink
- Institute of Neuroanatomy, Faculty of Medicine, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Gholamreza Hassanzadeh
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Arash Zaminy
- Giulan Neuroscience Research Center, Department of Anatomical Sciences, Faculty of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Malek Masoud Ansar
- Giulan Neuroscience Research Center, Department of Anatomical Sciences, Faculty of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Pardes Habib
- Department of Neurology, RWTH Aachen, 52074, Aachen, Germany
| | - Alexander Slowik
- Institute of Neuroanatomy, Faculty of Medicine, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Markus Kipp
- Department of Anatomy II, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Cordian Beyer
- Institute of Neuroanatomy, Faculty of Medicine, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany.,JARA-Brain, 52074, Aachen, Germany
| |
Collapse
|
32
|
|
33
|
Jones ZB, Ren Y. Sphingolipids in spinal cord injury. INTERNATIONAL JOURNAL OF PHYSIOLOGY, PATHOPHYSIOLOGY AND PHARMACOLOGY 2016; 8:52-69. [PMID: 27570580 PMCID: PMC4981650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 07/19/2016] [Indexed: 06/06/2023]
Abstract
Spinal cord injury (SCI) is a debilitating condition that affects millions of individuals worldwide. Despite progress over the last few decades, the molecular mechanisms of secondary SCI that continue to occur days and weeks after the original trauma remain poorly understood. As a result, current therapies for SCI are only marginally effective. Sphingolipids, a diverse class of bioactive lipids, have been shown to regulate SCI repair and key secondary injury processes such as apoptosis, ischemia and inflammation. This review will discuss the numerous roles of sphingolipids and highlight the potential of sphingolipid-targeted therapies for SCI.
Collapse
Affiliation(s)
- Zachary B Jones
- Department of Biomedical Sciences, Florida State UniversityTallahassee, FL, USA
| | - Yi Ren
- Department of Biomedical Sciences, Florida State UniversityTallahassee, FL, USA
- Institute of Inflammation and Diseases, The First Affiliated Hospital of Wenzhou Medical UniversityWenzhou, China
| |
Collapse
|
34
|
Cytokine and Growth Factor Activation In Vivo and In Vitro after Spinal Cord Injury. Mediators Inflamm 2016; 2016:9476020. [PMID: 27418745 PMCID: PMC4935915 DOI: 10.1155/2016/9476020] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 05/18/2016] [Indexed: 12/22/2022] Open
Abstract
Spinal cord injury results in a life-disrupting series of deleterious interconnected mechanisms encompassed by the primary and secondary injury. These events are mediated by the upregulation of genes with roles in inflammation, transcription, and signaling proteins. In particular, cytokines and growth factors are signaling proteins that have important roles in the pathophysiology of SCI. The balance between the proinflammatory and anti-inflammatory effects of these molecules plays a critical role in the progression and outcome of the lesion. The excessive inflammatory Th1 and Th17 phenotypes observed after SCI tilt the scale towards a proinflammatory environment, which exacerbates the deleterious mechanisms present after the injury. These mechanisms include the disruption of the spinal cord blood barrier, edema and ion imbalance, in particular intracellular calcium and sodium concentrations, glutamate excitotoxicity, free radicals, and the inflammatory response contributing to the neurodegenerative process which is characterized by demyelination and apoptosis of neuronal tissue.
Collapse
|
35
|
Wu J, Maoqiang L, Fan H, Zhenyu B, Qifang H, Xuepeng W, Liulong Z. Rutin attenuates neuroinflammation in spinal cord injury rats. J Surg Res 2016; 203:331-7. [DOI: 10.1016/j.jss.2016.02.041] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2015] [Revised: 01/22/2016] [Accepted: 02/26/2016] [Indexed: 01/09/2023]
|
36
|
Samantaray S, Das A, Matzelle DC, Yu SP, Wei L, Varma A, Ray SK, Banik NL. Administration of low dose estrogen attenuates persistent inflammation, promotes angiogenesis, and improves locomotor function following chronic spinal cord injury in rats. J Neurochem 2016; 137:604-17. [PMID: 26998684 DOI: 10.1111/jnc.13610] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Revised: 01/22/2016] [Accepted: 02/17/2016] [Indexed: 02/06/2023]
Abstract
Spinal cord injury (SCI) causes loss of neurological function and, depending upon the severity of injury, may lead to paralysis. Currently, no FDA-approved pharmacotherapy is available for SCI. High-dose methylprednisolone is widely used, but this treatment is controversial. We have previously shown that low doses of estrogen reduces inflammation, attenuates cell death, and protects axon and myelin in SCI rats, but its effectiveness in recovery of function is not known. Therefore, the goal of this study was to investigate whether low doses of estrogen in post-SCI would reduce inflammation, protect cells and axons, and improve locomotor function during the chronic phase of injury. Injury (40 g.cm force) was induced at thoracic 10 in young adult male rats. Rats were treated with 10 or 100 μg 17β-estradiol (estrogen) for 7 days following SCI and compared with vehicle-treated injury and laminectomy (sham) controls. Histology (H&E staining), immunohistofluorescence, Doppler laser technique, and Western blotting were used to monitor tissue integrity, gliosis, blood flow, angiogenesis, the expression of angiogenic factors, axonal degeneration, and locomotor function (Basso, Beattie, and Bresnahan rating) following injury. To assess the progression of recovery, rats were sacrificed at 7, 14, or 42 days post injury. A reduction in glial reactivity, attenuation of axonal and myelin damage, protection of cells, increased expression of angiogenic factors and microvessel growth, and improved locomotor function were found following estrogen treatment compared with vehicle-treated SCI rats. These results suggest that treatment with a very low dose of estrogen has significant therapeutic implications for the improvement of locomotor function in chronic SCI. Experimental studies with low dose estrogen therapy in chronic spinal cord injury (SCI) demonstrated the potential for multi-active beneficial outcomes that could ameliorate the degenerative pathways in chronic SCI as shown in (a). Furthermore, the alterations in local spinal blood flow could be significantly alleviated with low dose estrogen therapy. This therapy led to the preservation of the structural integrity of the spinal cord (b), which in turn led to the improved functional recovery as shown (c).
Collapse
Affiliation(s)
- Supriti Samantaray
- Department of Neurology and Neurosurgery, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Arabinda Das
- Department of Neurology and Neurosurgery, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Denise C Matzelle
- Department of Neurology and Neurosurgery, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Shan P Yu
- Department of Anesthesia, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Ling Wei
- Department of Anesthesia, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Abhay Varma
- Department of Neurology and Neurosurgery, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Swapan K Ray
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, South Carolina, USA
| | - Naren L Banik
- Department of Neurology and Neurosurgery, Medical University of South Carolina, Charleston, South Carolina, USA.,Ralph H. Johnson Veterans Administration Medical Center, Charleston, South Carolina, USA
| |
Collapse
|
37
|
Dopamine D1 Receptor Agonist A-68930 Inhibits NLRP3 Inflammasome Activation, Controls Inflammation, and Alleviates Histopathology in a Rat Model of Spinal Cord Injury. Spine (Phila Pa 1976) 2016; 41:E330-4. [PMID: 26966979 DOI: 10.1097/brs.0000000000001287] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
STUDY DESIGN A randomized experimental study. OBJECTIVE The aim of this study was to investigate the therapeutic efficacy and molecular mechanisms of dopamine D1 receptor agonist A-68930 in spinal cord injury (SCI) rats. SUMMARY OF BACKGROUND DATA The inflammation induced by SCI includes maturation and secretion of pro-inflammatory cytokines interleukin (IL)-1β and IL-18 mediated by nucleotide-binding domain -like receptor protein 3 (NLRP3) inflammasome. Dopamine D1 receptor agonist A-68930 has been reported to exert neuroprotective effect via suppressing NLRP3 inflammasome activation in some central nervous injury models. However, whether A-68930 can exert nueroprotection in rat SCI models through inhibition of NLRP3 inflammasome activation has yet to be investigated. METHODS Eighty female Sprague-Dawley rats were randomly divided into 4 groups: sham group, SCI group, SCI + Vehicle (Veh) group, SCI + A-68930 group. The influences of A-68930 on the proinflammatory cytokines levels, histological changes, and locomotion scale were estimated. RESULTS SCI significantly promoted NLRP3 inflammasome activation and increased proinflammatory cytokines productions in SCI group as compared with sham group. A-68930 administration significantly inhibited NLRP3 inflammasome activation and reduced inflammatory cytokines levels. Moreover, A-68930 administration attenuated histopathology and promoted locomotion recovery. CONCLUSION A-68930 can attenuate tissue damage and improve neurological function recovery, and the mechanism may be related to the inhibition of NLRP3 inflammasome activation.
Collapse
|
38
|
Quercetin suppresses NLRP3 inflammasome activation and attenuates histopathology in a rat model of spinal cord injury. Spinal Cord 2016; 54:592-6. [DOI: 10.1038/sc.2015.227] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Revised: 10/28/2015] [Accepted: 11/21/2015] [Indexed: 12/14/2022]
|
39
|
Sámano C, Kaur J, Nistri A. A study of methylprednisolone neuroprotection against acute injury to the rat spinal cord in vitro. Neuroscience 2015; 315:136-49. [PMID: 26701292 DOI: 10.1016/j.neuroscience.2015.12.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 11/30/2015] [Accepted: 12/01/2015] [Indexed: 12/21/2022]
Abstract
Methylprednisolone sodium succinate (MPSS) has been proposed as a first-line treatment for acute spinal cord injury (SCI). Its clinical use remains, however, controversial because of the modest benefits and numerous side-effects. We investigated if MPSS could protect spinal neurons and glia using an in vitro model of the rat spinal cord that enables recording reflexes, fictive locomotion and morphological analysis of damage. With this model, a differential lesion affecting mainly either neurons or glia can be produced via kainate-evoked excitotoxicity or application of a pathological medium (lacking O2 and glucose), respectively. MPSS (6-10 μM) applied for 24 h after 1-h pathological medium protected astrocytes and oligodendrocytes especially in the ventrolateral white matter. This effect was accompanied by the return of slow, alternating oscillations (elicited by NMDA and 5-hydroxytryptamine (5-HT)) reminiscent of a sluggish fictive locomotor pattern. MPSS was, however, unable to reverse even a moderate neuronal loss and the concomitant suppression of fictive locomotion evoked by kainate (0.1 mM; 1 h). These results suggest that MPSS could, at least in part, contrast damage to spinal glia induced by a dysmetabolic state (associated to oxygen and glucose deprivation) and facilitate reactivation of spinal networks. Conversely, when even a minority of neurons was damaged by excitotoxicity, MPSS did not protect them nor did it restore network function in the current experimental model.
Collapse
Affiliation(s)
- C Sámano
- Departamento de Ciencias Naturales, Universidad Autónoma Metropolitana, Unidad Cuajimalpa, Mexico City, Mexico
| | - J Kaur
- Neuroscience Department, International School for Advanced Studies (SISSA), Trieste, Italy
| | - A Nistri
- Neuroscience Department, International School for Advanced Studies (SISSA), Trieste, Italy; SPINAL (Spinal Person Injury Neurorehabilitation Applied Laboratory) Laboratory, Istituto di Medicina Fisica e Riabilitazione, Udine, Italy.
| |
Collapse
|
40
|
Cox A, Varma A, Barry J, Vertegel A, Banik N. Nanoparticle Estrogen in Rat Spinal Cord Injury Elicits Rapid Anti-Inflammatory Effects in Plasma, Cerebrospinal Fluid, and Tissue. J Neurotrauma 2015; 32:1413-21. [PMID: 25845398 DOI: 10.1089/neu.2014.3730] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Persons with spinal cord injury (SCI) are in need of effective therapeutics. Estrogen (E2), as a steroid hormone, is a highly pleiotropic agent; with anti-inflammatory, anti-apoptotic, and neurotrophic properties, it is ideal for use in treatment of patients with SCI. Safety concerns around the use of high doses of E2 have limited clinical application, however. To address these concerns, low doses of E2 (25 μg and 2.5 μg) were focally delivered to the injured spinal cord using nanoparticles. A per-acute model (6 h after injury) was used to assess nanoparticle release of E2 into damaged spinal cord tissue; in addition, E2 was evaluated as a rapid anti-inflammatory. To assess inflammation, 27-plex cytokine/chemokine arrays were conducted in plasma, cerebrospinal fluid (CSF), and spinal cord tissue. A particular focus was placed on IL-6, GRO-KC, and MCP-1 as these have been identified from CSF in human studies as potential biomarkers in SCI. S100β, an additional proposed biomarker, was also assessed in spinal cord tissue only. Tissue concentrations of E2 were double those found in the plasma, indicating focal release. E2 showed rapid anti-inflammatory effects, significantly reducing interleukin (IL)-6, GRO-KC, MCP-1, and S100β in one or all compartments. Numerous additional targets of rapid E2 modulation were identified including: leptin, MIP-1α, IL-4, IL-2, IL-10, IFNγ, tumor necrosis factor-α, etc. These data further elucidate the rapid anti-inflammatory effects E2 exerts in an acute rat SCI model, have identified additional targets of estrogen efficacy, and suggest nanoparticle delivered estrogen may provide a safe and efficacious treatment option in persons with acute SCI.
Collapse
Affiliation(s)
- April Cox
- 1 Department of Neurology and Neurosurgery, Medical University of South Carolina , Charleston, South Carolina
| | - Abhay Varma
- 1 Department of Neurology and Neurosurgery, Medical University of South Carolina , Charleston, South Carolina
| | - John Barry
- 2 Department of Bioengineering, Clemson University , Clemson, South Carolina
| | - Alexey Vertegel
- 2 Department of Bioengineering, Clemson University , Clemson, South Carolina
| | - Naren Banik
- 1 Department of Neurology and Neurosurgery, Medical University of South Carolina , Charleston, South Carolina.,3 Ralph H. Johnson VA Medical Center , Charleston, South Carolina
| |
Collapse
|