1
|
Wu W, He Y, Chen Y, Fu Y, He S, Liu K, Qu JY. In vivo imaging in mouse spinal cord reveals that microglia prevent degeneration of injured axons. Nat Commun 2024; 15:8837. [PMID: 39397028 PMCID: PMC11471772 DOI: 10.1038/s41467-024-53218-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 10/02/2024] [Indexed: 10/15/2024] Open
Abstract
Microglia, the primary immune cells in the central nervous system, play a critical role in regulating neuronal function and fate through their interaction with neurons. Despite extensive research, the specific functions and mechanisms of microglia-neuron interactions remain incompletely understood. In this study, we demonstrate that microglia establish direct contact with myelinated axons at Nodes of Ranvier in the spinal cord of mice. The contact associated with neuronal activity occurs in a random scanning pattern. In response to axonal injury, microglia rapidly transform their contact into a robust wrapping form, preventing acute axonal degeneration from extending beyond the nodes. This wrapping behavior is dependent on the function of microglial P2Y12 receptors, which may be activated by ATP released through axonal volume-activated anion channels at the nodes. Additionally, voltage-gated sodium channels (NaV) and two-pore-domain potassium (K2P) channels contribute to the interaction between nodes and glial cells following injury, and inhibition of NaV delays axonal degeneration. Through in vivo imaging, our findings reveal a neuroprotective role of microglia during the acute phase of single spinal cord axon injury, achieved through neuron-glia interaction.
Collapse
Grants
- ITCPD/17-9 Innovation and Technology Commission (ITF)
- ITCPD/17-9 Innovation and Technology Commission (ITF)
- 32101211, 32192400 National Natural Science Foundation of China (National Science Foundation of China)
- 82171384 National Natural Science Foundation of China (National Science Foundation of China)
- the Hong Kong Research Grants Council through grants (16102122, 16102123, 16102421, 16102518, 16102920, T13-607/12R, T13-605/18W, T13-602/21N, C6002-17GF, C6001-19E);the Area of Excellence Scheme of the University Grants Committee (AoE/M-604/16, AOE/M-09/12) and the Hong Kong University of Science & Technology (HKUST) through grant 30 for 30 Research Initiative Scheme.
- Guangdong Basic and Applied Basic Research Foundation 2024A1515012414 Shenzhen Medical Research Fund (B2301004)
- Guangzhou Key Projects of Brain Science and Brain-Like Intelligence Technology (20200730009), Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions (2019SHIBS0001);the Area of Excellence Scheme of the University Grants Committee (AoE/M-604/16); Hong Kong Research Grants Council through grants (T13-602/21N, C6034-21G)
Collapse
Affiliation(s)
- Wanjie Wu
- Department of Electronic and Computer Engineering, The Hong Kong University of Science and Technology, Hong Kong, P. R. China
| | - Yingzhu He
- Department of Electronic and Computer Engineering, The Hong Kong University of Science and Technology, Hong Kong, P. R. China
| | - Yujun Chen
- Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong, P. R. China
| | - Yiming Fu
- Department of Electronic and Computer Engineering, The Hong Kong University of Science and Technology, Hong Kong, P. R. China
| | - Sicong He
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Kai Liu
- Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong, P. R. China.
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, Hong Kong, P. R. China.
- StateKey Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, P. R. China.
- Biomedical Research Institute, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, China.
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, Shenzhen, Guangdong, China.
- HKUST Shenzhen Research Institute, Guangdong, China.
- Shenzhen-Hong Kong Institute of Brain Science, Guangdong, China.
| | - Jianan Y Qu
- Department of Electronic and Computer Engineering, The Hong Kong University of Science and Technology, Hong Kong, P. R. China.
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, Hong Kong, P. R. China.
- Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Hong Kong, P. R. China.
| |
Collapse
|
2
|
Yu D, Zeng X, Aljuboori ZS, Dennison R, Wu L, Anderson JA, Teng YD. T12-L3 Nerve Transfer-Induced Locomotor Recovery in Rats with Thoracolumbar Contusion: Essential Roles of Sensory Input Rerouting and Central Neuroplasticity. Cells 2023; 12:2804. [PMID: 38132124 PMCID: PMC10741684 DOI: 10.3390/cells12242804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 11/28/2023] [Accepted: 12/05/2023] [Indexed: 12/23/2023] Open
Abstract
Locomotor recovery after spinal cord injury (SCI) remains an unmet challenge. Nerve transfer (NT), the connection of a functional/expendable peripheral nerve to a paralyzed nerve root, has long been clinically applied, aiming to restore motor control. However, outcomes have been inconsistent, suggesting that NT-induced neurological reinstatement may require activation of mechanisms beyond motor axon reinnervation (our hypothesis). We previously reported that to enhance rat locomotion following T13-L1 hemisection, T12-L3 NT must be performed within timeframes optimal for sensory nerve regrowth. Here, T12-L3 NT was performed for adult female rats with subacute (7-9 days) or chronic (8 weeks) mild (SCImi: 10 g × 12.5 mm) or moderate (SCImo: 10 g × 25 mm) T13-L1 thoracolumbar contusion. For chronic injuries, T11-12 implantation of adult hMSCs (1-week before NT), post-NT intramuscular delivery of FGF2, and environmentally enriched/enlarged (EEE) housing were provided. NT, not control procedures, qualitatively improved locomotion in both SCImi groups and animals with subacute SCImo. However, delayed NT did not produce neurological scale upgrading conversion for SCImo rats. Ablation of the T12 ventral/motor or dorsal/sensory root determined that the T12-L3 sensory input played a key role in hindlimb reanimation. Pharmacological, electrophysiological, and trans-synaptic tracing assays revealed that NT strengthened integrity of the propriospinal network, serotonergic neuromodulation, and the neuromuscular junction. Besides key outcomes of thoracolumbar contusion modeling, the data provides the first evidence that mixed NT-induced locomotor efficacy may rely pivotally on sensory rerouting and pro-repair neuroplasticity to reactivate neurocircuits/central pattern generators. The finding describes a novel neurobiology mechanism underlying NT, which can be targeted for development of innovative neurotization therapies.
Collapse
Affiliation(s)
- Dou Yu
- Department of Physical Medicine and Rehabilitation, Harvard Medical School, Boston, MA 02129, USA
- Department of Neurosurgery, Harvard Medical School, Boston, MA 02115, USA
- Laboratory of SCI, Stem Cell and Recovery Neurobiology Research, Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital, Mass General Brigham, Harvard Medical School, Boston, MA 02129, USA
| | - Xiang Zeng
- Department of Physical Medicine and Rehabilitation, Harvard Medical School, Boston, MA 02129, USA
- Department of Neurosurgery, Harvard Medical School, Boston, MA 02115, USA
- Laboratory of SCI, Stem Cell and Recovery Neurobiology Research, Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital, Mass General Brigham, Harvard Medical School, Boston, MA 02129, USA
| | - Zaid S. Aljuboori
- Department of Physical Medicine and Rehabilitation, Harvard Medical School, Boston, MA 02129, USA
- Department of Neurosurgery, Harvard Medical School, Boston, MA 02115, USA
- Laboratory of SCI, Stem Cell and Recovery Neurobiology Research, Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital, Mass General Brigham, Harvard Medical School, Boston, MA 02129, USA
| | - Rachel Dennison
- Department of Physical Medicine and Rehabilitation, Harvard Medical School, Boston, MA 02129, USA
- Department of Neurosurgery, Harvard Medical School, Boston, MA 02115, USA
- Laboratory of SCI, Stem Cell and Recovery Neurobiology Research, Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital, Mass General Brigham, Harvard Medical School, Boston, MA 02129, USA
| | - Liquan Wu
- Department of Physical Medicine and Rehabilitation, Harvard Medical School, Boston, MA 02129, USA
- Department of Neurosurgery, Harvard Medical School, Boston, MA 02115, USA
- Laboratory of SCI, Stem Cell and Recovery Neurobiology Research, Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital, Mass General Brigham, Harvard Medical School, Boston, MA 02129, USA
| | - Jamie A. Anderson
- Department of Physical Medicine and Rehabilitation, Harvard Medical School, Boston, MA 02129, USA
- Department of Neurosurgery, Harvard Medical School, Boston, MA 02115, USA
- Laboratory of SCI, Stem Cell and Recovery Neurobiology Research, Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital, Mass General Brigham, Harvard Medical School, Boston, MA 02129, USA
| | - Yang D. Teng
- Department of Physical Medicine and Rehabilitation, Harvard Medical School, Boston, MA 02129, USA
- Department of Neurosurgery, Harvard Medical School, Boston, MA 02115, USA
- Laboratory of SCI, Stem Cell and Recovery Neurobiology Research, Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital, Mass General Brigham, Harvard Medical School, Boston, MA 02129, USA
- Neurotrauma Recovery Research, Spaulding Rehabilitation Hospital Network, Mass General Brigham, Harvard Medical School, Boston, MA 02129, USA
| |
Collapse
|
3
|
Davis JA, Grau JW. Protecting the injured central nervous system: Do anesthesia or hypothermia ameliorate secondary injury? Exp Neurol 2023; 363:114349. [PMID: 36775099 DOI: 10.1016/j.expneurol.2023.114349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 01/13/2023] [Accepted: 02/06/2023] [Indexed: 02/12/2023]
Abstract
Traumatic injury to the central nervous system (CNS) and stroke initiate a cascade of processes that expand the area of tissue loss. The current review considers recent studies demonstrating that the induction of an anesthetic state or cooling the affected tissue (hypothermia) soon after injury can have a therapeutic effect. We first provide an overview of the neurobiological processes that fuel tissue loss after traumatic brain injury (TBI), spinal cord injury (SCI) and stroke. We then examine the rehabilitative effectiveness of therapeutic anesthesia across a variety of drug categories through a systematic review of papers in the PubMed database. We also review the therapeutic benefits hypothermia, another treatment that quells neural activity. We conclude by considering factors related to the safety, efficacy and timing of treatment, as well as the mechanisms of action. Clinical implications are also discussed.
Collapse
Affiliation(s)
- Jacob A Davis
- Cellular and Behavioral Neuroscience, Department of Psychology, Texas A&M University, College Station, TX 77843, USA.
| | - James W Grau
- Cellular and Behavioral Neuroscience, Department of Psychology, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
4
|
Katikou P, Gokbulut C, Kosker AR, Campàs M, Ozogul F. An Updated Review of Tetrodotoxin and Its Peculiarities. Mar Drugs 2022; 20:md20010047. [PMID: 35049902 PMCID: PMC8780202 DOI: 10.3390/md20010047] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 12/28/2021] [Accepted: 12/30/2021] [Indexed: 12/19/2022] Open
Abstract
Tetrodotoxin (TTX) is a crystalline, weakly basic, colorless organic substance and is one of the most potent marine toxins known. Although TTX was first isolated from pufferfish, it has been found in numerous other marine organisms and a few terrestrial species. Moreover, tetrodotoxication is still an important health problem today, as TTX has no known antidote. TTX poisonings were most commonly reported from Japan, Thailand, and China, but today the risk of TTX poisoning is spreading around the world. Recent studies have shown that TTX-containing fish are being found in other regions of the Pacific and in the Indian Ocean, as well as the Mediterranean Sea. This review aims to summarize pertinent information available to date on the structure, origin, distribution, mechanism of action of TTX and analytical methods used for the detection of TTX, as well as on TTX-containing organisms, symptoms of TTX poisoning, and incidence worldwide.
Collapse
Affiliation(s)
- Panagiota Katikou
- Ministry of Rural Development and Food, Directorate of Research, Innovation and Education, Hapsa & Karatasou 1, 54626 Thessaloniki, Greece
- Correspondence: (P.K.); (F.O.)
| | - Cengiz Gokbulut
- Department of Pharmacology, Faculty of Medicine, Balikesir University, Balikesir 10145, Turkey;
| | - Ali Rıza Kosker
- Department of Seafood Processing Technology, Faculty of Fisheries, Cukurova University, Adana 01330, Turkey;
| | - Mònica Campàs
- IRTA, Ctra Poble Nou km 5.5, 43540 Sant Carles de la Ràpita, Spain;
| | - Fatih Ozogul
- Department of Seafood Processing Technology, Faculty of Fisheries, Cukurova University, Adana 01330, Turkey;
- Correspondence: (P.K.); (F.O.)
| |
Collapse
|
5
|
Bucciarelli GM, Lechner M, Fontes A, Kats LB, Eisthen HL, Shaffer HB. From Poison to Promise: The Evolution of Tetrodotoxin and Its Potential as a Therapeutic. Toxins (Basel) 2021; 13:toxins13080517. [PMID: 34437388 PMCID: PMC8402337 DOI: 10.3390/toxins13080517] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 07/17/2021] [Accepted: 07/20/2021] [Indexed: 11/16/2022] Open
Abstract
Tetrodotoxin (TTX) is a potent neurotoxin that was first identified in pufferfish but has since been isolated from an array of taxa that host TTX-producing bacteria. However, determining its origin, ecosystem roles, and biomedical applications has challenged researchers for decades. Recognized as a poison and for its lethal effects on humans when ingested, TTX is primarily a powerful sodium channel inhibitor that targets voltage-gated sodium channels, including six of the nine mammalian isoforms. Although lethal doses for humans range from 1.5-2.0 mg TTX (blood level 9 ng/mL), when it is administered at levels far below LD50, TTX exhibits therapeutic properties, especially to treat cancer-related pain, neuropathic pain, and visceral pain. Furthermore, TTX can potentially treat a variety of medical ailments, including heroin and cocaine withdrawal symptoms, spinal cord injuries, brain trauma, and some kinds of tumors. Here, we (i) describe the perplexing evolution and ecology of tetrodotoxin, (ii) review its mechanisms and modes of action, and (iii) offer an overview of the numerous ways it may be applied as a therapeutic. There is much to be explored in these three areas, and we offer ideas for future research that combine evolutionary biology with therapeutics. The TTX system holds great promise as a therapeutic and understanding the origin and chemical ecology of TTX as a poison will only improve its general benefit to humanity.
Collapse
Affiliation(s)
- Gary M. Bucciarelli
- Department of Ecology and Evolutionary Biology & UCLA La Kretz Center for California Conservation Science, Institute of the Environment and Sustainability, University of California, Los Angeles, CA 90095, USA; (M.L.); (H.B.S.)
- Correspondence:
| | - Maren Lechner
- Department of Ecology and Evolutionary Biology & UCLA La Kretz Center for California Conservation Science, Institute of the Environment and Sustainability, University of California, Los Angeles, CA 90095, USA; (M.L.); (H.B.S.)
| | - Audrey Fontes
- Natural Science Division, Pepperdine University, Malibu, CA 90263, USA; (A.F.); (L.B.K.)
| | - Lee B. Kats
- Natural Science Division, Pepperdine University, Malibu, CA 90263, USA; (A.F.); (L.B.K.)
| | - Heather L. Eisthen
- Department of Integrative Biology, Michigan State University, East Lansing, MI 48824, USA;
| | - H. Bradley Shaffer
- Department of Ecology and Evolutionary Biology & UCLA La Kretz Center for California Conservation Science, Institute of the Environment and Sustainability, University of California, Los Angeles, CA 90095, USA; (M.L.); (H.B.S.)
| |
Collapse
|
6
|
Pizzolato C, Gunduz MA, Palipana D, Wu J, Grant G, Hall S, Dennison R, Zafonte RD, Lloyd DG, Teng YD. Non-invasive approaches to functional recovery after spinal cord injury: Therapeutic targets and multimodal device interventions. Exp Neurol 2021; 339:113612. [DOI: 10.1016/j.expneurol.2021.113612] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 12/24/2020] [Accepted: 01/11/2021] [Indexed: 12/16/2022]
|
7
|
Jeffery ND, Olby NJ, Moore SA. Clinical Trial Design-A Review-With Emphasis on Acute Intervertebral Disc Herniation. Front Vet Sci 2020; 7:583. [PMID: 33134333 PMCID: PMC7512142 DOI: 10.3389/fvets.2020.00583] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 07/20/2020] [Indexed: 11/13/2022] Open
Abstract
There is a clear need for new methods of treatment of acute disc herniation in dogs, most obviously to address the permanent loss of function that can arise because of the associated spinal cord injury. Clinical trials form the optimal method to introduce new therapies into everyday clinical practice because they are a reliable source of unbiased evidence of effectiveness. Although many designs are available, parallel cohort trials are most widely applicable to acute disc herniation in dogs. In this review another key trial design decision—that between pragmatic and explanatory approaches—is highlighted and used as a theme to illustrate the close relationship between trial objective and design. Acute disc herniation, and acute spinal cord injury, is common in dogs and there is a multitude of candidate interventions that could be trialed. Most current obstacles to large-scale clinical trials in dogs can be overcome by collaboration and cooperation amongst interested veterinarians.
Collapse
Affiliation(s)
- Nick D Jeffery
- Department of Small Animal Clinical Sciences, Texas A&M University, College Station, TX, United States
| | - Natasha J Olby
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
| | - Sarah A Moore
- Department of Veterinary Clinical Sciences, The Ohio State University, Columbus, OH, United States
| | | |
Collapse
|
8
|
Teng YD. Functional Multipotency of Stem Cells and Recovery Neurobiology of Injured Spinal Cords. Cell Transplant 2019; 28:451-459. [PMID: 31134830 PMCID: PMC6628559 DOI: 10.1177/0963689719850088] [Citation(s) in RCA: 19] [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: 02/21/2019] [Revised: 03/31/2019] [Accepted: 04/19/2019] [Indexed: 02/06/2023] Open
Abstract
This invited concise review was written for the special issue of Cell Transplantation to celebrate the 25th anniversary of the American Society for Neural Therapy and Repair (ASNTR). I aimed to present a succinct summary of two interweaved lines of research work carried out by my team members and collaborators over the past decade. Since the middle of the 20th century, biomedical research has been driven overwhelmingly by molecular technology-based focal endeavors. Our investigative undertakings, however, were orchestrated to define and propose novel theoretical frameworks to enhance the field's ability to overcome complex neurological disorders. The effort has engendered two important academic concepts: Functional Multipotency of Stem Cells, and Recovery Neurobiology of Injured Spinal Cords. Establishing these theories was facilitated by academic insight gleaned from stem cell-based multimodal cross-examination studies using tactics of material science, systems neurobiology, glial biology, and neural oncology. It should be emphasized that the collegial environment cultivated by the mission of the ASNTR greatly promoted the efficacy of inter-laboratory collaborations. Notably, our findings have shed new light on fundamentals of stem cell biology and adult mammalian spinal cord neurobiology. Moreover, the novel academic leads have enabled determination of potential therapeutic targets to restore function for spinal cord injury and neurodegenerative diseases.
Collapse
Affiliation(s)
- Yang D. Teng
- Department of Physical Medicine and Rehabilitation, Harvard Medical
School/Spaulding Rehabilitation Hospital Network, Charlestown, USA
- Department of Neurosurgery, Harvard Medical School/Brigham and Women’s
Hospital, Boston, USA
- Division of SCI Research, Veterans Affairs Boston Healthcare System, Boston,
USA
| |
Collapse
|
9
|
Page JC, Park J, Chen Z, Cao P, Shi R. Parallel Evaluation of Two Potassium Channel Blockers in Restoring Conduction in Mechanical Spinal Cord Injury in Rat. J Neurotrauma 2018; 35:1057-1068. [PMID: 29228863 DOI: 10.1089/neu.2017.5297] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Myelin damage is a hallmark of spinal cord injury (SCI), and potassium channel blocker (PCB) is proven effective to restore axonal conduction and regain neurological function. Aiming to improve this therapy beyond the U.S. Food and Drug Administration-approved 4-aminopyridine (4-AP), we have developed multiple new PCBs, with 4-aminopyridine-3-methanol (4-AP-3-MeOH) being the most potent and effective. The current study evaluated two PCBs, 4-AP-3-MeOH and 4-AP, in parallel in both ex vivo and in vivo rat mechanical SCI models. Specifically, 4-AP-3-MeOH induced significantly greater augmentation of axonal conduction than 4-AP in both acute and chronic injury. 4-AP-3-MeOH had no negative influence on the electrical responsiveness of rescued axons whereas 4-AP-recruited axons displayed a reduced ability to follow multiple stimuli. In addition, 4-AP-3-MeOH can be applied intraperitoneally at a dose that is at least 5 times higher (5 mg/kg) than that of 4-AP (1 mg/kg) in vivo. Further, 5 mg/kg of 4-AP-3-MeOH significantly improved motor function whereas both 4-AP-3-MeOH (1 and 5 mg/kg) and, to a lesser degree, 4-AP (1 mg/kg) alleviated neuropathic pain-like behavior when applied in rats 2 weeks post-SCI. Based on these and other findings, we conclude that 4-AP-3-MeOH appears to be more advantageous over 4-AP in restoring axonal conduction because of the combination of its higher efficacy in enhancing the amplitude of compound action potential, lesser negative effect on axonal responsiveness to multiple stimuli, and wider therapeutic range in both ex vivo and in vivo application. As a result, 4-AP-3-MeOH has emerged as a strong alternative to 4-AP that can complement the effectiveness, and even partially overcome the shortcomings, of 4-AP in the treatment of neurotrauma and degenerative diseases where myelin damage is implicated.
Collapse
Affiliation(s)
- Jessica C Page
- 1 Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University , West Lafayette, Indiana
| | - Jonghyuck Park
- 2 Weldon School of Biomedical Engineering, Purdue University , West Lafayette, Indiana
| | - Zhe Chen
- 3 Department of Orthopedics, Rui-Jin Hospital, School of Medicine, Shanghai Jiao-tong University , Institute of Trauma and Orthopedics, Shanghai, China
| | - Peng Cao
- 3 Department of Orthopedics, Rui-Jin Hospital, School of Medicine, Shanghai Jiao-tong University , Institute of Trauma and Orthopedics, Shanghai, China
| | - Riyi Shi
- 1 Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University , West Lafayette, Indiana.,2 Weldon School of Biomedical Engineering, Purdue University , West Lafayette, Indiana
| |
Collapse
|
10
|
Acute spinal cord injury: A review of pathophysiology and potential of non-steroidal anti-inflammatory drugs for pharmacological intervention. J Chem Neuroanat 2018; 87:25-31. [DOI: 10.1016/j.jchemneu.2017.08.001] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 08/07/2017] [Accepted: 08/07/2017] [Indexed: 12/21/2022]
|
11
|
Kjell J, Olson L. Rat models of spinal cord injury: from pathology to potential therapies. Dis Model Mech 2017; 9:1125-1137. [PMID: 27736748 PMCID: PMC5087825 DOI: 10.1242/dmm.025833] [Citation(s) in RCA: 243] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
A long-standing goal of spinal cord injury research is to develop effective spinal cord repair strategies for the clinic. Rat models of spinal cord injury provide an important mammalian model in which to evaluate treatment strategies and to understand the pathological basis of spinal cord injuries. These models have facilitated the development of robust tests for assessing the recovery of locomotor and sensory functions. Rat models have also allowed us to understand how neuronal circuitry changes following spinal cord injury and how recovery could be promoted by enhancing spontaneous regenerative mechanisms and by counteracting intrinsic inhibitory factors. Rat studies have also revealed possible routes to rescuing circuitry and cells in the acute stage of injury. Spatiotemporal and functional studies in these models highlight the therapeutic potential of manipulating inflammation, scarring and myelination. In addition, potential replacement therapies for spinal cord injury, including grafts and bridges, stem primarily from rat studies. Here, we discuss advantages and disadvantages of rat experimental spinal cord injury models and summarize knowledge gained from these models. We also discuss how an emerging understanding of different forms of injury, their pathology and degree of recovery has inspired numerous treatment strategies, some of which have led to clinical trials. Summary: In this Review, we discuss the advantages and disadvantages of the rat for studies of experimental spinal cord injury and summarize the knowledge gained from such studies.
Collapse
Affiliation(s)
- Jacob Kjell
- Department of Physiological Genomics, Ludwig-Maximilians-Universität München, Munich 80336, Germany
| | - Lars Olson
- Department of Neuroscience, Karolinska Institutet, Stockholm 171 77, Sweden
| |
Collapse
|
12
|
Nardone R, Florea C, Höller Y, Brigo F, Versace V, Lochner P, Golaszewski S, Trinka E. Rodent, large animal and non-human primate models of spinal cord injury. ZOOLOGY 2017; 123:101-114. [PMID: 28720322 DOI: 10.1016/j.zool.2017.06.004] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Revised: 06/02/2017] [Accepted: 06/02/2017] [Indexed: 01/05/2023]
Abstract
In this narrative review we aimed to assess the usefulness of the different animal models in identifying injury mechanisms and developing therapies for humans suffering from spinal cord injury (SCI). Results obtained from rodent studies are useful but, due to the anatomical, molecular and functional differences, confirmation of these findings in large animals or non-human primates may lead to basic discoveries that cannot be made in rodent models and that are more useful for developing treatment strategies in humans. SCI in dogs can be considered as intermediate between rodent models and human clinical trials, but the primate models could help to develop appropriate methods that might be more relevant to humans. Ideally, an animal model should meet the requirements of availability and repeatability as well as reproduce the anatomical features and the clinical pathological changing process of SCI. An animal model that completely simulates SCI in humans does not exist. The different experimental models of SCI have advantages and disadvantages for investigating the different aspects of lesion development, recovery mechanisms and potential therapeutic interventions. The potential advantages of non-human primate models include genetic similarities, similar caliber/length of the spinal cord as well as biological and physiological responses to injury which are more similar to humans. Among the potential disadvantages, high operating costs, infrastructural requirements and ethical concerns should be considered. The translation from experimental repair strategies to clinical applications needs to be investigated in future carefully designed studies.
Collapse
Affiliation(s)
- Raffaele Nardone
- Department of Neurology, Christian Doppler Klinik, Paracelsus Medical University, Ignaz-Harrer-Str. 79, A-5020, Salzburg, Austria; Department of Neurology, Franz Tappeiner Hospital, Via Rossini 5, I-39012, Merano, Italy; Spinal Cord Injury and Tissue Regeneration Center, Paracelsus Medical University, Ignaz-Harrer-Str. 79, A-5020, Salzburg, Austria.
| | - Cristina Florea
- Department of Neurology, Christian Doppler Klinik, Paracelsus Medical University, Ignaz-Harrer-Str. 79, A-5020, Salzburg, Austria
| | - Yvonne Höller
- Department of Neurology, Christian Doppler Klinik, Paracelsus Medical University, Ignaz-Harrer-Str. 79, A-5020, Salzburg, Austria
| | - Francesco Brigo
- Department of Neurology, Franz Tappeiner Hospital, Via Rossini 5, I-39012, Merano, Italy; Department of Neurosciences, Biomedicine and Movement Sciences, Section of Clinical Neurology, University of Verona, Piazzale L.A. Scuro, I-37134 Verona, Italy
| | - Viviana Versace
- Department of Neurorehabilitation, Hospital of Vipiteno, Via Santa Margherita 24, I-39049, Italy
| | - Piergiorgio Lochner
- Department of Neurology, Saarland University Medical Center, Kirrberger-Str. 100, D-66421 Homburg, Germany
| | - Stefan Golaszewski
- Department of Neurology, Christian Doppler Klinik, Paracelsus Medical University, Ignaz-Harrer-Str. 79, A-5020, Salzburg, Austria
| | - Eugen Trinka
- Department of Neurology, Christian Doppler Klinik, Paracelsus Medical University, Ignaz-Harrer-Str. 79, A-5020, Salzburg, Austria; Spinal Cord Injury and Tissue Regeneration Center, Paracelsus Medical University, Ignaz-Harrer-Str. 79, A-5020, Salzburg, Austria
| |
Collapse
|
13
|
Ahmad M, Zakaria A, Almutairi KM. Effectiveness of minocycline and FK506 alone and in combination on enhanced behavioral and biochemical recovery from spinal cord injury in rats. Pharmacol Biochem Behav 2016; 145:45-54. [PMID: 27106204 DOI: 10.1016/j.pbb.2016.04.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Revised: 04/15/2016] [Accepted: 04/18/2016] [Indexed: 11/15/2022]
Abstract
Injury to the spinal cord results in immediate physical damage (primary injury) followed by a prolonged posttraumatic inflammatory disorder (secondary injury). The present study aimed to investigate the neuroprotective effects of minocycline and FK506 (Tacrolimus) individually and in combination on recovery from experimental spinal cord injury (SCI). Young adult male rats were subjected to experimental SCI by weight compression method. Minocycline (50mg/kg) and FK506 (1mg/kg) were administered orally in combination and individually to the SCI group daily for three weeks. During these three weeks, the recovery was measured using behavioral motor parameters (including BBB, Tarlov and other scorings) every other day for 29days after SCI. Thereafter, the animals were sacrificed and the segment of the spinal cord centered at the injury site was removed for the histopathological studies as well as for biochemical analysis of monoamines such as 5-hydroxytryptamine (5-HT) and 5-hydroxy-indolacetic acid (5-HIAA) and some oxidative stress indices, such as thiobarbituric acid-reactive substances (TBARS), total glutathione (GSH) and myeloperoxidase (MPO). All behavioral results indicated that both drugs induced significant recovery from SCI with respect to time. The biochemical and histopathological results supported the behavioral findings, revealing significant recovery in the regeneration of the injured spinal tissues, the monoamine levels, and the oxidative stress indices. Overall, the effects of the tested drugs for SCI recovery were as follows: FK506+minocycline>minocycline>FK506 in all studied parameters. Thus, minocycline and FK506 may prove to be a potential therapy cocktail to treat acute SCI. However, further studies are warranted.
Collapse
Affiliation(s)
- Mohammad Ahmad
- Department of Medical Surgical Nursing, College of Nursing, King Saud University, Riyadh, Saudi Arabia.
| | - Abdulrahim Zakaria
- Department of Rehabilitation Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Khalid M Almutairi
- Department of Community Health Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
14
|
Vardjan N, Horvat A, Anderson JE, Yu D, Croom D, Zeng X, Lužnik Z, Kreft M, Teng YD, Kirov SA, Zorec R. Adrenergic activation attenuates astrocyte swelling induced by hypotonicity and neurotrauma. Glia 2016; 64:1034-49. [PMID: 27018061 DOI: 10.1002/glia.22981] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Revised: 02/18/2016] [Accepted: 02/18/2016] [Indexed: 12/29/2022]
Abstract
Edema in the central nervous system can rapidly result in life-threatening complications. Vasogenic edema is clinically manageable, but there is no established medical treatment for cytotoxic edema, which affects astrocytes and is a primary trigger of acute post-traumatic neuronal death. To test the hypothesis that adrenergic receptor agonists, including the stress stimulus epinephrine protects neural parenchyma from damage, we characterized its effects on hypotonicity-induced cellular edema in cortical astrocytes by in vivo and in vitro imaging. After epinephrine administration, hypotonicity-induced swelling of astrocytes was markedly reduced and cytosolic 3'-5'-cyclic adenosine monophosphate (cAMP) was increased, as shown by a fluorescence resonance energy transfer nanosensor. Although, the kinetics of epinephrine-induced cAMP signaling was slowed in primary cortical astrocytes exposed to hypotonicity, the swelling reduction by epinephrine was associated with an attenuated hypotonicity-induced cytosolic Ca(2+) excitability, which may be the key to prevent astrocyte swelling. Furthermore, in a rat model of spinal cord injury, epinephrine applied locally markedly reduced neural edema around the contusion epicenter. These findings reveal new targets for the treatment of cellular edema in the central nervous system.
Collapse
Affiliation(s)
- Nina Vardjan
- Celica, BIOMEDICAL, Tehnološki park 24, Ljubljana, 1000, Slovenia.,Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloška 4, Ljubljana, 1000, Slovenia
| | - Anemari Horvat
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloška 4, Ljubljana, 1000, Slovenia
| | - Jamie E Anderson
- Departments of Neurosurgery and Physical Medicine & Rehabilitation, Harvard Medical School, Boston, Massachusetts
| | - Dou Yu
- Departments of Neurosurgery and Physical Medicine & Rehabilitation, Harvard Medical School, Boston, Massachusetts
| | - Deborah Croom
- Brain and Behaviour Discovery Institute, Medical College of Georgia, Augusta, Georgia.,Department of Neurosurgery, Medical College of Georgia, Augusta, Georgia
| | - Xiang Zeng
- Departments of Neurosurgery and Physical Medicine & Rehabilitation, Harvard Medical School, Boston, Massachusetts
| | - Zala Lužnik
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloška 4, Ljubljana, 1000, Slovenia
| | - Marko Kreft
- Celica, BIOMEDICAL, Tehnološki park 24, Ljubljana, 1000, Slovenia.,Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloška 4, Ljubljana, 1000, Slovenia.,Department of Biology, Biotechnical Faculty, University of Ljubljana, Večna pot 111, Ljubljana, 1000, Slovenia
| | - Yang D Teng
- Departments of Neurosurgery and Physical Medicine & Rehabilitation, Harvard Medical School, Boston, Massachusetts.,Division of SCI Research, VA Boston Healthcare System, Boston, Massachusetts
| | - Sergei A Kirov
- Brain and Behaviour Discovery Institute, Medical College of Georgia, Augusta, Georgia.,Department of Neurosurgery, Medical College of Georgia, Augusta, Georgia
| | - Robert Zorec
- Celica, BIOMEDICAL, Tehnološki park 24, Ljubljana, 1000, Slovenia.,Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloška 4, Ljubljana, 1000, Slovenia
| |
Collapse
|
15
|
Jin Y, Bouyer J, Shumsky JS, Haas C, Fischer I. Transplantation of neural progenitor cells in chronic spinal cord injury. Neuroscience 2016; 320:69-82. [PMID: 26852702 DOI: 10.1016/j.neuroscience.2016.01.066] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Revised: 01/07/2016] [Accepted: 01/29/2016] [Indexed: 01/24/2023]
Abstract
Previous studies demonstrated that neural progenitor cells (NPCs) transplanted into a subacute contusion injury improve motor, sensory, and bladder function. In this study we tested whether transplanted NPCs can also improve functional recovery after chronic spinal cord injury (SCI) alone or in combination with the reduction of glial scar and neurotrophic support. Adult rats received a T10 moderate contusion. Thirteen weeks after the injury they were divided into four groups and received either: 1. Medium (control), 2. NPC transplants, 3. NPC+lentivirus vector expressing chondroitinase, or 4. NPC+lentivirus vectors expressing chondroitinase and neurotrophic factors. During the 8 weeks post-transplantation the animals were tested for functional recovery and eventually analyzed by anatomical and immunohistochemical assays. The behavioral tests for motor and sensory function were performed before and after injury, and weekly after transplantation, with some animals also tested for bladder function at the end of the experiment. Transplant survival in the chronic injury model was variable and showed NPCs at the injury site in 60% of the animals in all transplantation groups. The NPC transplants comprised less than 40% of the injury site, without significant anatomical or histological differences among the groups. All groups also showed similar patterns of functional deficits and recovery in the 12 weeks after injury and in the 8 weeks after transplantation using the Basso, Beattie, and Bresnahan rating score, the grid test, and the Von Frey test for mechanical allodynia. A notable exception was group 4 (NPC together with chondroitinase and neurotrophins), which showed a significant improvement in bladder function. This study underscores the therapeutic challenges facing transplantation strategies in a chronic SCI in which even the inclusion of treatments designed to reduce scarring and increase neurotrophic support produce only modest functional improvements. Further studies will have to identify the combination of acute and chronic interventions that will augment the survival and efficacy of neural cell transplants.
Collapse
Affiliation(s)
- Y Jin
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia PA 19129, United States.
| | - J Bouyer
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia PA 19129, United States
| | - J S Shumsky
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia PA 19129, United States
| | - C Haas
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia PA 19129, United States
| | - I Fischer
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia PA 19129, United States.
| |
Collapse
|
16
|
Transcriptional analysis reveals distinct subtypes in amyotrophic lateral sclerosis: implications for personalized therapy. Future Med Chem 2016; 7:1335-59. [PMID: 26144267 DOI: 10.4155/fmc.15.60] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is an incurable disease, caused by the loss of the upper and lower motor neurons. The lack of therapeutic progress is mainly due to the insufficient understanding of complexity and heterogeneity underlying the pathogenic mechanisms of ALS. Recently, we analyzed whole-genome expression profiles of motor cortex of sporadic ALS patients, classifying them into two subgroups characterized by differentially expressed genes and pathways. Some of the deregulated genes encode proteins, which are primary targets of drugs currently in preclinical or clinical studies for several clinical conditions, including neurodegenerative diseases. In this review, we discuss in-depth the potential role of these candidate targets in ALS pathogenesis, highlighting their possible relevance for personalized ALS treatments.
Collapse
|
17
|
Alvarez P, Levine JD. Antihyperalgesic effect of tetrodotoxin in rat models of persistent muscle pain. Neuroscience 2015; 311:499-507. [PMID: 26548414 DOI: 10.1016/j.neuroscience.2015.10.059] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 10/26/2015] [Accepted: 10/30/2015] [Indexed: 11/29/2022]
Abstract
Persistent muscle pain is a common and disabling symptom for which available treatments have limited efficacy. Since tetrodotoxin (TTX) displays a marked antinociceptive effect in models of persistent cutaneous pain, we tested its local antinociceptive effect in rat models of muscle pain induced by inflammation, ergonomic injury and chemotherapy-induced neuropathy. While local injection of TTX (0.03-1 μg) into the gastrocnemius muscle did not affect the mechanical nociceptive threshold in naïve rats, exposure to the inflammogen carrageenan produced a marked muscle mechanical hyperalgesia, which was dose-dependently inhibited by TTX. This antihyperalgesic effect was still significant at 24h. TTX also displayed a robust antinociceptive effect on eccentric exercise-induced mechanical hyperalgesia in the gastrocnemius muscle, a model of ergonomic pain. Finally, TTX produced a small but significant inhibition of neuropathic muscle pain induced by systemic administration of the cancer chemotherapeutic agent oxaliplatin. These results indicate that TTX-sensitive sodium currents in nociceptors play a central role in diverse states of skeletal muscle nociceptive sensitization, supporting the suggestion that therapeutic interventions based on TTX may prove useful in the treatment of muscle pain.
Collapse
Affiliation(s)
- P Alvarez
- Department of Oral and Maxillofacial Surgery, University of California San Francisco, San Francisco, CA, USA
| | - J D Levine
- Department of Oral and Maxillofacial Surgery, University of California San Francisco, San Francisco, CA, USA; Department of Medicine, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
18
|
|
19
|
Riluzole as a neuroprotective drug for spinal cord injury: from bench to bedside. Molecules 2015; 20:7775-89. [PMID: 25939067 PMCID: PMC6272473 DOI: 10.3390/molecules20057775] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Revised: 04/16/2015] [Accepted: 04/24/2015] [Indexed: 12/20/2022] Open
Abstract
Spinal cord injury (SCI) is a devastating event resulting in permanent loss of neurological function. To date, effective therapies for SCI have not been established. With recent progress in neurobiology, however, there is hope that drug administration could improve outcomes after SCI. Riluzole is a benzothiazole anticonvulsant with neuroprotective effects. It has been approved by the U.S. Food and Drug Administration as a safe and well-tolerated treatment for patients with amyotrophic lateral sclerosis. The mechanism of action of riluzole involves the inhibition of pathologic glutamatergic transmission in synapses of neurons via sodium channel blockade. There is convincing evidence that riluzole diminishes neurological tissue destruction and promotes functional recovery in animal SCI models. Based on these results, a phase I/IIa clinical trial with riluzole was conducted for patients with SCI between 2010 and 2011. This trial demonstrated significant improvement in neurological outcomes and showed it to be a safe drug with no serious adverse effects. Currently, an international, multi-center clinical trial (Riluzole in Acute Spinal Cord Injury Study: RISCIS) in phase II/III is in progress with riluzole for patients with SCI (clinicaltrials.gov, registration number NCT01597518). This article reviews the pharmacology and neuroprotective mechanisms of riluzole, and focuses on existing preclinical evidence, and emerging clinical data in the treatment of SCI.
Collapse
|
20
|
Nagoshi N, Fehlings MG. Investigational drugs for the treatment of spinal cord injury: review of preclinical studies and evaluation of clinical trials from Phase I to II. Expert Opin Investig Drugs 2015; 24:645-58. [PMID: 25645889 DOI: 10.1517/13543784.2015.1009629] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Efforts in basic research have clarified mechanisms involved in spinal cord injury (SCI), and resulted in positive findings using experimental treatments including cell transplantation and drug administration preclinically. Based on accumulated results, various clinical trials have begun for human SCI. AREAS COVERED In this review, the authors focus on five investigational drugs: riluzole, minocycline, Rho protein antagonist, magnesium chloride in polyethylene glycol formulation, and basic fibroblast growth factor. All drugs have established safety and tolerability from Phase I clinical trials, and are now in Phase II. They have been proven to have neuroprotective and/or neuroregenerative effects in animal models of SCI. EXPERT OPINION To date, diverse drugs have been translated into clinical trials, but none have reached clinical application. A key gap was the lack of reliable biomarkers for SCI to fast-track Phase I/II trials. Furthermore, problems were often due to lack of adequate outcome assessments for both animal models and SCI patients. In order to advance clinical trials more quickly and with greater success, more clinically relevant animal models should be used in basic research. Clinically, it is indispensable to use appropriate outcome measurements and to construct a wide network among clinical centers to validate the efficacy of drugs.
Collapse
Affiliation(s)
- Narihito Nagoshi
- University Health Network, Toronto Western Hospital, Krembil Neuroscience Center , Toronto, ON M5T 2S8 , Canada +1 416 603 5229 ; +1 416 603 6274 ;
| | | |
Collapse
|
21
|
Wu W, Wang P, Cheng JX, Xu XM. Assessment of white matter loss using bond-selective photoacoustic imaging in a rat model of contusive spinal cord injury. J Neurotrauma 2014; 31:1998-2002. [PMID: 24850066 PMCID: PMC4245875 DOI: 10.1089/neu.2014.3349] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
White matter (WM) loss is a critical event after spinal cord injury (SCI). Conventionally, such loss has been measured with histological and histochemical approaches, although the procedures are complex and may cause artifact. Recently, coherent Raman microscopy has been proven to be an emerging technology to study de- and remyelination of the injured spinal cord; however, limited penetration depth and small imaging field prevent it from comprehensive assessments of large areas of damaged tissues. Here, we report the use of bond-selective photoacoustic (PA) imaging with 1730-nm excitation, where the first overtone vibration of CH2 bond is located, to assess WM loss after a contusive SCI in adult rats. By employing the first overtone vibration of CH2 bond as the contrast, the mapping of the WM in an intact spinal cord was achieved in a label-free three-dimensional manner, and the physiological change of the spinal cord before and after injury was observed. Moreover, the recovery of the spinal cord from contusive injury with the treatment of a neuroprotective nanomedicine ferulic-acid-conjugated glycol chitosan (FA-GC) was also observed. Our study suggests that bond-selective PA imaging is a valuable tool to assess the progression of WM pathology after SCI as well as neuroprotective therapeutics in a label-free manner.
Collapse
Affiliation(s)
- Wei Wu
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Goldman and Campbell Brain and Spine, Indiana University School of Medicine, Indianapolis, Indiana
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana
| | - Pu Wang
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana
| | - Ji-Xin Cheng
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana
| | - Xiao-Ming Xu
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Goldman and Campbell Brain and Spine, Indiana University School of Medicine, Indianapolis, Indiana
| |
Collapse
|
22
|
Jung GY, Lee JY, Rhim H, Oh TH, Yune TY. An increase in voltage-gated sodium channel current elicits microglial activation followed inflammatory responsesin vitroandin vivoafter spinal cord injury. Glia 2013; 61:1807-21. [DOI: 10.1002/glia.22559] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2013] [Revised: 07/03/2013] [Accepted: 07/12/2013] [Indexed: 12/19/2022]
Affiliation(s)
- Gil Y. Jung
- Age-Related and Brain Diseases Research Center, School of Medicine; Kyung Hee University; Seoul Korea
| | - Jee Y. Lee
- Age-Related and Brain Diseases Research Center, School of Medicine; Kyung Hee University; Seoul Korea
- Neurodegeneration Control Research Center, School of Medicine; Kyung Hee University; Seoul Korea
| | - Hyewhon Rhim
- Center for Neuroscience, Korea Institute of Science & Technology; Seoul Korea
| | - Tae H. Oh
- Age-Related and Brain Diseases Research Center, School of Medicine; Kyung Hee University; Seoul Korea
| | - Tae Y. Yune
- Age-Related and Brain Diseases Research Center, School of Medicine; Kyung Hee University; Seoul Korea
- Neurodegeneration Control Research Center, School of Medicine; Kyung Hee University; Seoul Korea
- Department of Biochemistry and Molecular Biology, School of Medicine; Kyung Hee University; Seoul Korea
| |
Collapse
|
23
|
Fehlings MG, Wilson JR, Frankowski RF, Toups EG, Aarabi B, Harrop JS, Shaffrey CI, Harkema SJ, Guest JD, Tator CH, Burau KD, Johnson MW, Grossman RG. Riluzole for the treatment of acute traumatic spinal cord injury: rationale for and design of the NACTN Phase I clinical trial. J Neurosurg Spine 2013; 17:151-6. [PMID: 22985381 DOI: 10.3171/2012.4.aospine1259] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
In the immediate period after traumatic spinal cord injury (SCI) a variety of secondary injury mechanisms combine to gradually expand the initial lesion size, potentially leading to diminished neurological outcomes at long-term follow-up. Riluzole, a benzothiazole drug, which has neuroprotective properties based on sodium channel blockade and mitigation of glutamatergic toxicity, is currently an approved drug that attenuates the extent of neuronal degeneration in patients with amyotrophic lateral sclerosis. Moreover, several preclinical SCI studies have associated riluzole administration with improved functional outcomes and increased neural tissue preservation. Based on these findings, riluzole has attracted considerable interest as a potential neuroprotective drug for the treatment of SCI. Currently, a Phase I trial evaluating the safety and pharmacokinetic profile of riluzole in human SCI patients is being conducted by the North American Clinical Trials Network (NACTN) for Treatment of Spinal Cord Injury. The current review summarizes the existing preclinical and clinical literature on riluzole, provides a detailed description of the Phase I trial, and suggests potential opportunities for future investigation. Clinical trial registration no.: NCT00876889.
Collapse
Affiliation(s)
- Michael G Fehlings
- Department of Surgery, Division of Neurosurgery and Spinal Program, University of Toronto, Toronto Western Hospital, Toronto, Ontario, Canada.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Wilson JR, Fehlings MG. Riluzole for acute traumatic spinal cord injury: a promising neuroprotective treatment strategy. World Neurosurg 2013; 81:825-9. [PMID: 23295632 DOI: 10.1016/j.wneu.2013.01.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2012] [Accepted: 01/02/2013] [Indexed: 10/27/2022]
Abstract
BACKGROUND Over the years, understanding of the specific secondary injury mechanisms that follow traumatic spinal cord injury (SCI) has improved. These pathologic mechanisms collectively serve to increase the extent of neural tissue injury, reducing prospects for neurologic recovery. An enhanced understanding of the pathobiology of SCI has permitted investigation of therapies targeting specific elements of this pathologic cascade. It is now known that the continuous posttraumatic activation of neuronal voltage-gated sodium ion channels leads to increased rates of cell death through the development of cellular swelling, acidosis, and glutaminergic excitotoxicity. The objective herein is to provide an update regarding the current status of the potential neuroprotective drug riluzole in the treatment of traumatic SCI. METHODS Narrative review and summary paper. RESULTS Riluzole is a sodium channel-blocking benzothiazole anticonvulsant drug that is approved by the U.S. Food and Drug Administration for the treatment of amyotrophic lateral sclerosis and has shown efficacy in preclinical models of SCI in reducing the extent of sodium and glutamate mediated secondary injury. This drug is currently under early stages of clinical investigation in SCI and shows promise as an acute neuroprotective therapy in this context. CONCLUSION This article reviews the biologic rationale, existing preclinical evidence, and emerging clinical data for riluzole in the treatment of traumatic SCI.
Collapse
Affiliation(s)
- Jefferson R Wilson
- Department of Surgery, Division of Neurosurgery and Spinal Program, University of Toronto, Toronto Western Hospital, Toronto, Ontario, Canada
| | - Michael G Fehlings
- Department of Surgery, Division of Neurosurgery and Spinal Program, University of Toronto, Toronto Western Hospital, Toronto, Ontario, Canada.
| |
Collapse
|
25
|
Gensel JC, Tovar CA, Bresnahan JC, Beattie MS. Topiramate treatment is neuroprotective and reduces oligodendrocyte loss after cervical spinal cord injury. PLoS One 2012; 7:e33519. [PMID: 22428066 PMCID: PMC3302770 DOI: 10.1371/journal.pone.0033519] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2011] [Accepted: 02/15/2012] [Indexed: 11/21/2022] Open
Abstract
Excess glutamate release and associated neurotoxicity contributes to cell death after spinal cord injury (SCI). Indeed, delayed administration of glutamate receptor antagonists after SCI in rodents improves tissue sparing and functional recovery. Despite their therapeutic potential, most glutamate receptor antagonists have detrimental side effects and have largely failed clinical trials. Topiramate is an AMPA-specific, glutamate receptor antagonists that is FDA-approved to treat CNS disorders. In the current study we tested whether topiramate treatment is neuroprotective after cervical contusion injury in rats. We report that topiramate, delivered 15-minutes after SCI, increases tissue sparing and preserves oligodendrocytes and neurons when compared to vehicle treatment. In addition, topiramate is more effective than the AMPA-receptor antagonist, NBQX. To the best of our knowledge, this is the first report documenting a neuroprotective effect of topiramate treatment after spinal cord injury.
Collapse
Affiliation(s)
- John C Gensel
- Center for Brain and Spinal Cord Repair, Department of Neuroscience, The Ohio State University College of Medicine, Columbus, Ohio, United States of America.
| | | | | | | |
Collapse
|
26
|
Valproate reduces CHOP levels and preserves oligodendrocytes and axons after spinal cord injury. Neuroscience 2011; 178:33-44. [DOI: 10.1016/j.neuroscience.2011.01.012] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2010] [Revised: 01/05/2011] [Accepted: 01/06/2011] [Indexed: 01/12/2023]
|
27
|
Gensel JC, Donnelly DJ, Popovich PG. Spinal cord injury therapies in humans: an overview of current clinical trials and their potential effects on intrinsic CNS macrophages. Expert Opin Ther Targets 2011; 15:505-18. [PMID: 21281256 DOI: 10.1517/14728222.2011.553605] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Macrophage activation is a hallmark of spinal cord injury (SCI) pathology. CNS macrophages, derived from resident microglia and blood monocytes, are ubiquitous throughout the injured spinal cord, and respond to signals in the lesion environment by changing their phenotype and function. Depending on their phenotype and activation status, macrophages may initiate secondary injury mechanisms and/or promote CNS regeneration and repair. AREAS COVERED This review provides a comprehensive overview of current SCI clinical trials that are intended to promote neuroprotection, axon regeneration or cell replacement. None of these potential therapies were developed with the goal of influencing macrophage function; however, it is likely that each will have direct or indirect effects on CNS macrophages. The potential impact of each trial is discussed in the context of CNS macrophage biology. EXPERT OPINION Activation of CNS macrophages is an inevitable consequence of traumatic SCI. Given that these cells are exquisitely sensitive to changes in microenvironment, any intervention that affects tissue integrity and/or the composition of the cellular milieu will undoubtedly affect CNS macrophages. Thus, it is important to understand how current clinical trials will affect intrinsic CNS macrophages.
Collapse
Affiliation(s)
- John C Gensel
- The Ohio State University College of Medicine, Center for Brain and Spinal Cord Repair, 795 Biomedical Research Tower, 460 West 12th Avenue, Columbus, OH 43210-1239, USA
| | | | | |
Collapse
|
28
|
Hunanyan AS, Alessi V, Patel S, Pearse DD, Matthews G, Arvanian VL. Alterations of action potentials and the localization of Nav1.6 sodium channels in spared axons after hemisection injury of the spinal cord in adult rats. J Neurophysiol 2010; 105:1033-44. [PMID: 21177993 DOI: 10.1152/jn.00810.2010] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Previously, we reported a pronounced reduction in transmission through surviving axons contralateral to chronic hemisection (HX) of adult rat spinal cord. To examine the cellular and molecular mechanisms responsible for this diminished transmission, we recorded intracellularly from lumbar lateral white matter axons in deeply anesthetized adult rats in vivo and measured the propagation of action potentials (APs) through rubrospinal/reticulospinal tract (RST/RtST) axons contralateral to chronic HX at T10. We found decreased excitability in these axons, manifested by an increased rheobase to trigger APs and longer latency for AP propagation passing the injury level, without significant differences in axonal resting membrane potential and input resistance. These electrophysiological changes were associated with altered spatial localization of Nav1.6 sodium channels along axons: a subset of axons contralateral to the injury exhibited a diffuse localization (>10 μm spread) of Nav1.6 channels, a pattern characteristic of demyelinated axons (Craner MJ, Newcombe J, Black JA, Hartle C, Cuzner ML, Waxman SG. Proc Natl Acad Sci USA 101: 8168-8173, 2004b). This result was substantiated by ultrastructural changes seen with electron microscopy, in which an increased number of large-caliber, demyelinated RST axons were found contralateral to the chronic HX. Therefore, an increased rheobase, pathological changes in the distribution of Nav1.6 sodium channels, and the demyelination of contralateral RST axons are likely responsible for their decreased conduction chronically after HX and thus may provide novel targets for strategies to improve function following incomplete spinal cord injury.
Collapse
Affiliation(s)
- Arsen S Hunanyan
- Northport Veterans Affairs Medical Center, 79 Middleville Road, Bld. 62, Northport, NY 11768, USA
| | | | | | | | | | | |
Collapse
|
29
|
Mechanisms of Axonal Spheroid Formation in Central Nervous System Wallerian Degeneration. J Neuropathol Exp Neurol 2010; 69:455-72. [DOI: 10.1097/nen.0b013e3181da84db] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
30
|
Yu CG, Yezierski RP, Joshi A, Raza K, Li Y, Geddes JW. Involvement of ERK2 in traumatic spinal cord injury. J Neurochem 2010; 113:131-42. [PMID: 20067580 DOI: 10.1111/j.1471-4159.2010.06579.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Activation of extracellular signal-regulated protein kinase 1/2 (ERK1/2) are implicated in the pathophysiology of spinal cord injury (SCI). However, the specific functions of individual ERK isoforms in neurodegeneration are largely unknown. We investigated the hypothesis that ERK2 activation may contribute to pathological and functional deficits following SCI and that ERK2 knockdown using RNA interference may provide a novel therapeutic strategy for SCI. Lentiviral ERK2 shRNA and siRNA were utilized to knockdown ERK2 expression in the spinal cord following SCI. Pre-injury intrathecal administration of ERK2 siRNA significantly reduced excitotoxic injury-induced activation of ERK2 (p < 0.001) and caspase 3 (p < 0.01) in spinal cord. Intraspinal administration of lentiviral ERK2 shRNA significantly reduced ERK2 expression in the spinal cord (p < 0.05), but did not alter ERK1 expression. Administration of the lentiviral ERK2 shRNA vector 1 week prior to severe spinal cord contusion injury resulted in a significant improvement in locomotor function (p < 0.05), total tissue sparing (p < 0.05), white matter sparing (p < 0.05), and gray matter sparing (p < 0.05) 6 weeks following severe contusive SCI. Our results suggest that ERK2 signaling is a novel target associated with the deleterious consequences of spinal injury.
Collapse
Affiliation(s)
- Chen-Guang Yu
- Spinal Cord and Brain Injury Research Center and Department of Anatomy and Neurobiology, University of Kentucky College of Medicine, Lexington, Kentucky 40536-0509, USA.
| | | | | | | | | | | |
Collapse
|
31
|
Wang JA, Lin W, Morris T, Banderali U, Juranka PF, Morris CE. Membrane trauma and Na+ leak from Nav1.6 channels. Am J Physiol Cell Physiol 2009; 297:C823-34. [PMID: 19657055 DOI: 10.1152/ajpcell.00505.2008] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
During brain trauma, white matter experiences shear and stretch forces that, without severing axons, nevertheless trigger their secondary degeneration. In central nervous system (CNS) trauma models, voltage-gated sodium channel (Nav) blockers are neuroprotective. This, plus the rapid tetrodotoxin-sensitive Ca2+ overload of stretch-traumatized axons, points to "leaky" Nav channels as a pivotal early lesion in brain trauma. Direct effects of mechanical trauma on neuronal Nav channels have not, however, been tested. Here, we monitor immediate responses of recombinant neuronal Nav channels to stretch, using patch-clamp and Na+-dye approaches. Trauma constituted either bleb-inducing aspiration of cell-attached oocyte patches or abrupt uniaxial stretch of cells on an extensible substrate. Nav1.6 channel transient current displayed irreversible hyperpolarizing shifts of steady-state inactivation [availability(V)] and of activation [g(V)] and, thus, of window current. Left shift increased progressively with trauma intensity. For moderately intense patch trauma, a approximately 20-mV hyperpolarizing shift was registered. Nav1.6 voltage sensors evidently see lower energy barriers posttrauma, probably because of the different bilayer mechanics of blebbed versus intact membrane. Na+ dye-loaded human embryonic kidney (HEK) cells stably transfected with alphaNav1.6 were subjected to traumatic brain injury-like stretch. Cytoplasmic Na+ levels abruptly increased and the trauma-induced influx had a significant tetrodotoxin-sensitive component. Nav1.6 channel responses to cell and membrane trauma are therefore consistent with the hypothesis that mechanically induced Nav channel leak is a primary lesion in traumatic brain injury. Nav1.6 is the CNS node of Ranvier Nav isoform. When, during head trauma, nodes experienced bleb-inducing membrane damage of varying intensities, nodal Nav1.6 channels should immediately "leak" over a broadly left-smeared window current range.
Collapse
Affiliation(s)
- Jun A Wang
- Neuroscience, Ottawa Hospital Research Institute, The Ottawa Hospital, Ottawa, Ontario, Canada
| | | | | | | | | | | |
Collapse
|
32
|
Yu D, Neeley WL, Pritchard CD, Slotkin JR, Woodard EJ, Langer R, Teng YD. Blockade of peroxynitrite-induced neural stem cell death in the acutely injured spinal cord by drug-releasing polymer. Stem Cells 2009; 27:1212-22. [PMID: 19418456 DOI: 10.1002/stem.26] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Therapeutic impact of neural stem cells (NSCs) for acute spinal cord injury (SCI) has been limited by the rapid loss of donor cells. Neuroinflammation is likely the cause. As there are close temporal-spatial correlations between the inducible nitric oxide (NO) synthase expression and the donor NSC death after neurotrauma, we reasoned that NO-associated radical species might be the inflammatory effectors which eliminate NSC grafts and kill host neurons. To test this hypothesis, human NSCs (hNSCs: 5 x 10(4) to 2 x 10(6) per milliliter) were treated in vitro with "plain" medium, 20 microM glutamate, or donors of NO and peroxynitrite (ONOO(-); 100 and 400 microM of spermine or DETA NONOate, and SIN-1, respectively). hNSC apoptosis primarily resulted from SIN-1 treatment, showing ONOO(-)-triggered protein nitration and the activation of p38 MAPK, cytochrome c release, and caspases. Therefore, cell death following post-SCI (p.i.) NO surge may be mediated through conversion of NO into ONOO(-). We subsequently examined such causal relationship in a rat model of dual penetrating SCI using a retrievable design of poly-lactic-co-glycolic acid (PLGA) scaffold seeded with hNSCs that was shielded by drug-releasing polymer. Besides confirming the ONOO(-)-induced cell death signaling, we demonstrated that cotransplantation of PLGA film embedded with ONOO(-) scavenger, manganese (III) tetrakis (4-benzoic acid) porphyrin, or uric acid (1 micromol per film), markedly protected hNSCs 24 hours p.i. (total: n = 10). Our findings may provide a bioengineering approach for investigating mechanisms underlying the host microenvironment and donor NSC interaction and help formulate strategies for enhancing graft and host cell survival after SCI.
Collapse
Affiliation(s)
- Dou Yu
- Department of Neurosurgery, Harvard Medical School, The Brigham and Women's Hospital and Children's Hospital Boston, Boston, MA 02115, USA
| | | | | | | | | | | | | |
Collapse
|
33
|
Systemic polyethylene glycol promotes neurological recovery and tissue sparing in rats after cervical spinal cord injury. J Neuropathol Exp Neurol 2009; 68:661-76. [PMID: 19458542 DOI: 10.1097/nen.0b013e3181a72605] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Polyethylene glycol (PEG) has been reported to possess fusogenic properties that may confer neuroprotection after spinal cord injury (SCI), but there is uncertainty regarding the mechanisms of PEG in vivo and the robustness of its protective effects. We hypothesized that PEG promotes preservation of cytoskeletal proteins associated with white matter protection and neurobehavioral recovery after SCI. In proof-of-principle experiments using a pin-drop organotypic culture model of SCI, PEG attenuated neural cell death. Adult rats underwent 35-g clip compression SCI at C8 and were randomized postinjury to receive intravenous 30% PEG or sterile Ringer's lactate solution. Confocal microscopy and high-performance liquid chromatography of fluorescein-conjugated PEG permitted in vivo quantification of PEG concentrations in the injured and uninjured spinal cord. Western blot, immunohistochemistry, and terminal deoxynucleotidyl transferase mediated dUTP nick end labeling staining demonstrated that PEG reduced 200-kd neurofilament degradation and apoptotic cell death. Polyethylene glycol also promoted spinal cord tissue sparing based on retrograde axonal Fluoro-Gold tracing and morphometric histological assessment. Polyethylene glycol also promoted significant, although modest, neurobehavioral recovery after SCI. Collectively, these results indicate that PEG protects key axonal cytoskeletal proteins after SCI, and that the protection is associated with axonal preservation. The modest extent of locomotor recovery after treatment with PEG suggests, however, that this compound may notconfer sufficient neuroprotection to be used clinically as a single treatment.
Collapse
|
34
|
Yu CG, Joshi A, Geddes JW. Intraspinal MDL28170 microinjection improves functional and pathological outcome following spinal cord injury. J Neurotrauma 2008; 25:833-40. [PMID: 18627259 DOI: 10.1089/neu.2007.0490] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Although calpain (calcium-activated cysteine protease) inhibition represents a rational therapeutic target for spinal cord injury (SCI), few studies have reported improved functional outcomes with post-injury administration of calpain inhibitors. This reflects the weak potency and limited aqueous solubility of current calpain inhibitors. Previously, we demonstrated that intraspinal microinjection of the calpain inhibitor MDL28170 resulted in greater inhibition of calpain activity as compared to systemic administration of the same compound. In the present study, we evaluated the ability of intraspinal MDL28170 microinjection to spare spinal tissue and locomotor dysfunction following SCI. Contusion SCI was produced in female Long-Evans rats using the Infinite Horizon impactor at the 200-kdyn force setting. Open-field locomotion was evaluated until 6 weeks post-injury. Histological assessment of tissue sparing was performed at 6 weeks after SCI. The results demonstrate that MDL28170, administered with a single post-injury intraspinal microinjection (50 nmoles), significantly improves both locomotor function and pathological outcome measures following SCI.
Collapse
Affiliation(s)
- Chen-Guang Yu
- Spinal Cord and Brain Injury Research Center, Department of Anatomy and Neurobiology, University of Kentucky, Lexington, Kentucky, USA
| | | | | |
Collapse
|
35
|
Neural and anatomical abnormalities of the gastrointestinal system resulting from contusion spinal cord injury. Neuroscience 2008; 154:1627-38. [DOI: 10.1016/j.neuroscience.2008.04.071] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2008] [Revised: 04/15/2008] [Accepted: 04/25/2008] [Indexed: 11/21/2022]
|
36
|
Ouyang H, Galle B, Li J, Nauman E, Shi R. Biomechanics of spinal cord injury: a multimodal investigation using ex vivo guinea pig spinal cord white matter. J Neurotrauma 2008; 25:19-29. [PMID: 18355155 DOI: 10.1089/neu.2007.0340] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The primary injury phase of traumatic spinal cord injury (SCI) was investigated using a novel compression injury model. Ventral white matter from adult guinea pigs was crushed to 25%, 50%, 70%, and 90% ex vivo. During compression, the physical deformation, applied force and the compound action potentials (CAP) were simultaneously recorded. In addition, axonal membrane continuity was analyzed with a horseradish peroxidase (HRP) exclusion assay. Experimental results showed both a CAP decrease and increased HRP uptake as a function of increased compression. The percent CAP reduction was also consistent to the percent HRP uptake, which implies that either metric could be used to assess acute axon damage. Analysis of the HRP stained axon distribution demonstrated a gradient of damage, with the highest levels of staining near the gray matter. The patterns of axon damage revealed by histology also coincided with higher levels of von Mises stress, which were predicted with a recently developed finite element model of ventral white matter. Numerical values obtained from the finite element model suggest stress magnitudes near 2 kPa are required to initiate anatomical tissue injury. We believe that data from this study could further elucidate the deformation-function relationship in acute spinal cord injury.
Collapse
Affiliation(s)
- Hui Ouyang
- Department of Medical Sciences, School of Veterinary Medicine, West Lafayette, Indiana, USA
| | | | | | | | | |
Collapse
|
37
|
Black JA, Waxman SG. Phenytoin protects central axons in experimental autoimmune encephalomyelitis. J Neurol Sci 2008; 274:57-63. [PMID: 18485368 DOI: 10.1016/j.jns.2008.04.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2008] [Revised: 03/20/2008] [Accepted: 04/02/2008] [Indexed: 10/22/2022]
Abstract
Axon degeneration is a major contributor to non-remitting deficits in multiple sclerosis (MS). Thus the development of therapies to provide protection of axons has elicited considerable interest. Voltage-gated sodium channels have been implicated in the injury cascade leading to axonal damage, and sodium-channel blockers have shown efficacy in ameliorating axonal damage in disease models following anoxia, trauma and damaging levels of nitric oxide (NO). Here we discuss studies in our laboratory that examined the protective effects of phenytoin, a well-characterized sodium-channel blocker, in the inflammatory/demyelinating disorder experimental autoimmune encephalomyelitis (EAE), a model of MS. Administration of phenytoin to C57/Bl6 mice inoculated with rat myelin oligodendrocyte glycoprotein (MOG) provides improved clinical status, preservation of axons, enhanced action potential conduction and reduced immune infiltrates compared to untreated mice with EAE. Moreover, continuous treatment with phenytoin provides these protective actions for at least 180 days post-MOG injection. The withdrawal of phenytoin from mice inoculated with MOG, however, is accompanied by acute exacerbation of EAE, with significant mortality and infiltration of immune cells in the CNS. Our studies demonstrate the efficacy of phenytoin as a neuroprotectant in EAE. Our results also, however, indicate that we need to learn more about the long-term effects of sodium-channel blockers, and of their withdrawal, in neuroinflammatory disorders.
Collapse
Affiliation(s)
- Joel A Black
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale School of Medicine, New Haven, CT 06510, United States.
| | | |
Collapse
|
38
|
Xu GY, Liu S, Hughes MG, McAdoo DJ. Glutamate-induced losses of oligodendrocytes and neurons and activation of caspase-3 in the rat spinal cord. Neuroscience 2008; 153:1034-47. [PMID: 18423997 DOI: 10.1016/j.neuroscience.2008.02.065] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2007] [Revised: 02/05/2008] [Accepted: 02/15/2008] [Indexed: 11/20/2022]
Abstract
The toxicity of released glutamate contributes substantially to secondary cell death following spinal cord injury (SCI). In this work, the extent and time courses of glutamate-induced losses of neurons and oligodendrocytes are established. Glutamate was administered into the spinal cords of anesthetized rats at approximately the concentration and duration of its release following SCI. Cells in normal tissue, in tissue exposed to artificial cerebrospinal fluid and in tissue exposed to glutamate were counted on a confocal system in control animals and from 6 h to 28 days after treatment to assess cell losses. Oligodendrocytes were identified by staining with antibody CC-1 and neurons by immunostaining for Neuronal Nuclei (NeuN) or Neurofilament H. The density of oligodendrocytes declined precipitously in the first 6 h after exposure to glutamate, and then relatively little from 24 h to 28 days post-exposure. Similarly, neuron densities first declined rapidly, but at a decreasing rate, from 0 h to 72 h post-glutamate exposure and did not change significantly from 72 h to 28 days thereafter. The nuclei of many cells strongly and specifically stained for activated caspase-3, an indicator of apoptosis, in response to exposure to glutamate. Caspase-3 was localized to the nucleus and may participate in apoptotic cell death. However, persistence of caspase-3 staining for at least a week after exposure to glutamate during little to no loss of oligodendrocytes and neurons demonstrates that elevation of caspase-3 does not necessarily lead to rapid cell death. Beyond about 48 h after exposure to glutamate, locomotor function began to recover while cell numbers stabilized or declined slowly, demonstrating that functional recovery in the experiments presented involves processes other than replacement of oligodendrocytes and/or neurons.
Collapse
Affiliation(s)
- G-Y Xu
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555-1043, USA
| | | | | | | |
Collapse
|
39
|
|
40
|
Yu CG, Geddes JW. Sustained calpain inhibition improves locomotor function and tissue sparing following contusive spinal cord injury. Neurochem Res 2007; 32:2046-53. [PMID: 17476592 DOI: 10.1007/s11064-007-9347-4] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2007] [Accepted: 04/03/2007] [Indexed: 10/23/2022]
Abstract
Following contusive spinal cord injury (SCI), calpain activity is dramatically increased and remains elevated for days to weeks. Although calpain inhibition has previously been demonstrated to be neuroprotective following spinal cord injury, most studies administered the calpain inhibitor at a single time point. We hypothesized that sustained calpain inhibition would improve functional and pathological outcomes, as compared to the results obtained with a single postinjury administration of the calpain inhibitor. Contusion SCI was produced in female Long-Evans rats using the Infinite Horizon spinal cord injury impactor at the 200 kdyn force setting. Open-field locomotor function was evaluated until 6 weeks postinjury. Histological assessment of lesion volume and tissue sparing was performed at 6 weeks after SCI. Calpain inhibitor MDL28170 administered as a single postinjury i.v. bolus (20 mg/kg) or as a daily i.p. dose (1 mg/kg) improved locomotor function, but did not increase tissue sparing. Combined i.v. and daily i.p. MDL28170 administration resulted in significant improvement in both functional and pathological outcome measures, supporting the calpain theory of SCI proposed by Dr. Banik and colleagues.
Collapse
Affiliation(s)
- Chen-Guang Yu
- Spinal Cord and Brain Injury Research Center and Department of Anatomy and Neurobiology, University of Kentucky, Lexington, KY 40536-0509, USA
| | | |
Collapse
|
41
|
Kang SK, Yeo JE, Kang KS, Phinney DG. Cytoplasmic extracts from adipose tissue stromal cells alleviates secondary damage by modulating apoptosis and promotes functional recovery following spinal cord injury. Brain Pathol 2007; 17:263-75. [PMID: 17465991 PMCID: PMC8095508 DOI: 10.1111/j.1750-3639.2007.00070.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Spinal cord injury (SCI) typically results from sustained trauma to the spinal cord, resulting in loss of neurologic function at the level of the injury. However, activation of various physiological mechanisms secondary to the initial trauma including edema, inflammation, excito-toxicity, excessive cytokine release and apoptosis may exacerbate the injury and/or retard natural repair mechanisms. Herein, we demonstrate that cytoplasmic extracts prepared from adipose tissue stromal cells (ATSCs) inhibits H(2)O(2)-mediated apoptosis of cultured spinal cord-derived neural progenitor cells (NPCs) resulting in increased cell survival. The ATSC extracts mediated this effect by decreasing caspase-3 and c-Jun-NH2-terminal kinase (SAPK/JNK) activity, inhibiting cytochrome c release from mitochondria and reducing Bax expression levels in cells. Direct injection of ATSC extracts mixed with Matrigel into the spinal cord immediately after SCI also resulted in reduced apoptotic cell death, astrogliosis and hypo-myelination but did not reduce the extent of microglia infiltration. Moreover, animals injected with the ATSC extract showed significant functional improvement of hind limbs as measured by the BBB (Basso, Beattie and Bresnahan) scale. Collectively, these studies show a prominent therapeutic effect of ATSC cytoplasmic extracts on SCI principally caused by an inhibition of apoptosis-mediated cell death, which spares white matter, oligodendrocytes and neurons at the site of injury. The ability of ATSC extracts to prevent secondary pathological events and improve neurologic function after SCI suggests that extracts prepared from autologous cells harvested from SCI patients may have clinical utility.
Collapse
Affiliation(s)
- Soo Kyung Kang
- Department of Physiology, College of Medicine, Pusan National University, 1-10 Ami-Dong, Busan 602-739, South Korea.
| | | | | | | |
Collapse
|
42
|
|
43
|
Rooney BA, Crown ED, Hulsebosch CE, McAdoo DJ. Preemptive analgesia with lidocaine prevents Failed Back Surgery Syndrome. Exp Neurol 2006; 204:589-96. [PMID: 17261281 DOI: 10.1016/j.expneurol.2006.12.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2006] [Revised: 12/01/2006] [Accepted: 12/12/2006] [Indexed: 11/17/2022]
Abstract
Failed Back Surgery Syndrome (FBSS) is commonly encountered in pain-treatment settings in the United States. We tested whether potential key factors in this syndrome, such as extracellular concentrations of excitatory amino acids (EAAs), are increased in the dorsal horn by synaptic release due to unintentional stretch and/or deformation/compression/transection of dorsal spinal structures during surgery. We hypothesized that pharmacological nerve block as a form of preemptive analgesia prior to any insult to dorsal root neurons will prevent an abnormally high increase in extracellular concentrations of EAAs in the dorsal horn and ultimately the establishment of central sensitization during back surgery. The L4 and L5 dorsal roots were cut bilaterally near the spinal cord to provide an adequate model to test for preemptive analgesia. Amino acid concentrations were measured by dorsal horn microdialysis sampling; EAAs aspartate and glutamate were significantly increased by 80% and 65% respectively, as were other amino acids compared to sham control values. Topical application of 1% Lidocaine, a voltage-gated Na(+) channel blocker, for 10 min prior to L4 and L5 bilateral dorsal rhizotomy (BDR) significantly attenuated the increase in EAA concentrations such that their values were not different from sham controls. Behavioral tests demonstrated significant hindlimb mechanical allodynia after BDRs that was significantly attenuated by Lidocaine pretreatment. Thus, Lidocaine pretreatment could offer a safe measure for prevention of chronic pain for back surgical procedures if given by intramuscular injection, topical administration onto spinal nerves and/or the dorsal spinal surface during surgical procedures that include nerve entrapment release, intervertebral disc modification and laminectomies.
Collapse
Affiliation(s)
- B A Rooney
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555-1043, USA.
| | | | | | | |
Collapse
|
44
|
Vitellaro-Zuccarello L, Mazzetti S, Madaschi L, Bosisio P, Gorio A, De Biasi S. Erythropoietin-mediated preservation of the white matter in rat spinal cord injury. Neuroscience 2006; 144:865-77. [PMID: 17141961 DOI: 10.1016/j.neuroscience.2006.10.023] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2006] [Revised: 10/12/2006] [Accepted: 10/12/2006] [Indexed: 12/21/2022]
Abstract
We investigated the effect of a single administration of recombinant human erythropoietin (rhEPO) on the preservation of the ventral white matter of rats at 4 weeks after contusive spinal cord injury (SCI), a time at which functional recovery is significantly improved in comparison to the controls [Gorio A, Necati Gokmen N, Erbayraktar S, Yilmaz O, Madaschi L, Cichetti C, Di Giulio AM, Enver Vardar E, Cerami A, Brines M (2002) Recombinant human erythropoietin counteracts secondary injury and markedly enhances neurological recovery from experimental spinal cord trauma. Proc Natl Acad Sci U S A 99:9450-9455; Gorio A, Madaschi L, Di Stefano B, Carelli S, Di Giulio AM, De Biasi S, Coleman T, Cerami A, Brines M (2005) Methylprednisolone neutralizes the beneficial effects of erythropoietin in experimental spinal cord injury. Proc Natl Acad Sci U S A 102:16379-16384]. Specifically, we examined, by morphological and cytochemical methods combined with light, confocal and electron microscopy, i) myelin preservation, ii) activation of adult oligodendrocyte progenitors (OPCs) identified for the expression of NG2 transmembrane proteoglycan, iii) changes in the amount of the chondroitin sulfate proteoglycans neurocan, versican and phosphacan and of their glycosaminoglycan component labeled with Wisteria floribunda lectin, and iv) ventral horn density of the serotonergic plexus as a marker of descending motor control axons. Injured rats received either saline or a single dose of rhEPO within 30 min after SCI. The results showed that the significant improvement of functional outcome observed in rhEPO-treated rats was associated with a better preservation of myelin in the ventral white matter. Moreover, the significant increase of both the number of NG2-positive OPCs and the labeling for Nogo-A, a marker of differentiated oligodendrocytes, suggested that rhEPO treatment could result in the generation of new myelinating oligodendrocytes. Sparing of fiber tracts in the ventral white matter was confirmed by the increased density of the serotonergic plexus around motor neurons. As for chondroitin sulfate proteoglycans, only phosphacan, increased in saline-treated rats, returned to normal levels in rhEPO group, probably reflecting a better maintenance of glial-axolemmal relationships along nerve fibers. In conclusion, this investigation expands previous studies supporting the pleiotropic neuroprotective effect of rhEPO on secondary degenerative response and its therapeutic potential for the treatment of SCI and confirms that the preservation of the ventral white matter, which contains descending motor pathways, may be critical for limiting functional deficit.
Collapse
Affiliation(s)
- L Vitellaro-Zuccarello
- Dipartimento di Scienze Biomolecolari e Biotecnologie, Università degli Studi di Milano, Via Celoria 26, I-20133 Milano, Italy.
| | | | | | | | | | | |
Collapse
|
45
|
Ouardouz M, Malek S, Coderre E, Stys PK. Complex interplay between glutamate receptors and intracellular Ca2+ stores during ischaemia in rat spinal cord white matter. J Physiol 2006; 577:191-204. [PMID: 16945971 PMCID: PMC2000677 DOI: 10.1113/jphysiol.2006.116798] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Electrophysiological recordings of propagated compound action potentials (CAPs) and axonal Ca(2+) measurements using confocal microscopy were used to study the interplay between AMPA receptors and intracellullar Ca(2+) stores in rat spinal dorsal columns subjected to in vitro combined oxygen and glucose deprivation (OGD). Removal of Ca(2+) or Na(+) from the perfusate was protective after 30 but not 60 min of OGD. TTX was ineffective with either exposure, consistent with its modest effect on ischaemic depolarization. In contrast, AMPA antagonists were very protective, even after 60 min of OGD where 0Ca(2+) + EGTA perfusate was ineffective. Similarly, blocking ryanodine receptor-mediated Ca(2+) mobilization from internal stores (0Ca(2+) + nimodipine or 0Ca(2+) + ryanodine), or inositol 1,4,5-trisphosphate (IP(3))-dependent Ca(2+) release (block of group 1 metabotropic glutamate receptors with 1-aminoindan-1,5-dicarboxylic acid, inhibition of phospholipase C with U73122 or IP(3) receptor block with 2APB; each in 0Ca(2+)) were each very protective, with the combination resulting in virtually complete functional recovery after 1 h OGD (97 +/- 32% CAP recovery versus 4 +/- 6% in artificial cerebrospinal fluid). AMPA induced a rise in Ca(2+) concentration in normoxic axons, which was greatly reduced by blocking ryanodine receptors. Our data therefore suggest a novel and surprisingly complex interplay between AMPA receptors and Ca(2+) mobilization from intracellular Ca(2+) stores. We propose that AMPA receptors may not only allow Ca(2+) influx from the extracellular space, but may also significantly influence Ca(2+) release from intra-axonal Ca(2+) stores. In dorsal column axons, AMPA receptor-dependent mechanisms appear to exert a greater influence than voltage-gated Na(+) channels on functional outcome following OGD.
Collapse
Affiliation(s)
- Mohamed Ouardouz
- Division of Neuroscience, Ottawa Health Research Institute, 725 Parkdale Avenue, Ottawa, Ontario, Canada K1Y 4E9
| | | | | | | |
Collapse
|
46
|
Kyung KS, Gon JH, Geun KY, Sup JJ, Suk WJ, Ho KJ. 6-Shogaol, a natural product, reduces cell death and restores motor function in rat spinal cord injury. Eur J Neurosci 2006; 24:1042-52. [PMID: 16930431 DOI: 10.1111/j.1460-9568.2006.04908.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Spinal cord injury (SCI) results in progressive waves of secondary injuries, which via the activation of a barrage of noxious pathological mechanisms exacerbate the injury to the spinal cord. Secondary injuries are associated with edema, inflammation, excitotoxicity, excessive cytokine release, caspase activation and cell apoptosis. This study was aimed at investigating the possible neuroprotective effects of 6-shogaol purified from Zingiber officinale by comparing an experimental SCI rat group with SCI control rats. Shogaol attenuated apoptotic cell death, including poly(ADP-ribose) polymerase activity, and reduced astrogliosis and hypomyelination which occurs in areas of active cell death in the spinal cords of SCI rats. The foremost protective effect of shogaol in SCI would therefore be manifested in the suppression of the acute secondary apoptotic cell death. However, it does not attenuate active microglia and macrophage infiltration. This finding is supported by a lack of histopathological changes in the areas of the lesion in the shogaol-treated SCI rats. Moreover, shogaol-mediated neuroprotection has been linked with shogaol's attenuation of p38 mitogen-activated protein kinase, p-SAPK/JNK and signal transducer, and with transcription-3 activation. Our results demonstrate that shogaol administrated immediately after SCI significantly diminishes functional deficits. The shogaol-treated group recovered hindlimb reflexes more rapidly and a higher percentage of these rats regained responses compared with the untreated injured rats. The overall hindlimb functional improvement of hindlimbs, as measured by the Basso, Beattie and Bresnahan scale, was significantly enhanced in the shogaol-treated group relative to the SCI control rats. Our data show that the therapeutic outcome of shogaol probably results from its comprehensive effects of blocking apoptotic cell death, resulting in the protection of white matter, oligodendrocytes and neurons, and inhibiting astrogliosis. Our finding that the administration of shogaol prevents secondary pathological events in traumatic SCIs and promotes recovery of motor functions in an animal model raises the issue of whether shogaol could be used therapeutically in humans after SCI.
Collapse
Affiliation(s)
- Kang Soo Kyung
- Department of Physiology, School of Medicine, Pusan National University, 1-10 Ami-Dong, Seo-Gu, Busan, South Korea.
| | | | | | | | | | | |
Collapse
|
47
|
Jeffery ND, Smith PM, Lakatos A, Ibanez C, Ito D, Franklin RJM. Clinical canine spinal cord injury provides an opportunity to examine the issues in translating laboratory techniques into practical therapy. Spinal Cord 2006; 44:584-93. [PMID: 16520817 DOI: 10.1038/sj.sc.3101912] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
STUDY DESIGN Review. OBJECTIVES To highlight the value of investigating the effects of putative therapeutic interventions in clinical spinal cord injury (SCI) in domestic dogs. SETTING England, UK. METHODS Many experimental interventions in laboratory rodents have been shown to ameliorate the functional deficits caused by SCI; the challenge now is to determine whether they can be translated into useful clinical techniques. Important differences between clinical SCI in human patients and that in laboratory rodents are in the size of the spinal cord and heterogeneity of injury severity. A further key issue is whether the statistical difference in outcome in the laboratory will translate into a useful difference in clinical outcome. Here, we stress the value of investigating the effects of putative therapies in clinical SCI in domestic dogs. The causes of injury, ability to categorise the severity and methods available to measure outcome are very similar between canine and human patients. Furthermore, postmortem tissue more rapidly becomes available from dogs because of their short lifespan than from human patients. RESULTS The role that investigation of canine SCI might play is illustrated by our preliminary trials on intraspinal transplantation of olfactory glial cells for severe SCI. CONCLUSIONS This canine translational model provides a means of 'filtering' putative treatments before human application.
Collapse
Affiliation(s)
- N D Jeffery
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | | | | | | | | | | |
Collapse
|
48
|
Wrathall JR, Emch GS. Effect of injury severity on lower urinary tract function after experimental spinal cord injury. PROGRESS IN BRAIN RESEARCH 2006; 152:117-34. [PMID: 16198697 DOI: 10.1016/s0079-6123(05)52008-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Lower urinary tract dysfunction is a serious burden for patients following spinal cord injury. Patients are usually limited to treatment with urinary drainage catheters, which can lead to repeated urinary tract infections and lower quality of life. Most of the information previously obtained regarding lower urinary tract function after spinal cord injury has been in completely transected animals. After thoracic transection in the rat, plasticity of local lumbosacral spinal circuitry establishes a "reflex bladder," which results in partial recovery of micturition, albeit with reduced voiding efficiency. Since at least half of cord-injured patients exhibit neurologically incomplete injury, rat models of clinically relevant incomplete contusion injury have been developed. With respect to lower urinary tract function, recent anatomical and physiological studies have been performed after incomplete thoracic contusion injury. The results show greater recovery of lower urinary tract function that varies inversely with the severity of the initial trauma and is positively correlated with time after injury. Recovery, as measured by coordination of the bladder with the external urethral sphincter, occurs between 1 and 4 weeks after spinal cord injury. It is associated with normalization of: serotonin immunoreactivity and glutamate receptor subunit mRNA expression in the dorsolateral nucleus that innervates the external urethral sphincter muscle, the response to glutamatergic pharmacological probes administered at the lumbosacral spinal cord level, and c-Fos activation patterns in the lumbar spinal cord. Understanding the mechanisms involved in this recovery will provide a basis for enhancing lower urinary tract function in patients after incomplete spinal cord injury.
Collapse
Affiliation(s)
- Jean R Wrathall
- Department of Neuroscience, Georgetown University Medical Center, TRB EP04, Washington, DC 20057, USA
| | | |
Collapse
|
49
|
McAdoo DJ, Hughes MG, Nie L, Shah B, Clifton C, Fullwood S, Hulsebosch CE. The effect of glutamate receptor blockers on glutamate release following spinal cord injury. Lack of evidence for an ongoing feedback cascade of damage --> glutamate release --> damage --> glutamate release --> etc. Brain Res 2005; 1038:92-9. [PMID: 15748877 DOI: 10.1016/j.brainres.2005.01.024] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2004] [Revised: 01/03/2005] [Accepted: 01/09/2005] [Indexed: 11/25/2022]
Abstract
It is widely hypothesized that excitotoxicity of released glutamate following a CNS insult is propagated by the cyclic cascade: glutamate release --> damage --> glutamate release --> further damage --> etc. We tested this hypothesis by determining the effects of attempting to interrupt the loop by administering glutamate receptor antagonists and Na(+)-channel blockers on glutamate release following spinal cord injury (SCI). The effects of administering the NMDA receptor blockers MK-801 and memantine, the AMPA/kainate receptor blockers NBQX and GYKI 52466, the AMPA receptor desensitization blocker cyclothiazide and the sodium channel blockers riluzole, mexiletine and QX-314 on post-SCI were determined. Agents were administered into the site of injury by direct injection, by microdialysis or systemically. None of these agents had an appreciable effect on glutamate release following SCI. Thus, it is unlikely that the above cascade produces significant secondary glutamate release and ongoing damage following SCI, although such cascades may worsen other CNS insults. We attribute our results to overwhelming effects of much greater release by direct mechanical damage and reversal of transport following SCI.
Collapse
Affiliation(s)
- David J McAdoo
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555-1043, USA.
| | | | | | | | | | | | | |
Collapse
|
50
|
Xu GY, Hughes MG, Zhang L, Cain L, McAdoo DJ. Administration of glutamate into the spinal cord at extracellular concentrations reached post-injury causes functional impairments. Neurosci Lett 2005; 384:271-6. [PMID: 15925447 DOI: 10.1016/j.neulet.2005.04.100] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2005] [Revised: 03/31/2005] [Accepted: 04/30/2005] [Indexed: 11/15/2022]
Abstract
In vivo experiments addressing the role of released glutamate in damage caused by neurotrauma seldom administer glutamate itself because it usually produces relatively little damage when administered into central nervous system (CNS) tissue in vivo. However, because of recent observations that glutamate administered into the spinal cord at the levels attained following spinal cord injury (SCI) kills neurons and oligodendrocytes, we tested the effects of administering glutamate at those concentrations on locomotor function. The Basso-Beattie-Bresnahan (BBB) test and activity box measures demonstrated that those glutamate concentrations produce lasting functional impairments. Several parameters provided by the activity box provided sensitive measures of the degree of post-SCI impairment, demonstrating their substantial potential for evaluating outcomes of SCI. Results obtained also enhance evidence that glutamate toxicity contributes to secondary damage following SCI and suggest that damage to white matter is an important contributor to such damage.
Collapse
Affiliation(s)
- Guo-Ying Xu
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555-1043, USA
| | | | | | | | | |
Collapse
|