1
|
Kadam SD, Hegarty SV. Development of KCC2 therapeutics to treat neurological disorders. Front Mol Neurosci 2024; 17:1503070. [PMID: 39720463 PMCID: PMC11666659 DOI: 10.3389/fnmol.2024.1503070] [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: 09/28/2024] [Accepted: 11/27/2024] [Indexed: 12/26/2024] Open
Abstract
KCC2 is CNS neuron-specific chloride extruder, essential for the establishment and maintenance of the transmembrane chloride gradient, thereby enabling synaptic inhibition within the CNS. Herein, we highlight KCC2 hypofunction as a fundamental and conserved pathology contributing to neuronal circuit excitation/inhibition (E/I) imbalances that underly epilepsies, chronic pain, neuro-developmental/-traumatic/-degenerative/-psychiatric disorders. Indeed, downstream of both acquired and genetic factors, multiple pathologies (e.g., hyperexcitability and inflammation) converge to impair KCC2-dependent inhibition in CNS. When KCC2 hypofunction occurs, affected neurons are disinhibited due to impaired inhibitory responses to GABA/glycine. This causes neuronal hyperexcitability, disinhibition within neuron circuits, and disrupted neurological functions. More recently, KCC2 was identified as a genetically-validated target for epilepsy, intellectual disability, and autism spectrum disorder, and pathogenic mutations in human SLC12A5 gene were linked to psychiatric/mood disorders. The broad therapeutic utility of KCC2-upmodulating drugs relates to its critical role in determining inhibitory activity of GABAergic neurotransmission, a mechanism widely targeted by several drugs. However, in cases of KCC2 hypofunction GABAergic neurotransmission can be depolarizing/excitatory, thereby impairing endogenous neuronal inhibition while also limiting the effectiveness of existing therapeutics targeting/requiring GABAergic pathway inhibition. Several preclinical reports have shown that KCC2 upmodulating treatments rescue and increase the efficacy of anti-seizure and analgesic medications. Thus, a first-in-class KCC2-potentiating therapy would provide a novel mechanism for restoring physiological CNS inhibition and addressing drug resistance in patients with E/I imbalance pathologies. Herein, we discuss progress toward and further work needed to develop the first-in-class KCC2 therapeutics to treat neurological disorder patients.
Collapse
|
2
|
Bertomeu JB, Fioravanço LP, Ramis TR, Godinho DB, Nascimento AS, Lima GC, Furian AF, Oliveira MS, Fighera MR, Royes LFF. The Role of Ion-Transporting Proteins on Crosstalk Between the Skeletal Muscle and Central Nervous Systems Elicited by Physical Exercise. Mol Neurobiol 2024:10.1007/s12035-024-04613-7. [PMID: 39578339 DOI: 10.1007/s12035-024-04613-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 11/05/2024] [Indexed: 11/24/2024]
Abstract
A paradigm shift in the understanding of bidirectional interactions between peripheral and central nervous systems is essential for development of rehabilitation and preventive interventions based on physical exercise. Although a causal relationship has not been completely established, modulation of voltage-dependent ion channels (Ca2+, Cl-, K+, Na+, lactate-, H+) in skeletal and neuronal cells provides opportunities to maintain force production during exercise and reduce the risk of disease. However, there are caveats to consider when interpreting the effects of physical exercise on this bidirectional axis, since exercise protocol details (e.g., duration and intensity) have variable effects on this crosstalk. Therefore, an integrative perspective of the skeletal muscle and brain's communication pathway is discussed, and the role of physical exercise on such communication highway is explained in this review.
Collapse
Affiliation(s)
- Judit Borràs Bertomeu
- Graduate Program in Biological Sciences: Toxicological Biochemistry, Center in Natural and Exact Sciences, Federal University of Santa Maria, Santa Maria, RS, Brazil
- Exercise Biochemistry Laboratory (BIOEX), Department of Sports Methods and Techniques, Physical Education and Sports Center, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Letícia Paiva Fioravanço
- Graduate Program in Biological Sciences: Toxicological Biochemistry, Center in Natural and Exact Sciences, Federal University of Santa Maria, Santa Maria, RS, Brazil
- Exercise Biochemistry Laboratory (BIOEX), Department of Sports Methods and Techniques, Physical Education and Sports Center, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Thiago Rozales Ramis
- Graduate Program in Biological Sciences: Toxicological Biochemistry, Center in Natural and Exact Sciences, Federal University of Santa Maria, Santa Maria, RS, Brazil
- Exercise Biochemistry Laboratory (BIOEX), Department of Sports Methods and Techniques, Physical Education and Sports Center, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Douglas Buchmann Godinho
- Graduate Program in Biological Sciences: Toxicological Biochemistry, Center in Natural and Exact Sciences, Federal University of Santa Maria, Santa Maria, RS, Brazil
- Exercise Biochemistry Laboratory (BIOEX), Department of Sports Methods and Techniques, Physical Education and Sports Center, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Alexandre Seixas Nascimento
- Graduate Program in Biological Sciences: Toxicological Biochemistry, Center in Natural and Exact Sciences, Federal University of Santa Maria, Santa Maria, RS, Brazil
- Exercise Biochemistry Laboratory (BIOEX), Department of Sports Methods and Techniques, Physical Education and Sports Center, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Gabriel Corrêa Lima
- Graduate Program in Biological Sciences: Toxicological Biochemistry, Center in Natural and Exact Sciences, Federal University of Santa Maria, Santa Maria, RS, Brazil
- Exercise Biochemistry Laboratory (BIOEX), Department of Sports Methods and Techniques, Physical Education and Sports Center, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Ana Flavia Furian
- Graduate Program in Pharmacology, Health Sciences Center, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Mauro Schneider Oliveira
- Graduate Program in Pharmacology, Health Sciences Center, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Michele Rechia Fighera
- Graduate Program in Biological Sciences: Toxicological Biochemistry, Center in Natural and Exact Sciences, Federal University of Santa Maria, Santa Maria, RS, Brazil
- Exercise Biochemistry Laboratory (BIOEX), Department of Sports Methods and Techniques, Physical Education and Sports Center, Federal University of Santa Maria, Santa Maria, RS, Brazil
- Laboratory of Experimental and Clinical Neuropsychiatry, Department of Neuropsychiatry, Health Sciences Center, Federal University of Santa Maria, Santa Maria, RS, 97105-900, Brazil
| | - Luiz Fernando Freire Royes
- Graduate Program in Biological Sciences: Toxicological Biochemistry, Center in Natural and Exact Sciences, Federal University of Santa Maria, Santa Maria, RS, Brazil.
- Exercise Biochemistry Laboratory (BIOEX), Department of Sports Methods and Techniques, Physical Education and Sports Center, Federal University of Santa Maria, Santa Maria, RS, Brazil.
- Department of Sports Methods and Techniques, Center of Physical Education and , Sports, Federal University of Santa Maria - UFSM, Santa Maria, RS, 97105-900, Brazil.
| |
Collapse
|
3
|
Hassan OI, Takamiya S, Asgarihafshejani A, Fehlings MG. Bridging the gap: a translational perspective in spinal cord injury. Exp Biol Med (Maywood) 2024; 249:10266. [PMID: 39391076 PMCID: PMC11464315 DOI: 10.3389/ebm.2024.10266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 08/27/2024] [Indexed: 10/12/2024] Open
Abstract
Traumatic spinal cord injury (SCI) is a devastating and complex condition to treat with no curative options. In the past few decades, rapid advancements in our understanding of SCI pathophysiology as well as the mergence of new treatments has created more optimism. Focusing on clinical translation, this paper provides a comprehensive overview of SCI through its epidemiology, pathophysiology, currently employed management strategies, and emerging therapeutic approaches. Additionally, it emphasizes the importance of addressing the heavy quality of life (QoL) challenges faced by SCI patients and their desires, providing a basis to tailor patient-centric forms of care. Furthermore, this paper discusses the frequently encountered barriers in translation from preclinical models to clinical settings. It also seeks to summarize significant completed and ongoing SCI clinical trials focused on neuroprotective and neuroregenerative strategies. While developing a cohesive regenerative treatment strategy remains challenging, even modest improvements in sensory and motor function can offer meaningful benefits and motivation for patients coping with this highly debilitating condition.
Collapse
Affiliation(s)
- Omar Imad Hassan
- Division of Genetics and Development, Krembil Brain Institute, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Soichiro Takamiya
- Division of Genetics and Development, Krembil Brain Institute, University Health Network, Toronto, ON, Canada
| | - Azam Asgarihafshejani
- Division of Genetics and Development, Krembil Brain Institute, University Health Network, Toronto, ON, Canada
| | - Michael G. Fehlings
- Division of Genetics and Development, Krembil Brain Institute, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Division of Neurosurgery, Krembil Neuroscience Centre, Toronto Western Hospital, University Health Network, Toronto, ON, Canada
- Division of Neurosurgery and Spine Program, Department of Surgery, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
4
|
Li Q, Sandoval A, Moth J, Shang J, Liew JY, Dunn T, Yang Z, Su J, Henwood M, Williams P, Chen B. Reduction of prolonged excitatory neuron swelling after spinal cord injury improves locomotor recovery in mice. Sci Transl Med 2024; 16:eadn7095. [PMID: 39321270 DOI: 10.1126/scitranslmed.adn7095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 04/09/2024] [Accepted: 09/05/2024] [Indexed: 09/27/2024]
Abstract
Spinal cord injury (SCI) results in acute damage and triggers secondary injury responses with sustained neuronal loss and dysfunction. However, the underlying mechanisms for these delayed neuronal pathologies are not entirely understood. SCI results in the swelling of spinal neurons, but the contribution of cell swelling to neuronal loss and functional deficits after SCI has not been systematically characterized. In this study, we devised a three-dimensional image analysis pipeline to evaluate spinal neurons, examining their types, quantities, volumes, and spatial distribution in a double-lateral hemisection SCI mouse model. We found that both excitatory and inhibitory neurons swell and are lost, albeit with distinct temporal patterns. Inhibitory neurons demonstrated marked swelling and decline in number on day 2 after SCI, which resolved by day 14. In contrast, excitatory neurons maintained persistent swelling and continued cell loss for at least 35 days after SCI in mice. Excitatory neurons exhibited sustained expression of the Na+-K+-Cl- cotransporter 1 (NKCC1), whereas inhibitory neurons down-regulated the protein by day 14 after SCI. Treatment with a Food and Drug Administration-approved NKCC1 inhibitor, bumetanide, mitigated swelling of excitatory neurons and reduced their loss in the secondary injury phase after SCI. The administration of bumetanide after SCI in mouse improved locomotor recovery, with functional benefits persisting for at least 4 weeks after treatment cessation. This study advances our understanding of SCI-related pathology and introduces bumetanide as a potential treatment to mitigate sustained neuronal swelling and enhance recovery after SCI.
Collapse
Affiliation(s)
- Qiang Li
- Department of Neurobiology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Alfredo Sandoval
- Department of Neurobiology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - John Moth
- Department of Anesthesiology, Houston Methodist Hospital, Houston, TX 77030, USA
| | - Junkui Shang
- Department of Neurobiology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Jia Yi Liew
- Department of Neurobiology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Tiffany Dunn
- Department of Neurobiology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Zhiyun Yang
- F. M. Kirby Neurobiology Center, Boston Children's Hospital and Departments of Neurology and Ophthalmology, Harvard Medical School, Boston, MA 02115, USA
| | - Junfeng Su
- F. M. Kirby Neurobiology Center, Boston Children's Hospital and Departments of Neurology and Ophthalmology, Harvard Medical School, Boston, MA 02115, USA
| | - Melissa Henwood
- Department of Neurobiology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Philip Williams
- Department of Ophthalmology and Visual Sciences and Department of Neuroscience, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Bo Chen
- Department of Neurobiology, University of Texas Medical Branch, Galveston, TX 77555, USA
| |
Collapse
|
5
|
Ames S, Brooks J, Jones E, Morehouse J, Cortez-Thomas F, Desta D, Stirling DP. NKCC1 inhibition reduces periaxonal swelling, increases white matter sparing, and improves neurological recovery after contusive SCI. Neurobiol Dis 2024; 199:106611. [PMID: 39032797 DOI: 10.1016/j.nbd.2024.106611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 07/12/2024] [Accepted: 07/16/2024] [Indexed: 07/23/2024] Open
Abstract
Ultrastructural studies of contusive spinal cord injury (SCI) in mammals have shown that the most prominent acute changes in white matter are periaxonal swelling and separation of myelin away from their axon, axonal swelling, and axonal spheroid formation. However, the underlying cellular and molecular mechanisms that cause periaxonal swelling and the functional consequences are poorly understood. We hypothesized that periaxonal swelling and loss of connectivity between the axo-myelinic interface impedes neurological recovery by disrupting conduction velocity, and glial to axonal trophic support resulting in axonal swelling and spheroid formation. Utilizing in vivo longitudinal imaging of Thy1YFP+ axons and myelin labeled with Nile red, we reveal that periaxonal swelling significantly increases acutely following a contusive SCI (T13, 30 kdyn, IH Impactor) versus baseline recordings (laminectomy only) and often precedes axonal spheroid formation. In addition, using longitudinal imaging to determine the fate of myelinated fibers acutely after SCI, we show that ∼73% of myelinated fibers present with periaxonal swelling at 1 h post SCI and ∼ 51% of those fibers transition to axonal spheroids by 4 h post SCI. Next, we assessed whether cation-chloride cotransporters present within the internode contributed to periaxonal swelling and whether their modulation would increase white matter sparing and improve neurological recovery following a moderate contusive SCI (T9, 50 kdyn). Mechanistically, activation of the cation-chloride cotransporter KCC2 did not improve neurological recovery and acute axonal survival, but did improve chronic tissue sparing. In distinction, the NKKC1 antagonist bumetanide improved neurological recovery, tissue sparing, and axonal survival, in part through preventing periaxonal swelling and disruption of the axo-myelinic interface. Collectively, these data reveal a novel neuroprotective target to prevent periaxonal swelling and improve neurological recovery after SCI.
Collapse
Affiliation(s)
- Spencer Ames
- Kentucky Spinal Cord Injury Research Center, University of Louisville, School of Medicine, Louisville, KY, USA 40202
| | - Jesse Brooks
- Kentucky Spinal Cord Injury Research Center, University of Louisville, School of Medicine, Louisville, KY, USA 40202
| | - Emma Jones
- Kentucky Spinal Cord Injury Research Center, University of Louisville, School of Medicine, Louisville, KY, USA 40202
| | - Johnny Morehouse
- Kentucky Spinal Cord Injury Research Center, University of Louisville, School of Medicine, Louisville, KY, USA 40202
| | - Francisco Cortez-Thomas
- Kentucky Spinal Cord Injury Research Center, University of Louisville, School of Medicine, Louisville, KY, USA 40202; Bioengineering, University of Louisville, School of Medicine, Louisville, KY, USA 40202
| | - Dereje Desta
- Kentucky Spinal Cord Injury Research Center, University of Louisville, School of Medicine, Louisville, KY, USA 40202
| | - David P Stirling
- Kentucky Spinal Cord Injury Research Center, University of Louisville, School of Medicine, Louisville, KY, USA 40202; Departments of Neurological Surgery, University of Louisville, School of Medicine, Louisville, KY, USA 40202; Anatomical Sciences and Neurobiology, University of Louisville, School of Medicine, Louisville, KY, USA 40202.
| |
Collapse
|
6
|
Malloy DC, Côté MP. Multi-session transcutaneous spinal cord stimulation prevents chloride homeostasis imbalance and the development of hyperreflexia after spinal cord injury in rat. Exp Neurol 2024; 376:114754. [PMID: 38493983 PMCID: PMC11519955 DOI: 10.1016/j.expneurol.2024.114754] [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: 12/01/2023] [Revised: 02/28/2024] [Accepted: 03/11/2024] [Indexed: 03/19/2024]
Abstract
Spasticity is a complex and multidimensional disorder that impacts nearly 75% of individuals with spinal cord injury (SCI) and currently lacks adequate treatment options. This sensorimotor condition is burdensome as hyperexcitability of reflex pathways result in exacerbated reflex responses, co-contractions of antagonistic muscles, and involuntary movements. Transcutaneous spinal cord stimulation (tSCS) has become a popular tool in the human SCI research field. The likeliness for this intervention to be successful as a noninvasive anti-spastic therapy after SCI is suggested by a mild and transitory improvement in spastic symptoms following a single stimulation session, but it remains to be determined if repeated tSCS over the course of weeks can produce more profound effects. Despite its popularity, the neuroplasticity induced by tSCS also remains widely unexplored, particularly due to the lack of suitable animal models to investigate this intervention. Thus, the basis of this work was to use tSCS over multiple sessions (multi-session tSCS) in a rat model to target spasticity after SCI and identify the long-term physiological improvements and anatomical neuroplasticity occurring in the spinal cord. Here, we show that multi-session tSCS in rats with an incomplete (severe T9 contusion) SCI (1) decreases hyperreflexia, (2) increases the low frequency-dependent modulation of the H-reflex, (3) prevents potassium-chloride cotransporter isoform 2 (KCC2) membrane downregulation in lumbar motoneurons, and (4) generally augments motor output, i.e., EMG amplitude in response to single pulses of tSCS, particularly in extensor muscles. Together, this work displays that multi-session tSCS can target and diminish spasticity after SCI as an alternative to pharmacological interventions and begins to highlight the underlying neuroplasticity contributing to its success in improving functional recovery.
Collapse
Affiliation(s)
- Dillon C Malloy
- Marion Murray Spinal Cord Research Center, Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, United States of America.
| | - Marie-Pascale Côté
- Marion Murray Spinal Cord Research Center, Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, United States of America.
| |
Collapse
|
7
|
He Y, Xu Y, Hai M, Feng Y, Liu P, Chen Z, Duan W. Exoskeleton-Assisted Rehabilitation and Neuroplasticity in Spinal Cord Injury. World Neurosurg 2024; 185:45-54. [PMID: 38320651 DOI: 10.1016/j.wneu.2024.01.167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 01/29/2024] [Accepted: 01/30/2024] [Indexed: 02/08/2024]
Abstract
Spinal cord injury (SCI) results in neurological deficits below the level of injury, causing motor dysfunction and various severe multisystem complications. Rehabilitative training plays a crucial role in the recovery of individuals with SCI, and exoskeleton serves as an emerging and promising tool for rehabilitation, especially in promoting neuroplasticity and alleviating SCI-related complications. This article reviews the classifications and research progresses of medical exoskeletons designed for SCI patients and describes their performances in practical application separately. Meanwhile, we discuss their mechanisms for enhancing neuroplasticity and functional remodeling, as well as their palliative impacts on secondary complications. The potential trends in exoskeleton design are raised according to current progress and requirements on SCI rehabilitation.
Collapse
Affiliation(s)
- Yana He
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China; School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Yuxuan Xu
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China; School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Minghang Hai
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China; School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Yang Feng
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China; School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Penghao Liu
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China; Lab of Spinal Cord Injury and Functional Reconstruction, China International Neuroscience Institute(CHINA-INI), Beijing, China
| | - Zan Chen
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China; Lab of Spinal Cord Injury and Functional Reconstruction, China International Neuroscience Institute(CHINA-INI), Beijing, China
| | - Wanru Duan
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China; Lab of Spinal Cord Injury and Functional Reconstruction, China International Neuroscience Institute(CHINA-INI), Beijing, China.
| |
Collapse
|
8
|
Kerzonkuf M, Verneuil J, Brocard C, Dingu N, Trouplin V, Ramirez Franco JJ, Bartoli M, Brocard F, Bras H. Knockdown of calpain1 in lumbar motoneurons reduces spasticity after spinal cord injury in adult rats. Mol Ther 2024; 32:1096-1109. [PMID: 38291756 PMCID: PMC11163198 DOI: 10.1016/j.ymthe.2024.01.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 12/23/2023] [Accepted: 01/23/2024] [Indexed: 02/01/2024] Open
Abstract
Spasticity, affecting ∼75% of patients with spinal cord injury (SCI), leads to hyperreflexia, muscle spasms, and cocontractions of antagonist muscles, greatly affecting their quality of life. Spasticity primarily stems from the hyperexcitability of motoneurons below the lesion, driven by an upregulation of the persistent sodium current and a downregulation of chloride extrusion. This imbalance results from the post-SCI activation of calpain1, which cleaves Nav1.6 channels and KCC2 cotransporters. Our study was focused on mitigating spasticity by specifically targeting calpain1 in spinal motoneurons. We successfully transduced lumbar motoneurons in adult rats with SCI using intrathecal administration of adeno-associated virus vector serotype 6, carrying a shRNA sequence against calpain1. This approach significantly reduced calpain1 expression in transduced motoneurons, leading to a noticeable decrease in spasticity symptoms, including hyperreflexia, muscle spasms, and cocontractions in hindlimb muscles, which are particularly evident in the second month post-SCI. In addition, this decrease, which prevented the escalation of spasticity to a severe grade, paralleled the restoration of KCC2 levels in transduced motoneurons, suggesting a reduced proteolytic activity of calpain1. These findings demonstrate that inhibiting calpain1 in motoneurons is a promising strategy for alleviating spasticity in SCI patients.
Collapse
Affiliation(s)
- Marjorie Kerzonkuf
- Institut des Neurosciences de la Timone (UMR7289), Aix-Marseille Université and CNRS, Marseille, France
| | - Jérémy Verneuil
- Institut des Neurosciences de la Timone (UMR7289), Aix-Marseille Université and CNRS, Marseille, France
| | - Cécile Brocard
- Institut des Neurosciences de la Timone (UMR7289), Aix-Marseille Université and CNRS, Marseille, France
| | - Nejada Dingu
- Institut des Neurosciences de la Timone (UMR7289), Aix-Marseille Université and CNRS, Marseille, France
| | - Virginie Trouplin
- Institut des Neurosciences de la Timone (UMR7289), Aix-Marseille Université and CNRS, Marseille, France
| | - Jose Jorge Ramirez Franco
- Institut des Neurosciences de la Timone (UMR7289), Aix-Marseille Université and CNRS, Marseille, France
| | - Marc Bartoli
- Institut Marseille Maladies Rares (MarMaRa), Aix-Marseille Université and INSERM, Marseille, France
| | - Frédéric Brocard
- Institut des Neurosciences de la Timone (UMR7289), Aix-Marseille Université and CNRS, Marseille, France.
| | - Hélène Bras
- Institut des Neurosciences de la Timone (UMR7289), Aix-Marseille Université and CNRS, Marseille, France.
| |
Collapse
|
9
|
Zhang X, Zhang Y, Su Q, Liu Y, Li Z, Yong VW, Xue M. Ion Channel Dysregulation Following Intracerebral Hemorrhage. Neurosci Bull 2024; 40:401-414. [PMID: 37755675 PMCID: PMC10912428 DOI: 10.1007/s12264-023-01118-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 06/14/2023] [Indexed: 09/28/2023] Open
Abstract
Injury to the brain after intracerebral hemorrhage (ICH) results from numerous complex cellular mechanisms. At present, effective therapy for ICH is limited and a better understanding of the mechanisms of brain injury is necessary to improve prognosis. There is increasing evidence that ion channel dysregulation occurs at multiple stages in primary and secondary brain injury following ICH. Ion channels such as TWIK-related K+ channel 1, sulfonylurea 1 transient receptor potential melastatin 4 and glutamate-gated channels affect ion homeostasis in ICH. They in turn participate in the formation of brain edema, disruption of the blood-brain barrier, and the generation of neurotoxicity. In this review, we summarize the interaction between ions and ion channels, the effects of ion channel dysregulation, and we discuss some therapeutics based on ion-channel modulation following ICH.
Collapse
Affiliation(s)
- Xiangyu Zhang
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China
- Academy of Medical Science, Zhengzhou University, Zhengzhou, 450000, China
| | - Yan Zhang
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China
- Academy of Medical Science, Zhengzhou University, Zhengzhou, 450000, China
| | - Qiuyang Su
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China
- Academy of Medical Science, Zhengzhou University, Zhengzhou, 450000, China
| | - Yang Liu
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China
- Academy of Medical Science, Zhengzhou University, Zhengzhou, 450000, China
| | - Zhe Li
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China
- Academy of Medical Science, Zhengzhou University, Zhengzhou, 450000, China
| | - V Wee Yong
- Hotchkiss Brain Institute and Department of Clinical Neurosciences, University of Calgary, Calgary, AB, T2N 1N4, Canada.
| | - Mengzhou Xue
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China.
- Academy of Medical Science, Zhengzhou University, Zhengzhou, 450000, China.
| |
Collapse
|
10
|
Grau JW, Hudson KE, Johnston DT, Partipilo SR. Updating perspectives on spinal cord function: motor coordination, timing, relational processing, and memory below the brain. Front Syst Neurosci 2024; 18:1184597. [PMID: 38444825 PMCID: PMC10912355 DOI: 10.3389/fnsys.2024.1184597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Accepted: 01/29/2024] [Indexed: 03/07/2024] Open
Abstract
Those studying neural systems within the brain have historically assumed that lower-level processes in the spinal cord act in a mechanical manner, to relay afferent signals and execute motor commands. From this view, abstracting temporal and environmental relations is the province of the brain. Here we review work conducted over the last 50 years that challenges this perspective, demonstrating that mechanisms within the spinal cord can organize coordinated behavior (stepping), induce a lasting change in how pain (nociceptive) signals are processed, abstract stimulus-stimulus (Pavlovian) and response-outcome (instrumental) relations, and infer whether stimuli occur in a random or regular manner. The mechanisms that underlie these processes depend upon signal pathways (e.g., NMDA receptor mediated plasticity) analogous to those implicated in brain-dependent learning and memory. New data show that spinal cord injury (SCI) can enable plasticity within the spinal cord by reducing the inhibitory effect of GABA. It is suggested that the signals relayed to the brain may contain information about environmental relations and that spinal cord systems can coordinate action in response to descending signals from the brain. We further suggest that the study of stimulus processing, learning, memory, and cognitive-like processing in the spinal cord can inform our views of brain function, providing an attractive model system. Most importantly, the work has revealed new avenues of treatment for those that have suffered a SCI.
Collapse
Affiliation(s)
- James W. Grau
- Lab of Dr. James Grau, Department of Psychological and Brain Sciences, Cellular and Behavioral Neuroscience, Texas A&M University, College Station, TX, United States
| | | | | | | |
Collapse
|
11
|
Malloy DC, Côté MP. Multi-session transcutaneous spinal cord stimulation prevents chloridehomeostasis imbalance and the development of spasticity after spinal cordinjury in rat. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.24.563419. [PMID: 37961233 PMCID: PMC10634766 DOI: 10.1101/2023.10.24.563419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Spasticity is a complex and multidimensional disorder that impacts nearly 75% of individuals with spinal cord injury (SCI) and currently lacks adequate treatment options. This sensorimotor condition is burdensome as hyperexcitability of reflex pathways result in exacerbated reflex responses, co-contractions of antagonistic muscles, and involuntary movements. Transcutaneous spinal cord stimulation (tSCS) has become a popular tool in the human SCI research field. The likeliness for this intervention to be successful as a noninvasive anti-spastic therapy after SCI is suggested by a mild and transitory improvement in spastic symptoms following a single stimulation session, but it remains to be determined if repeated tSCS over the course of weeks can produce more profound effects. Despite its popularity, the neuroplasticity induced by tSCS also remains widely unexplored, particularly due to the lack of suitable animal models to investigate this intervention. Thus, the basis of this work was to use tSCS over multiple sessions (multi-session tSCS) in a rat model to target spasticity after SCI and identify the long-term physiological improvements and anatomical neuroplasticity occurring in the spinal cord. Here, we show that multi-session tSCS in rats with an incomplete (severe T9 contusion) SCI (1) decreases hyperreflexia, (2) increases the low frequency-dependent modulation of the H-reflex, (3) prevents potassium-chloride cotransporter isoform 2 (KCC2) membrane downregulation in lumbar motoneurons, and (4) generally augments motor output, i.e., EMG amplitude in response to single pulses of tSCS, particularly in extensor muscles. Together, this work displays that multi-session tSCS can target and diminish spasticity after SCI as an alternative to pharmacological interventions and begins to highlight the underlying neuroplasticity contributing to its success in improving functional recovery.
Collapse
Affiliation(s)
- Dillon C. Malloy
- Marion Murray Spinal Cord Research Center, Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129
| | - Marie-Pascale Côté
- Marion Murray Spinal Cord Research Center, Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129
| |
Collapse
|
12
|
Hugues N, Pin-Barre C, Brioche T, Pellegrino C, Berton E, Rivera C, Laurin J. High-intensity training with short and long intervals regulate cortical neurotrophic factors, apoptosis markers and chloride homeostasis in rats with stroke. Physiol Behav 2023; 266:114190. [PMID: 37055005 DOI: 10.1016/j.physbeh.2023.114190] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 03/29/2023] [Accepted: 04/04/2023] [Indexed: 04/15/2023]
Abstract
BACKGROUND/PURPOSE The optimal endurance exercise parameters remain to be defined to potentiate long-term functional recovery after stroke. We aim to assess the effects of individualized high-intensity interval training (HIIT) with either long or short intervals on neurotrophic factors and their receptors, apoptosis markers and the two-main cation-chloride cotransporters in the ipsi- and contralesional cerebral cortices in rats with cerebral ischemia. Endurance performance and sensorimotor functions were also assessed METHODS: : Rats with a 2-hour transient middle cerebral artery occlusion (tMCAO) performed work-matched HIIT4 (intervals: 4min) or HIIT1 (intervals: 1min) on treadmill for 2 weeks. Incremental exercises and sensorimotor tests were performed at day 1 (D1), D8, and D15 after tMCAO. Molecular analyses were achieved in both the paretic and non-paretic triceps brachii muscles and the ipsi- and contralesional cortices at D17 RESULTS: : Gains in endurance performance are in a time-dependent manner from the first week of training. This enhancement is supported by the upregulation of metabolic markers in both triceps brachii muscles. Both regimens alter the expression of neurotrophic markers and chloride homeostasis in a specific manner in the ipsi- and contralesional cortices. HIIT acts on apoptosis markers by promoting anti-apoptotic proteins in the ipsilesional cortex CONCLUSION: : HIIT regimens seem to be of clinical relevance in the critical period of stroke rehabilitation by strongly improving aerobic performance. Also, the observed cortical changes suggest an influence of HIIT on neuroplasticity in both ipsi- and contralesional hemispheres. Such neurotrophic markers might be considered as biomarkers of functional recovery in individuals with stroke.
Collapse
Affiliation(s)
- Nicolas Hugues
- Aix Marseille Univ, INSERM, INMED, Marseille, France; Aix Marseille Univ, CNRS, ISM, Marseille, France
| | | | - Thomas Brioche
- Université de Montpellier, INRAE, DMEM, Montpellier, France
| | | | - Eric Berton
- Aix Marseille Univ, CNRS, ISM, Marseille, France
| | | | - Jérôme Laurin
- Aix Marseille Univ, INSERM, INMED, Marseille, France.
| |
Collapse
|
13
|
Fogarty MJ. Inhibitory Synaptic Influences on Developmental Motor Disorders. Int J Mol Sci 2023; 24:ijms24086962. [PMID: 37108127 PMCID: PMC10138861 DOI: 10.3390/ijms24086962] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 04/05/2023] [Accepted: 04/06/2023] [Indexed: 04/29/2023] Open
Abstract
During development, GABA and glycine play major trophic and synaptic roles in the establishment of the neuromotor system. In this review, we summarise the formation, function and maturation of GABAergic and glycinergic synapses within neuromotor circuits during development. We take special care to discuss the differences in limb and respiratory neuromotor control. We then investigate the influences that GABAergic and glycinergic neurotransmission has on two major developmental neuromotor disorders: Rett syndrome and spastic cerebral palsy. We present these two syndromes in order to contrast the approaches to disease mechanism and therapy. While both conditions have motor dysfunctions at their core, one condition Rett syndrome, despite having myriad symptoms, has scientists focused on the breathing abnormalities and their alleviation-to great clinical advances. By contrast, cerebral palsy remains a scientific quagmire or poor definitions, no widely adopted model and a lack of therapeutic focus. We conclude that the sheer abundance of diversity of inhibitory neurotransmitter targets should provide hope for intractable conditions, particularly those that exhibit broad spectra of dysfunction-such as spastic cerebral palsy and Rett syndrome.
Collapse
Affiliation(s)
- Matthew J Fogarty
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN 55902, USA
| |
Collapse
|
14
|
Caron G, Bilchak J, Marie-Pascale Côté. Bumetanide increases postsynaptic inhibition after chronic SCI and decreases presynaptic inhibition with step-training. J Physiol 2023; 601:1425-1447. [PMID: 36847245 PMCID: PMC10106440 DOI: 10.1113/jp283753] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 02/16/2023] [Indexed: 03/01/2023] Open
Abstract
Current anti-spastic medication significantly compromises motor recovery after spinal cord injury (SCI), indicating a critical need for alternative interventions. Because a shift in chloride homeostasis decreases spinal inhibition and contributes to hyperreflexia after SCI, we investigated the effect of bumetanide, an FDA-approved sodium-potassium-chloride intruder (NKCC1) antagonist, on presynaptic and postsynaptic inhibition. We compared its effect with step-training as it is known to improve spinal inhibition by restoring chloride homeostasis. In SCI rats, a prolonged bumetanide treatment increased postynaptic inhibition but not presynaptic inhibition of the plantar H-reflex evoked by posterior biceps and semitendinosus (PBSt) group I afferents. By using in vivo intracellular recordings of motoneurons, we further show that a prolonged bumetanide increased postsynaptic inhibition by hyperpolarizing the reversal potential for inhibitory postsynaptic potentials (IPSPs) after SCI. However, in step-trained SCI rats an acute delivery of bumetanide decreased presynaptic inhibition of the H-reflex, but not postsynaptic inhibition. These results suggest that bumetanide might be a viable option to improve postsynaptic inhibition after SCI, but it also decreases the recovery of presynaptic inhibition with step-training. We discuss whether the effects of bumetanide are mediated by NKCC1 or by off-target effects. KEY POINTS: After spinal cord injury (SCI), chloride homeostasis is dysregulated over time in parallel with the decrease in presynaptic inhibition of Ia afferents and postsynaptic inhibition of motoneurons, and the development of spasticity. While step-training counteracts these effects, it cannot always be implemented in the clinic because of comorbidities. An alternative intervention is to use pharmacological strategies to decrease spasticity without hindering the recovery of motor function with step-training. Here we found that, after SCI, a prolonged bumetanide (an FDA-approved antagonist of the sodium-potassium-chloride intruder, NKCC1) treatment increases postsynaptic inhibition of the H-reflex, and it hyperpolarizes the reversal potential for inhibitory postsynaptic potentials in motoneurons. However, in step-trained SCI, an acute delivery of bumetanide decreases presynaptic inhibition of the H-reflex, but not postsynaptic inhibition. Our results suggest that bumetanide has the potential to decrease spastic symptoms related to a decrease in postsynaptic but not presynaptic inhibition after SCI.
Collapse
Affiliation(s)
- Guillaume Caron
- Marion Murray Spinal Cord Research Center, Department of Neurobiology and Anatomy, Drexel University, Philadelphia, PA 19129
| | - Jadwiga Bilchak
- Marion Murray Spinal Cord Research Center, Department of Neurobiology and Anatomy, Drexel University, Philadelphia, PA 19129
| | - Marie-Pascale Côté
- Marion Murray Spinal Cord Research Center, Department of Neurobiology and Anatomy, Drexel University, Philadelphia, PA 19129
| |
Collapse
|
15
|
Talifu Z, Pan Y, Gong H, Xu X, Zhang C, Yang D, Gao F, Yu Y, Du L, Li J. The role of KCC2 and NKCC1 in spinal cord injury: From physiology to pathology. Front Physiol 2022; 13:1045520. [PMID: 36589461 PMCID: PMC9799334 DOI: 10.3389/fphys.2022.1045520] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 12/05/2022] [Indexed: 12/23/2022] Open
Abstract
The balance of ion concentrations inside and outside the cell is an essential homeostatic mechanism in neurons and serves as the basis for a variety of physiological activities. In the central nervous system, NKCC1 and KCC2, members of the SLC12 cation-chloride co-transporter (CCC) family, participate in physiological and pathophysiological processes by regulating intracellular and extracellular chloride ion concentrations, which can further regulate the GABAergic system. Over recent years, studies have shown that NKCC1 and KCC2 are essential for the maintenance of Cl- homeostasis in neural cells. NKCC1 transports Cl- into cells while KCC2 transports Cl- out of cells, thereby regulating chloride balance and neuronal excitability. An imbalance of NKCC1 and KCC2 after spinal cord injury will disrupt CI- homeostasis, resulting in the transformation of GABA neurons from an inhibitory state into an excitatory state, which subsequently alters the spinal cord neural network and leads to conditions such as spasticity and neuropathic pain, among others. Meanwhile, studies have shown that KCC2 is also an essential target for motor function reconstruction after spinal cord injury. This review mainly introduces the physiological structure and function of NKCC1 and KCC2 and discusses their pathophysiological roles after spinal cord injury.
Collapse
Affiliation(s)
- Zuliyaer Talifu
- School of Rehabilitation, Capital Medical University, Beijing, China,Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China,Chinese Institute of Rehabilitation Science, Beijing, China,Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China,Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China,School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao, China
| | - Yunzhu Pan
- School of Rehabilitation, Capital Medical University, Beijing, China,Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China,Chinese Institute of Rehabilitation Science, Beijing, China,Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China,Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China,School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao, China
| | - Han Gong
- School of Rehabilitation, Capital Medical University, Beijing, China,Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China,Chinese Institute of Rehabilitation Science, Beijing, China,Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China,Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China
| | - Xin Xu
- School of Rehabilitation, Capital Medical University, Beijing, China,Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China,Chinese Institute of Rehabilitation Science, Beijing, China,Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China,Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China
| | - Chunjia Zhang
- School of Rehabilitation, Capital Medical University, Beijing, China,Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China,Chinese Institute of Rehabilitation Science, Beijing, China,Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China,Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China
| | - Degang Yang
- School of Rehabilitation, Capital Medical University, Beijing, China,Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China,Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China,Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China
| | - Feng Gao
- School of Rehabilitation, Capital Medical University, Beijing, China,Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China,Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China,Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China
| | - Yan Yu
- School of Rehabilitation, Capital Medical University, Beijing, China,Chinese Institute of Rehabilitation Science, Beijing, China,Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China,Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China
| | - Liangjie Du
- School of Rehabilitation, Capital Medical University, Beijing, China,Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China,Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China,Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China,*Correspondence: Liangjie Du, ; Jianjun Li,
| | - Jianjun Li
- School of Rehabilitation, Capital Medical University, Beijing, China,Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China,Chinese Institute of Rehabilitation Science, Beijing, China,Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China,Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China,School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao, China,*Correspondence: Liangjie Du, ; Jianjun Li,
| |
Collapse
|
16
|
Hashemizadeh S, Gharaylou Z, Hosseindoost S, Sardari M, Omidi A, Hosseini ravandi H, Hadjighassem M. Long-term administration of bumetanide improve functional recovery after spinal cord injury in rats. Front Pharmacol 2022; 13:932487. [PMID: 36339604 PMCID: PMC9628211 DOI: 10.3389/fphar.2022.932487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 08/24/2022] [Indexed: 11/16/2022] Open
Abstract
Ion disturbances are among the most remarkable deficits in spinal cord injury (SCI). GABA is an integral part of neural interaction. Action of the GABAA receptor depends on the amount of intracellular chloride. Homeostasis of chloride is controlled by two co-transporters, NKCC1 and KCC2. Previous studies revealed that NKCC1 are disturbed in SCI. In this study, NKCC1 is highly expressed in the epicenter of the lesioned spinal cord at 3 hours after induction of the lesion and reached the peak around 6 hours after SCI. Bumetanide (2 and 4 mg/day), as a specific NKCC1 inhibitor, was used at 3 hours post SCI for 28 days. The functional recovery outcomes were measured by the Basso-Beattie-Bresnahan (BBB) locomotor rating scale, ladder walking test, and hot plate test. The rats that received bumetanide 4 mg/day exhibited improved recovery of locomotor function, reduction of NKCC1 gene expression, and upregulation of GAP protein levels 28 days post SCI. Histological tissue evaluations confirmed bumetanide's neuroprotective and regenerative effects. This study provides novel evidence for the benefits of bumetanide in early administration after SCI.
Collapse
Affiliation(s)
- Shiva Hashemizadeh
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
- School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Saereh Hosseindoost
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
- Pain Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Sardari
- Department of Animal Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Ameneh Omidi
- Department of Anatomical Sciences, Faculty of Medical Science, Tarbiat Modares University, Tehran, Iran
| | | | - Mahmoudreza Hadjighassem
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
- School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
17
|
Grau JW, Hudson KE, Tarbet MM, Strain MM. Behavioral studies of spinal conditioning: The spinal cord is smarter than you think it is. JOURNAL OF EXPERIMENTAL PSYCHOLOGY. ANIMAL LEARNING AND COGNITION 2022; 48:435-457. [PMID: 35901417 PMCID: PMC10391333 DOI: 10.1037/xan0000332] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
In 1988 Robert Rescorla published an article in the Annual Review of Neuroscience that addressed the circumstances under which learning occurs, some key methodological issues, and what constitutes an example of learning. The article has inspired a generation of neuroscientists, opening the door to a wider range of learning phenomena. After reviewing the historical context for his article, its key points are briefly reviewed. The perspective outlined enabled the study of learning in simpler preparations, such as the spinal cord. The period after 1988 revealed that pain (nociceptive) stimuli can induce a lasting sensitization of spinal cord circuits, laying down a kind of memory mediated by signal pathways analogous to those implicated in brain dependent learning and memory. Evidence suggests that the spinal cord is sensitive to instrumental response-outcome (R-O) relations, that learning can induce a peripheral modification (muscle memory) that helps maintain the learned response, and that learning can promote adaptive plasticity (a form of metaplasticity). Conversely, exposure to uncontrollable stimulation disables the capacity to learn. Spinal cord neurons can also abstract that stimuli occur in a regular (predictable) manner, a capacity that appears linked to a neural oscillator (central pattern generator). Disrupting communication with the brain has been shown to transform how GABA affects neuronal function (an example of ionic plasticity), releasing a brake that enables plasticity. We conclude by presenting a framework for understanding these findings and the implications for the broader study of learning. (PsycInfo Database Record (c) 2022 APA, all rights reserved).
Collapse
Affiliation(s)
- James W. Grau
- Cellular and Behavioral Neuroscience, Department of Psychology, Texas A&M University, College Station, TX, 77843 USA
| | - Kelsey E. Hudson
- Cellular and Behavioral Neuroscience, Department of Psychology, Texas A&M University, College Station, TX, 77843 USA
| | - Megan M. Tarbet
- McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX
| | - Misty M. Strain
- Department of Cellular and Integrative Physiology, University of Texas Health San Antonio, San Antonio, TX 78229
| |
Collapse
|
18
|
Lewis NE, Tabarestani TQ, Cellini BR, Zhang N, Marrotte EJ, Wang H, Laskowitz DT, Abd-El-Barr MM, Faw TD. Effect of Acute Physical Interventions on Pathophysiology and Recovery After Spinal Cord Injury: A Comprehensive Review of the Literature. Neurospine 2022; 19:671-686. [PMID: 36203293 PMCID: PMC9537860 DOI: 10.14245/ns.2244476.238] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 07/20/2022] [Indexed: 12/14/2022] Open
Abstract
Physical rehabilitation is essential for enhancing recovery in individuals with spinal cord injury (SCI); however, aside from early surgical intervention and hemodynamic management, there are no proven interventions for promoting recovery in the acute phase. In general, early rehabilitation is considered beneficial, but optimal parameters and potential contraindications for implementing rehabilitation at very early time points are unclear. Moreover, clinical trials to date are limited to studies initiating rehabilitation 2 weeks after injury and later. To address these gaps, this article reviews the preclinical literature on physical interventions initiated within the first 8 days postinjury. Effects of early rehabilitation on molecular and structural nervous system changes, behavioral function, and body systems are considered. Most studies utilized treadmill or cycle training as the primary intervention. Treadmill training initiated within the first 3 days and terminated by 1 week after injury worsened autonomic function, inflammation, and locomotor outcomes, while swim training during this period increased microvascular dysfunction. In contrast, lower-intensity rehabilitation such as reach training, ladder training, or voluntary wheel or ball training showed benefits when implemented during the first 3 days. Rehabilitation initiated at 4 days postinjury was also associated with enhanced motor recovery. Cycling appears to have the greatest risk-benefit ratio; however, the effects of cycle training in the first 3 days were not investigated. Overall, research suggests that lower intensity or voluntary rehabilitation during the hyperacute phase is more appropriate until at least 4 days postinjury, at which point higher-intensity activity becomes safer and more beneficial for recovery.
Collapse
Affiliation(s)
- Nicholle E. Lewis
- Doctor of Physical Therapy Division, Department of Orthopaedic Surgery, Duke University, Durham, NC, USA
| | | | - Brianna R. Cellini
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
| | - Nina Zhang
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
| | - Eric J. Marrotte
- Department of Neurology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Haichen Wang
- Department of Neurology, Duke University, Durham, NC, USA
| | | | | | - Timothy D. Faw
- Doctor of Physical Therapy Division, Department of Orthopaedic Surgery, Duke University, Durham, NC, USA,Duke Institute for Brain Sciences, Duke University, Durham, NC, USA,Corresponding Author Timothy D. Faw Doctor of Physical Therapy Division, Department of Orthopaedic Surgery, Duke University, 311 Research Drive, Durham, NC 21170, USA
| |
Collapse
|
19
|
Samejima S, Henderson R, Pradarelli J, Mondello SE, Moritz CT. Activity-dependent plasticity and spinal cord stimulation for motor recovery following spinal cord injury. Exp Neurol 2022; 357:114178. [PMID: 35878817 DOI: 10.1016/j.expneurol.2022.114178] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 06/22/2022] [Accepted: 07/16/2022] [Indexed: 02/07/2023]
Abstract
Spinal cord injuries lead to permanent physical impairment despite most often being anatomically incomplete disruptions of the spinal cord. Remaining connections between the brain and spinal cord create the potential for inducing neural plasticity to improve sensorimotor function, even many years after injury. This narrative review provides an overview of the current evidence for spontaneous motor recovery, activity-dependent plasticity, and interventions for restoring motor control to residual brain and spinal cord networks via spinal cord stimulation. In addition to open-loop spinal cord stimulation to promote long-term neuroplasticity, we also review a more targeted approach: closed-loop stimulation. Lastly, we review mechanisms of spinal cord neuromodulation to promote sensorimotor recovery, with the goal of advancing the field of rehabilitation for physical impairments following spinal cord injury.
Collapse
Affiliation(s)
- Soshi Samejima
- International Collaboration on Repair Discoveries, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada; Department of Medicine, Division of Physical Medicine and Rehabilitation, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Richard Henderson
- Department of Electrical and Computer Engineering, University of Washington, Seattle, WA, USA; Department of Rehabilitation Medicine, University of Washington, Seattle, WA, USA
| | - Jared Pradarelli
- Department of Rehabilitation Medicine, University of Washington, Seattle, WA, USA
| | - Sarah E Mondello
- Department of Rehabilitation Medicine, University of Washington, Seattle, WA, USA
| | - Chet T Moritz
- Department of Electrical and Computer Engineering, University of Washington, Seattle, WA, USA; Department of Rehabilitation Medicine, University of Washington, Seattle, WA, USA; Center for Neurotechnology, Seattle, WA, USA; Department of Physiology and Biophysics, University of Washington, Seattle, WA, USA.
| |
Collapse
|
20
|
Sánchez-Ventura J, Lane MA, Udina E. The Role and Modulation of Spinal Perineuronal Nets in the Healthy and Injured Spinal Cord. Front Cell Neurosci 2022; 16:893857. [PMID: 35669108 PMCID: PMC9163449 DOI: 10.3389/fncel.2022.893857] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 04/21/2022] [Indexed: 11/13/2022] Open
Abstract
Rather than being a stable scaffold, perineuronal nets (PNNs) are a dynamic and specialized extracellular matrix involved in plasticity modulation. They have been extensively studied in the brain and associated with neuroprotection, ionic buffering, and neural maturation. However, their biological function in the spinal cord and the effects of disrupting spinal PNNs remain elusive. The goal of this review is to summarize the current knowledge of spinal PNNs and their potential in pathological conditions such as traumatic spinal cord injury (SCI). We also highlighted interventions that have been used to modulate the extracellular matrix after SCI, targeting the glial scar and spinal PNNs, in an effort to promote regeneration and stabilization of the spinal circuits, respectively. These concepts are discussed in the framework of developmental and neuroplastic changes in PNNs, drawing similarities between immature and denervated neurons after an SCI, which may provide a useful context for future SCI research.
Collapse
Affiliation(s)
- Judith Sánchez-Ventura
- Department of Cell Biology, Physiology and Immunology, Institute of Neuroscience, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Bellaterra, Spain
| | - Michael A. Lane
- Department of Neurobiology and Anatomy, College of Medicine, Drexel University, Philadelphia, PA, United States
- The Marion Murray Spinal Cord Research Center, College of Medicine, Drexel University, Philadelphia, PA, United States
| | - Esther Udina
- Department of Cell Biology, Physiology and Immunology, Institute of Neuroscience, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Bellaterra, Spain
- *Correspondence: Esther Udina
| |
Collapse
|
21
|
Song W, Martin JH. Trans-Spinal Direct Current Stimulation Targets Ca 2+ Channels to Induce Persistent Motor Unit Responses. Front Neurosci 2022; 16:856948. [PMID: 35546896 PMCID: PMC9081846 DOI: 10.3389/fnins.2022.856948] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 03/04/2022] [Indexed: 01/13/2023] Open
Abstract
Trans-spinal direct current stimulation (tsDCS) is a neuromodulatory approach to augment spinal cord activity to improve function after neurological disease and injury. Little is known about the mechanisms underlying tsDCS actions on the motor system. The purpose of this study is to determine the role for a persistent inward current (PIC)-like response in motoneurons in mediating tsDCS actions. We recorded single motor units from the extensor and flexor carpi radialis muscles in healthy sedated rats and measured unit activity changes produced by cervical enlargement cathodal and anodal tsDCS (c-tsDCS; a-tsDCS). Both c-tsDCS and a-tsDCS immediately increased spontaneous motor unit firing during stimulation. After c-tsDCS was stopped, spontaneous firing persisted for a substantial period (165 ± 5s), yet after a-tsDCS activity shortly returned to baseline (27 ± 7s). Administration of the L-type calcium channel blocker Nimodipine reduced spontaneous motor unit firing during c-tsDCS and blocked the persistent response. By contrast, Nimodipine did not change unit firing during a-tsDCS but the short persistent response was blocked. Computer simulation using a two-compartment neuronal model replicated the main experimental observations: larger and more persistent responses during and after c-tsDCS than a-tsDCS. Using reduced Ca2+ conductance to model Nimodipine action, a reduced response during c-tsDCS and elimination of the persistent response was observed. Our experimental findings, supported by computer simulation, show that c-tsDCS can target Ca2+ conductances to augment motoneuron activity. As tsDCS is well-tolerated in humans, this knowledge informs therapeutic treatment strategies to achieve rehabilitation goals after injury; in particular, to increase muscle force.
Collapse
Affiliation(s)
- Weiguo Song
- Department of Molecular, Cellular, and Biomedical Sciences, Center for Discovery and Innovation, City University of New York School of Medicine, New York, NY, United States.,Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Manhasset, NY, United States
| | - John H Martin
- Department of Molecular, Cellular, and Biomedical Sciences, Center for Discovery and Innovation, City University of New York School of Medicine, New York, NY, United States.,Neuroscience Program, Graduate Center of the City University of New York, New York, NY, United States
| |
Collapse
|
22
|
Liao YH, Chen MX, Chen SC, Luo KX, Wang B, Ao LJ, Liu Y. Low-Intensity Focused Ultrasound Alleviates Spasticity and Increases Expression of the Neuronal K-Cl Cotransporter in the L4–L5 Sections of Rats Following Spinal Cord Injury. Front Cell Neurosci 2022; 16:882127. [PMID: 35634464 PMCID: PMC9133482 DOI: 10.3389/fncel.2022.882127] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 04/19/2022] [Indexed: 11/13/2022] Open
Abstract
Low-intensity focused ultrasound (LIFU) has been shown to provide effective activation of the spinal cord neurocircuits. The aim of this study was to investigate the effects of LIFU in order to alleviate spasticity following spinal cord injury (SCI) by activating the spinal neurocircuits and increasing the expression of the neuronal K-Cl cotransporter KCC2. Adult male Sprague Dawley (SD) rats (220–300 g) were randomly divided into a sham control group, a LIFU− group, and a LIFU+ group. The mechanical threshold hold (g) was used to evaluate the behavioral characteristics of spasm. Electromyography (EMG) was used to assess activation of the spinal cord neurocircuits and muscle spontaneous contraction. Spasticity was assessed by frequency-dependent depression (FDD). The expression of KCC2 of the lumbar spinal cord was determined via western blot (WB) and immunofluorescence (IF) staining. The spinal cord neurocircuits were activated by LIFU simulation, which significantly reduced the mechanical threshold (g), FDD, and EMG recordings (s) after 4 weeks of treatment. WB and IF staining both demonstrated that the expression of KCC2 was reduced in the LIFU− group (P < 0.05). After 4 weeks of LIFU stimulation, expression of KCC2 had significantly increased (P < 0.05) in the LIFU+ group compared with the LIFU− group. Thus, we hypothesized that LIFU treatment can alleviate spasticity effectively and upregulate the expression of KCC2 in the L4–L5 section of SCI rats.
Collapse
Affiliation(s)
- Ye-Hui Liao
- School of Rehabilitation, Kunming Medical University, Kunming, China
- Department of Orthopaedics, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Mo-Xian Chen
- School of Rehabilitation, Kunming Medical University, Kunming, China
| | - Shao-Chun Chen
- School of Rehabilitation, Kunming Medical University, Kunming, China
| | - Kai-Xuan Luo
- School of Rehabilitation, Kunming Medical University, Kunming, China
| | - Bing Wang
- School of Rehabilitation, Kunming Medical University, Kunming, China
| | - Li-Juan Ao
- School of Rehabilitation, Kunming Medical University, Kunming, China
- *Correspondence: Li-Juan Ao
| | - Yao Liu
- School of Rehabilitation, Kunming Medical University, Kunming, China
- Yao Liu
| |
Collapse
|
23
|
Lam P, Vinnakota C, Guzmán BCF, Newland J, Peppercorn K, Tate WP, Waldvogel HJ, Faull RLM, Kwakowsky A. Beta-Amyloid (Aβ 1-42) Increases the Expression of NKCC1 in the Mouse Hippocampus. Molecules 2022; 27:2440. [PMID: 35458638 PMCID: PMC9027496 DOI: 10.3390/molecules27082440] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/22/2022] [Accepted: 04/06/2022] [Indexed: 11/16/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder with an increasing need for developing disease-modifying treatments as current therapies only provide marginal symptomatic relief. Recent evidence suggests the γ-aminobutyric acid (GABA) neurotransmitter system undergoes remodeling in AD, disrupting the excitatory/inhibitory (E/I) balance in the brain. Altered expression levels of K-Cl-2 (KCC2) and N-K-Cl-1 (NKCC1), which are cation-chloride cotransporters (CCCs), have been implicated in disrupting GABAergic activity by regulating GABAA receptor signaling polarity in several neurological disorders, but these have not yet been explored in AD. NKCC1 and KCC2 regulate intracellular chloride [Cl-]i by accumulating and extruding Cl-, respectively. Increased NKCC1 expression in mature neurons has been reported in these disease conditions, and bumetanide, an NKCC1 inhibitor, is suggested to show potential therapeutic benefits. This study used primary mouse hippocampal neurons to explore if KCC2 and NKCC1 expression levels are altered following beta-amyloid (Aβ1-42) treatment and the potential neuroprotective effects of bumetanide. KCC2 and NKCC1 expression levels were also examined in 18-months-old male C57BL/6 mice following bilateral hippocampal Aβ1-42 stereotaxic injection. No change in KCC2 and NKCC1 expression levels were observed in mouse hippocampal neurons treated with 1 nM Aβ1-42, but NKCC1 expression increased 30-days post-Aβ1-42-injection in the CA1 region of the mouse hippocampus. Primary mouse hippocampal cultures were treated with 1 nM Aβ1-42 alone or with various concentrations of bumetanide (1 µM, 10 µM, 100 µM, 1 mM) to investigate the effect of the drug on cell viability. Aβ1-42 produced 53.1 ± 1.4% cell death after 5 days, and the addition of bumetanide did not reduce this. However, the drug at all concentrations significantly reduced cell viability, suggesting bumetanide is highly neurotoxic. In summary, these results suggest that chronic exposure to Aβ1-42 alters the balance of KCC2 and NKCC1 expression in a region-and layer-specific manner in mouse hippocampal tissue; therefore, this process most likely contributes to altered hippocampal E/I balance in this model. Furthermore, bumetanide induces hippocampal neurotoxicity, thus questioning its suitability for AD therapy. Further investigations are required to examine the effects of Aβ1-42 on KCC2 and NKCC1 expression and whether targeting CCCs might offer a therapeutic approach for AD.
Collapse
Affiliation(s)
- Patricia Lam
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand; (P.L.); (C.V.); (B.C.-F.G.); (J.N.); (H.J.W.); (R.L.M.F.)
| | - Chitra Vinnakota
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand; (P.L.); (C.V.); (B.C.-F.G.); (J.N.); (H.J.W.); (R.L.M.F.)
| | - Beatriz Calvo-Flores Guzmán
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand; (P.L.); (C.V.); (B.C.-F.G.); (J.N.); (H.J.W.); (R.L.M.F.)
| | - Julia Newland
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand; (P.L.); (C.V.); (B.C.-F.G.); (J.N.); (H.J.W.); (R.L.M.F.)
| | - Katie Peppercorn
- Department of Biochemistry, University of Otago, Dunedin 9054, New Zealand; (K.P.); (W.P.T.)
| | - Warren P. Tate
- Department of Biochemistry, University of Otago, Dunedin 9054, New Zealand; (K.P.); (W.P.T.)
| | - Henry J. Waldvogel
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand; (P.L.); (C.V.); (B.C.-F.G.); (J.N.); (H.J.W.); (R.L.M.F.)
| | - Richard L. M. Faull
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand; (P.L.); (C.V.); (B.C.-F.G.); (J.N.); (H.J.W.); (R.L.M.F.)
| | - Andrea Kwakowsky
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand; (P.L.); (C.V.); (B.C.-F.G.); (J.N.); (H.J.W.); (R.L.M.F.)
- Pharmacology and Therapeutics, Galway Neuroscience Centre, School of Medicine, National University of Ireland Galway, H91 W5P7 Galway, Ireland
| |
Collapse
|
24
|
Adapting Human-Based Transcutaneous Spinal Cord Stimulation to Develop a Clinically Relevant Animal Model. J Clin Med 2022; 11:jcm11072023. [PMID: 35407636 PMCID: PMC8999945 DOI: 10.3390/jcm11072023] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 03/30/2022] [Accepted: 03/31/2022] [Indexed: 02/01/2023] Open
Abstract
Transcutaneous spinal cord stimulation (tSCS) as a neuromodulatory strategy has received great attention as a method to promote functional recovery after spinal cord injury (SCI). However, due to the noninvasive nature of tSCS, investigations have primarily focused on human applications. This leaves a critical need for the development of a suitable animal model to further our understanding of this therapeutic intervention in terms of functional and neuroanatomical plasticity and to optimize stimulation protocols. The objective of this study is to establish a new animal model of thoracolumbar tSCS that (1) can accurately recapitulate studies in healthy humans and (2) can receive a repeated and stable tSCS treatment after SCI with minimal restraint, while the electrode remains consistently positioned. We show that our model displays bilateral evoked potentials in multisegmental leg muscles characteristically comparable to humans. Our data also suggest that tSCS mainly activates dorsal root structures like in humans, thereby accounting for the different electrode-to-body-size ratio between the two species. Finally, a repeated tSCS treatment protocol in the awake rat after a complete spinal cord transection is feasible, tolerable, and safe, even with minimal body restraint. Additionally, repeated tSCS was capable of modulating motor output after SCI, providing an avenue to further investigate stimulation-based neuroplasticity and optimize treatment.
Collapse
|
25
|
Cao T, Chen H, Huang W, Xu S, Liu P, Zou W, Pang M, Xu Y, Bai X, Liu B, Rong L, Cui ZK, Li M. hUC-MSC-mediated recovery of subacute spinal cord injury through enhancing the pivotal subunits β3 and γ2 of the GABA A receptor. Theranostics 2022; 12:3057-3078. [PMID: 35547766 PMCID: PMC9065192 DOI: 10.7150/thno.72015] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 03/12/2022] [Indexed: 12/19/2022] Open
Abstract
Rationale: Spinal cord injury (SCI) remains an incurable neurological disorder leading to permanent and profound neurologic deficits and disabilities. Human umbilical cord-derived mesenchymal stem cells (hUC-MSCs) are particularly appealing in SCI treatment to curtail damage, restore homeostasis and possible neural relay. However, the detailed mechanisms underlying hUC-MSC-mediated functional recovery of SCI have not been fully elucidated. The purpose of our current study is to identify novel therapeutic targets and depict the molecular mechanisms underlying the hUC-MSC-mediated recovery of subacute SCI. Methods: Adult female rats suffering from subacute incomplete thoracic SCI were treated with intrathecal transplantation of hUC-MSCs. The beneficial effects of hUC-MSCs on SCI repair were evaluated by a series of behavioral analyses, motor evoked potentials (MEPs) recording of hindlimb and immunohistochemistry. We carried out extensive transcriptome comparative analyses of spinal cord tissues at the lesion site from the subacute phase of SCI (sub-SCI) either treated without (+PBS) or with hUC-MSCs (+MSC) at 0 (sub-SCI), 1, 2, and 4 weeks post-transplantation (wpt), as well as normal spinal cord segments of intact/sham rats (Intact). Adeno-associated virus (AAV)-mediated neuron-specific expression system was employed to functionally screen specific γ-aminobutyric acid type A receptor (GABAAR) subunits promoting the functional recovery of SCI in vivo. The mature cortical axon scrape assay and transplantation of genetically modified MSCs with either overexpression or knockdown of brain-derived neurotrophic factor (BDNF) were employed to demonstrate that hUC-MSCs ameliorated the reduction of GABAAR subunits in the injured spinal cord via BDNF secretion in vitro and in vivo, respectively. Results: Comparative transcriptome analysis revealed the GABAergic synapse pathway is significantly enriched as a main target of hUC-MSC-activated genes in the injured spinal cord. Functional screening of the primary GABAAR subunits uncovered that Gabrb3 and Garbg2 harbored the motor and electrophysiological recovery-promoting competence. Moreover, targeting either of the two pivotal subunits β3 or γ2 in combination with/without the K+/Cl- cotransporter 2 (KCC2) reinforced the therapeutic effects. Mechanistically, BDNF secreted by hUC-MSCs contributed to the upregulation of GABAAR subunits (β3 & γ2) and KCC2 in the injured neurons. Conclusions: Our study identifies a novel mode for hUC-MSC-mediated locomotor recovery of SCI through synergistic upregulation of GABAAR β3 and γ2 along with KCC2 by BDNF secretion, indicating the significance of restoring the excitation/inhibition balance in the injured neurons for the reestablishment of neuronal circuits. This study also provides a potential combinatorial approach by targeting the pivotal subunit β3 or γ2 and KCC2, opening up possibilities for efficacious drug design.
Collapse
Affiliation(s)
- Tingting Cao
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Huan Chen
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Weiping Huang
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Sisi Xu
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Peilin Liu
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Weiwei Zou
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Mao Pang
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510515, China
- Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, 510630, China
- Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, 510630, China
| | - Ying Xu
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510515, China
- Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, 510630, China
- Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, 510630, China
| | - Xiaochun Bai
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Bin Liu
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510515, China
- Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, 510630, China
- Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, 510630, China
| | - Limin Rong
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510515, China
- Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, 510630, China
- Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, 510630, China
| | - Zhong-Kai Cui
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Mangmang Li
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| |
Collapse
|
26
|
Body Weight-Supported Treadmill Training Ameliorates Motoneuronal Hyperexcitability by Increasing GAD-65/67 and KCC2 Expression via TrkB Signaling in Rats with Incomplete Spinal Cord Injury. Neurochem Res 2022; 47:1679-1691. [PMID: 35320460 PMCID: PMC9124175 DOI: 10.1007/s11064-022-03561-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 01/29/2022] [Accepted: 02/17/2022] [Indexed: 12/11/2022]
Abstract
Spasticity is a typical consequence after spinal cord injury (SCI). The critical reasons are reducing the synthesis of Gamma-Aminobutyric Acid (GABA), glycine and potassium chloride co-transporter 2 (KCC2) inside the distal spinal cord. The current work aimed to test whether exercise training could increase the expression of glutamic acid decarboxylase 65/67 (GAD-65/67, the key enzymes in GABA synthesis) and KCC2 in the distal spinal cord via tropomyosin-related kinase B (TrkB) signaling. The experimental rats were randomly assigned to the following five groups: Sham, SCI/phosphate-buffered saline (PBS), SCI-treadmill training (TT)/PBS, SCI/TrkB-IgG, and SCI-TT/TrkB-IgG. After that, the model of T10 contusion SCI was used, then TrkB-IgG was used to prevent TrkB activity at 7 days post-SCI. Body weight-supported treadmill training started on the 8th day post-SCI for four weeks. The Hmax/Mmax ratio and the rate-dependent depression of H-reflex were used to assess the excitability of spinal motoneuronal networks. Western blotting and Immunohistochemistry techniques were utilized for measuring the expression of GAD-65, GAD-67, and KCC2. The findings revealed that exercise training could reduce motoneuronal excitability and boost GAD-65, GAD-67, and KCC2 production in the distal region of the spinal cord after SCI. The effects of exercise training were decreased after the TrkB signaling was inhibited. The present exploration demonstrated that exercise training increases GAD-65, GAD-67, and KCC2 expression in the spinal cord via TrkB signaling and that this method could also improve rats with motoneuronal hyperexcitability and spasticity induced by incomplete SCI.
Collapse
|
27
|
Mu JD, Ma LX, Zhang Z, Yu WY, Sun TY, Qian X, Tian Y, Wang JX. Acupuncture alleviates spinal hyperreflexia and motor dysfunction in post-ischemic stroke rats with spastic hypertonia via KCC2-mediated spinal GABA A activation. Exp Neurol 2022; 354:114027. [PMID: 35245503 DOI: 10.1016/j.expneurol.2022.114027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 01/18/2022] [Accepted: 02/24/2022] [Indexed: 11/17/2022]
Abstract
The majority of patients simultaneously develop motor dysfunction and spastic hypertonia after ischemic strokes, which can be associated with an increasing trend in motor impairments, seriously impeding the rehabilitation process. Evidence suggests that some deficits in the KCC2 expression in the spinal cord along with maladaptive endogenous plasticity via GABAA receptors are often involved in the pathology of spastic hypertonia after a stroke. In this respect, acupuncture has been commonly used in clinical settings for post-stroke patients' rehabilitation. Nevertheless, the mechanism of the modulating activity of this alternative medicine in the spinal pathways to relieve spasticity and improve functional recovery after a stroke has still remained unclear. Utilizing laser speckle imaging, functional assessments (viz. neurologic function scale, muscular tension scale, foot balance test, and gait analysis), H-reflex recording, TTC, Western blotting, RT-qPCR, ELISA, and immunofluorescence molecular assay, the study results illustrated that acupuncture could significantly alleviate the spinal hyperreflexia, decrease muscle tone, and enhance locomotor function by elevating the GABA, KCC2, and GABAAγ2 expressions in the lumbar spine of a rat model of post-ischemic stroke with spastic hypertonia. Furthermore, the KCC2 antagonist DIOA abolished the benefits induced by this practice. Overall, the data revealed that acupuncture is a promising therapeutic approach for spastic hypertonia after a stroke, and the positive outcomes in this sense could be achieved via activating the KCC2-mediated spinal GABAA signaling pathway.
Collapse
Affiliation(s)
- Jie-Dan Mu
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Liang-Xiao Ma
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China; The Key Unit of State Administration of Traditional Chinese Medicine, Evaluation of Characteristic Acupuncture Therapy, Beijing 100029, China.
| | - Zhou Zhang
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Wen-Yan Yu
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Tian-Yi Sun
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xu Qian
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yuan Tian
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Jun-Xiang Wang
- School of Nursing, Beijing University of Chinese Medicine, Beijing 100029, China
| |
Collapse
|
28
|
Wang S, Wang P, Yin R, Xiao M, Zhang Y, Reinhardt JD, Wang H, Xu G. Combination of repetitive transcranial magnetic stimulation and treadmill training reduces hyperreflexia by rebalancing motoneuron excitability in rats after spinal cord contusion. Neurosci Lett 2022; 775:136536. [PMID: 35183693 DOI: 10.1016/j.neulet.2022.136536] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 02/12/2022] [Accepted: 02/15/2022] [Indexed: 11/19/2022]
Abstract
Spasticity commonly emerges during the process of recovery after spinal cord injury (SCI) and critically exacerbates motor dysfunction. Given insufficient effects of individual therapies, we combined repetitive transcranial magnetic stimulation (rTMS) with treadmill training (Tr) in rats with SCI to investigate potential synergistic effects on alleviating spasticity and motor dysfunction. Animals were randomized into four groups: SCI only, rTMS, Tr, and rTMS plus Tr. At the study endpoint eight weeks after the start of interventions, the rTMS plus Tr group exhibited the largest decrease in maximal H-reflex amplitude/maximal M-wave amplitude ratio (effect size (ES): -0.082, 95% confidence interval (CI): -0.118 to -0.046, p < 0.001) as well as the greatest improvement in motor function measured with the Basso, Beattie, and Bresnahan locomotor scale (ES: 1.811, 95% CI: 1.018 to 2.603, p < 0.001; significantly different from all other groups at p < 0.01) and grid-walking test (ES: -5.1, 95% CI: -7.784 to -2.416, p < 0.001, significantly different from rTMS alone at p < 0.01). Pathological analyses demonstrated that the combined treatment facilitated the growth of serotonergic axons around the lesion site, and the upregulation of 5-hydroxytryptamine, potassium-chloride cotransporter-2, and glutamic acid decarboxylases 67 in the lumbar spinal cord distal to the injury site. All effects of combined treatment of rTMS and treadmill training were enhanced compared to treadmill training or rTMS alone. Treadmill training and rTMS intervention appear to have synergistic effects on hyperreflexia and locomotion likely related to a restored balance between facilitatory and inhibitory inputs to motoneurons.
Collapse
Affiliation(s)
- Shuangyan Wang
- Department of Rehabilitation Medicine, Zhongda Hospital, Southeast University Nanjing 210024, China; Nanjing Medical University, Center of Rehabilitation Medicine, 1st affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Pei Wang
- Department of Rehabilitation Medicine, Zhongda Hospital, Southeast University Nanjing 210024, China
| | - Ruian Yin
- Department of Rehabilitation Medicine, Zhongda Hospital, Southeast University Nanjing 210024, China
| | - Ming Xiao
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing 211166, China
| | - Yongjie Zhang
- Department of Human Anatomy, Nanjing Medical University, Nanjing 211166, China
| | - Jan D Reinhardt
- Institute for Disaster Management and Reconstruction, Sichuan University, Chengdu, China
| | - Hongxing Wang
- Department of Rehabilitation Medicine, Zhongda Hospital, Southeast University Nanjing 210024, China.
| | - Guangxu Xu
- Nanjing Medical University, Center of Rehabilitation Medicine, 1st affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.
| |
Collapse
|
29
|
Fauss GNK, Hudson KE, Grau JW. Role of Descending Serotonergic Fibers in the Development of Pathophysiology after Spinal Cord Injury (SCI): Contribution to Chronic Pain, Spasticity, and Autonomic Dysreflexia. BIOLOGY 2022; 11:234. [PMID: 35205100 PMCID: PMC8869318 DOI: 10.3390/biology11020234] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/27/2022] [Accepted: 01/29/2022] [Indexed: 12/12/2022]
Abstract
As the nervous system develops, nerve fibers from the brain form descending tracts that regulate the execution of motor behavior within the spinal cord, incoming sensory signals, and capacity to change (plasticity). How these fibers affect function depends upon the transmitter released, the receptor system engaged, and the pattern of neural innervation. The current review focuses upon the neurotransmitter serotonin (5-HT) and its capacity to dampen (inhibit) neural excitation. A brief review of key anatomical details, receptor types, and pharmacology is provided. The paper then considers how damage to descending serotonergic fibers contributes to pathophysiology after spinal cord injury (SCI). The loss of serotonergic fibers removes an inhibitory brake that enables plasticity and neural excitation. In this state, noxious stimulation can induce a form of over-excitation that sensitizes pain (nociceptive) circuits, a modification that can contribute to the development of chronic pain. Over time, the loss of serotonergic fibers allows prolonged motor drive (spasticity) to develop and removes a regulatory brake on autonomic function, which enables bouts of unregulated sympathetic activity (autonomic dysreflexia). Recent research has shown that the loss of descending serotonergic activity is accompanied by a shift in how the neurotransmitter GABA affects neural activity, reducing its inhibitory effect. Treatments that target the loss of inhibition could have therapeutic benefit.
Collapse
Affiliation(s)
| | | | - James W. Grau
- Department of Psychological and Brain Sciences, Texas A&M University, College Station, TX 77843, USA; (G.N.K.F.); (K.E.H.)
| |
Collapse
|
30
|
Pulverenti TS, Zaaya M, Grabowski M, Grabowski E, Islam MA, Li J, Murray LM, Knikou M. Neurophysiological Changes After Paired Brain and Spinal Cord Stimulation Coupled With Locomotor Training in Human Spinal Cord Injury. Front Neurol 2021; 12:627975. [PMID: 34040572 PMCID: PMC8141587 DOI: 10.3389/fneur.2021.627975] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 04/01/2021] [Indexed: 11/13/2022] Open
Abstract
Neurophysiological changes that involve activity-dependent neuroplasticity mechanisms via repeated stimulation and locomotor training are not commonly employed in research even though combination of interventions is a common clinical practice. In this randomized clinical trial, we established neurophysiological changes when transcranial magnetic stimulation (TMS) of the motor cortex was paired with transcutaneous thoracolumbar spinal (transspinal) stimulation in human spinal cord injury (SCI) delivered during locomotor training. We hypothesized that TMS delivered before transspinal (TMS-transspinal) stimulation promotes functional reorganization of spinal networks during stepping. In this protocol, TMS-induced corticospinal volleys arrive at the spinal cord at a sufficient time to interact with transspinal stimulation induced depolarization of alpha motoneurons over multiple spinal segments. We further hypothesized that TMS delivered after transspinal (transspinal-TMS) stimulation induces less pronounced effects. In this protocol, transspinal stimulation is delivered at time that allows transspinal stimulation induced action potentials to arrive at the motor cortex and affect descending motor volleys at the site of their origin. Fourteen individuals with motor incomplete and complete SCI participated in at least 25 sessions. Both stimulation protocols were delivered during the stance phase of the less impaired leg. Each training session consisted of 240 paired stimuli delivered over 10-min blocks. In transspinal-TMS, the left soleus H-reflex increased during the stance-phase and the right soleus H-reflex decreased at mid-swing. In TMS-transspinal no significant changes were found. When soleus H-reflexes were grouped based on the TMS-targeted limb, transspinal-TMS and locomotor training promoted H-reflex depression at swing phase, while TMS-transspinal and locomotor training resulted in facilitation of the soleus H-reflex at stance phase of the step cycle. Furthermore, both transspinal-TMS and TMS-transspinal paired-associative stimulation (PAS) and locomotor training promoted a more physiological modulation of motor activity and thus depolarization of motoneurons during assisted stepping. Our findings support that targeted non-invasive stimulation of corticospinal and spinal neuronal pathways coupled with locomotor training produce neurophysiological changes beneficial to stepping in humans with varying deficits of sensorimotor function after SCI.
Collapse
Affiliation(s)
- Timothy S Pulverenti
- Klab4Recovery Research Laboratory, Department of Physical Therapy, College of Staten Island, The City University of New York, New York, NY, United States
| | - Morad Zaaya
- Klab4Recovery Research Laboratory, Department of Physical Therapy, College of Staten Island, The City University of New York, New York, NY, United States
| | - Monika Grabowski
- Klab4Recovery Research Laboratory, Department of Physical Therapy, College of Staten Island, The City University of New York, New York, NY, United States
| | - Ewelina Grabowski
- Klab4Recovery Research Laboratory, Department of Physical Therapy, College of Staten Island, The City University of New York, New York, NY, United States
| | - Md Anamul Islam
- Klab4Recovery Research Laboratory, Department of Physical Therapy, College of Staten Island, The City University of New York, New York, NY, United States
| | - Jeffrey Li
- Klab4Recovery Research Laboratory, Department of Physical Therapy, College of Staten Island, The City University of New York, New York, NY, United States
| | - Lynda M Murray
- Klab4Recovery Research Laboratory, Department of Physical Therapy, College of Staten Island, The City University of New York, New York, NY, United States
| | - Maria Knikou
- Klab4Recovery Research Laboratory, Department of Physical Therapy, College of Staten Island, The City University of New York, New York, NY, United States.,Ph.D. Program in Biology and Collaborative Neuroscience Program, Graduate Center of the City University of New York and College of Staten Island, New York, NY, United States
| |
Collapse
|
31
|
Bilchak JN, Caron G, Côté MP. Exercise-Induced Plasticity in Signaling Pathways Involved in Motor Recovery after Spinal Cord Injury. Int J Mol Sci 2021; 22:ijms22094858. [PMID: 34064332 PMCID: PMC8124911 DOI: 10.3390/ijms22094858] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 04/28/2021] [Accepted: 04/29/2021] [Indexed: 02/06/2023] Open
Abstract
Spinal cord injury (SCI) leads to numerous chronic and debilitating functional deficits that greatly affect quality of life. While many pharmacological interventions have been explored, the current unsurpassed therapy for most SCI sequalae is exercise. Exercise has an expansive influence on peripheral health and function, and by activating the relevant neural pathways, exercise also ameliorates numerous disorders of the central nervous system (CNS). While the exact mechanisms by which this occurs are still being delineated, major strides have been made in the past decade to understand the molecular underpinnings of this essential treatment. Exercise rapidly and prominently affects dendritic sprouting, synaptic connections, neurotransmitter production and regulation, and ionic homeostasis, with recent literature implicating an exercise-induced increase in neurotrophins as the cornerstone that binds many of these effects together. The field encompasses vast complexity, and as the data accumulate, disentangling these molecular pathways and how they interact will facilitate the optimization of intervention strategies and improve quality of life for individuals affected by SCI. This review describes the known molecular effects of exercise and how they alter the CNS to pacify the injury environment, increase neuronal survival and regeneration, restore normal neural excitability, create new functional circuits, and ultimately improve motor function following SCI.
Collapse
|
32
|
Wang JX, Ma LX, Mu JD, Sun TY, Qian X, Yu WY, Tian Y, Zhang Z. Anti-spastic effect induced by waggle needling correlates with KCC2-GABA A pathway in post-stroke spasticity rats. Neurosci Lett 2021; 750:135810. [PMID: 33705929 DOI: 10.1016/j.neulet.2021.135810] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 03/02/2021] [Accepted: 03/03/2021] [Indexed: 12/30/2022]
Abstract
Although clinical efficacy of waggle needling has been confirmed, therapeutic mechanisms still remain poorly understood. Reduction of GABA was involved in the etiology of spasticity. Recently, accumulated evidences suggest that the inhibitory effect of GABA is determined by low intracellular chloride concentration, which is predominantly mediated by KCC2. This study was designed to investigate whether KCC2-GABAA pathway was involved in the mechanism underlying acupuncture intervention in rats with middle cerebral artery occlusion (MCAO). Three days after modeling, the rats received waggle needling, routine needling and placebo needling for 7 consecutive days. After treatment, the muscle spasticity, motor function and infarct volumes were tested. KCC2 and GABAAγ2 levels were detected via western blotting, RT-PCR and immunofluorescence. KCC2 antagonist and agonist were administered after the last intervention. We found that acupuncture, particularly waggle needling, could remarkably alleviate muscle spasticity, reverse motor deficits and reduce cerebral infraction in MCAO rats, possibly due to its effects on up-regulating expressions of KCC2 and GABAAγ2 in the cortical infarct regions. However, the effects were blocked by KCC2 antagonist. In summary, this study suggests that improvements in muscle spasticity and motor function induced by waggle needling correlates with the activation of KCC2-GABAA pathway.
Collapse
Affiliation(s)
- Jun-Xiang Wang
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Liang-Xiao Ma
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China.
| | - Jie-Dan Mu
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Tian-Yi Sun
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Xu Qian
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Wen-Yan Yu
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Yuan Tian
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Zhou Zhang
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
33
|
GABAergic Mechanisms Can Redress the Tilted Balance between Excitation and Inhibition in Damaged Spinal Networks. Mol Neurobiol 2021; 58:3769-3786. [PMID: 33826070 PMCID: PMC8279998 DOI: 10.1007/s12035-021-02370-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Accepted: 03/22/2021] [Indexed: 12/19/2022]
Abstract
Correct operation of neuronal networks depends on the interplay between synaptic excitation and inhibition processes leading to a dynamic state termed balanced network. In the spinal cord, balanced network activity is fundamental for the expression of locomotor patterns necessary for rhythmic activation of limb extensor and flexor muscles. After spinal cord lesion, paralysis ensues often followed by spasticity. These conditions imply that, below the damaged site, the state of balanced networks has been disrupted and that restoration might be attempted by modulating the excitability of sublesional spinal neurons. Because of the widespread expression of inhibitory GABAergic neurons in the spinal cord, their role in the early and late phases of spinal cord injury deserves full attention. Thus, an early surge in extracellular GABA might be involved in the onset of spinal shock while a relative deficit of GABAergic mechanisms may be a contributor to spasticity. We discuss the role of GABA A receptors at synaptic and extrasynaptic level to modulate network excitability and to offer a pharmacological target for symptom control. In particular, it is proposed that activation of GABA A receptors with synthetic GABA agonists may downregulate motoneuron hyperexcitability (due to enhanced persistent ionic currents) and, therefore, diminish spasticity. This approach might constitute a complementary strategy to regulate network excitability after injury so that reconstruction of damaged spinal networks with new materials or cell transplants might proceed more successfully.
Collapse
|
34
|
Bilchak JN, Yeakle K, Caron G, Malloy D, Côté MP. Enhancing KCC2 activity decreases hyperreflexia and spasticity after chronic spinal cord injury. Exp Neurol 2021; 338:113605. [PMID: 33453210 PMCID: PMC7904648 DOI: 10.1016/j.expneurol.2021.113605] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/21/2020] [Accepted: 01/09/2021] [Indexed: 02/03/2023]
Abstract
After spinal cord injury (SCI), the majority of individuals develop spasticity, a debilitating condition involving involuntary movements, co-contraction of antagonistic muscles, and hyperreflexia. By acting on GABAergic and Ca2+-dependent signaling, current anti-spastic medications lead to serious side effects, including a drastic decrease in motoneuronal excitability which impairs motor function and rehabilitation efforts. Exercise, in contrast, decreases spastic symptoms without decreasing motoneuron excitability. These functional improvements coincide with an increase in expression of the chloride co-transporter KCC2 in lumbar motoneurons. Thus, we hypothesized that spastic symptoms can be alleviated directly through restoration of chloride homeostasis and endogenous inhibition by increasing KCC2 activity. Here, we used the recently developed KCC2 enhancer, CLP257, to evaluate the effects of acutely increasing KCC2 extrusion capability on spastic symptoms after chronic SCI. Sprague Dawley rats received a spinal cord transection at T12 and were either bike-trained or remained sedentary for 5 weeks. Increasing KCC2 activity in the lumbar enlargement improved the rate-dependent depression of the H-reflex and reduced both phasic and tonic EMG responses to muscle stretch in sedentary animals after chronic SCI. Furthermore, the improvements due to this pharmacological treatment mirror those of exercise. Together, our results suggest that pharmacologically increasing KCC2 activity is a promising approach to decrease spastic symptoms in individuals with SCI. By acting to directly restore endogenous inhibition, this strategy has potential to avoid severe side effects and improve the quality of life of affected individuals.
Collapse
Affiliation(s)
- Jadwiga N Bilchak
- Marion Murray Spinal Cord Injury Research Center, Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, United States of America
| | - Kyle Yeakle
- Marion Murray Spinal Cord Injury Research Center, Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, United States of America
| | - Guillaume Caron
- Marion Murray Spinal Cord Injury Research Center, Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, United States of America
| | - Dillon Malloy
- Marion Murray Spinal Cord Injury Research Center, Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, United States of America
| | - Marie-Pascale Côté
- Marion Murray Spinal Cord Injury Research Center, Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, United States of America.
| |
Collapse
|
35
|
Jeffrey-Gauthier R, Bouyer J, Piché M, Côté MP, Leblond H. Locomotor deficits induced by lumbar muscle inflammation involve spinal microglia and are independent of KCC2 expression in a mouse model of complete spinal transection. Exp Neurol 2021; 338:113592. [PMID: 33388315 PMCID: PMC7904639 DOI: 10.1016/j.expneurol.2020.113592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 12/03/2020] [Accepted: 12/28/2020] [Indexed: 10/22/2022]
Abstract
Spinal cord injury (SCI) is associated with damage to musculoskeletal tissues of the spine. Recent findings show that pain and inflammatory processes caused by musculoskeletal injury mediate plastic changes in the spinal cord. These changes could impede the adaptive plastic changes responsible for functional recovery. The underlying mechanism remains unclear, but may involve the microglia-BDNF-KCC2 pathway, which is implicated in sensitization of dorsal horn neurons in neuropathic pain and in the regulation of spinal excitability by step-training. In the present study, we examined the effects of step-training and lumbar muscle inflammation induced by complete Freund's adjuvant (CFA) on treadmill locomotion in a mouse model of complete spinal transection. The impact on locomotor recovery of each of these interventions alone or in combination were examined in addition to changes in microglia and KCC2 expression in the dorsal and ventral horns of the sublesional spinal cord. Results show that angular motion at the hip, knee and ankle joint during locomotion were decreased by CFA injection and improved by step-training. Moreover, CFA injection enhanced the expression of the microglial marker Iba1 in both ventral and dorsal horns, with or without step-training. However, this change was not associated with a modulation of KCC2 expression, suggesting that locomotor deficits induced by inflammation are independent of KCC2 expression in the sublesional spinal cord. These results indicate that musculoskeletal injury hinders locomotor recovery after SCI and that microglia is involved in this effect.
Collapse
Affiliation(s)
- Renaud Jeffrey-Gauthier
- Department of Anatomy, Université du Québec à Trois-Rivières, 3351 boul. des Forges, C.P. 500, Trois-Rivières, QC G9A 5H7, Canada; CogNAC Research Group, Université du Québec à Trois-Rivières, 3351 boul. des Forges, C.P. 500, Trois-Rivières, QC G9A 5H7, Canada.
| | - Julien Bouyer
- Marion Murray Spinal Cord Research Center, Department of Neurobiology and Anatomy, Drexel University, Philadelphia, PA 19129, United States.
| | - Mathieu Piché
- Department of Anatomy, Université du Québec à Trois-Rivières, 3351 boul. des Forges, C.P. 500, Trois-Rivières, QC G9A 5H7, Canada; CogNAC Research Group, Université du Québec à Trois-Rivières, 3351 boul. des Forges, C.P. 500, Trois-Rivières, QC G9A 5H7, Canada.
| | - Marie-Pascale Côté
- Marion Murray Spinal Cord Research Center, Department of Neurobiology and Anatomy, Drexel University, Philadelphia, PA 19129, United States.
| | - Hugues Leblond
- Department of Anatomy, Université du Québec à Trois-Rivières, 3351 boul. des Forges, C.P. 500, Trois-Rivières, QC G9A 5H7, Canada; CogNAC Research Group, Université du Québec à Trois-Rivières, 3351 boul. des Forges, C.P. 500, Trois-Rivières, QC G9A 5H7, Canada.
| |
Collapse
|
36
|
Early escitalopram administration as a preemptive treatment strategy against spasticity after contusive spinal cord injury in rats. Sci Rep 2021; 11:7120. [PMID: 33782426 PMCID: PMC8007832 DOI: 10.1038/s41598-021-85961-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 02/23/2021] [Indexed: 11/29/2022] Open
Abstract
In the majority of spinal cord injury (SCI) patients, spasticity develops in the subacute phase and chronically persists with muscle hypertonia. Among various pathological conditions underlying spasticity, upregulated expression of 5-HT receptors (5-HTR) on the spinal motor neurons due to 5-HT denervation is considered one of crucial factors for hyperexcitability of the spinal circuit. As a 5-HT signal modulator, selective serotonin re-uptake inhibitors (SSRIs) are ordinarily prescribed for diseases associated with 5-HT in the CNS, and are known for their ability to increase 5-HT levels as well as to desensitize 5-HTR. Here, we hypothesized that early SSRI administration as a preemptive treatment strategy would effectively prevent the onset of spasticity. We used a rat model of contusive SCI and administered escitalopram during the first 4 weeks after injury, which is the period required for spasticity development in rodent models. We performed a swimming test to quantify spastic behaviors and conducted the Hoffman reflex test as well as histological analyses for 5-HT2AR and KCC2 expressions. Four weeks of escitalopram administration suppressed spastic behaviors during the swimming test and reduced the population of spasticity-strong rats. Moreover, the treatment resulted in decreased immunoreactivity of 5-HT2AR in the spinal motor neurons. Result of the H-reflex test and membrane expression of KCC2 were not significantly altered. In summary, early escitalopram administration could prevent the onset of spastic behaviors via regulation of 5-HT system after SCI, but could not modulate exaggerated spinal reflex. Our results suggest a novel application of SSRIs for preventative treatment of spasticity.
Collapse
|
37
|
Sánchez-Ventura J, Giménez-Llort L, Penas C, Udina E. Voluntary wheel running preserves lumbar perineuronal nets, enhances motor functions and prevents hyperreflexia after spinal cord injury. Exp Neurol 2020; 336:113533. [PMID: 33264633 DOI: 10.1016/j.expneurol.2020.113533] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 11/02/2020] [Accepted: 11/19/2020] [Indexed: 11/16/2022]
Abstract
Perineuronal nets (PNN) are a promising candidate to harness neural plasticity since their activity-dependent modulation allows to either stabilize the circuits or increase plasticity. Modulation of plasticity is the basis of rehabilitation strategies to reduce maladaptive plasticity after spinal cord injuries (SCI). Hence, it is important to understand how spinal PNN are affected after SCI and rehabilitation. Thus, this work aims to describe functional and PNN changes after thoracic SCI in mice, followed by different activity-dependent therapies: enriched environment, voluntary wheel and forced treadmill running. We found that the contusion provoked thermal hyperalgesia, hyperreflexia and locomotor impairment as measured by thermal plantar test, H wave recordings and the BMS score of locomotion, respectively. In the spinal cord, SCI reduced PNN density around lumbar motoneurons. In contrast, activity-based therapies increased motoneuron activity and reversed PNN decrease. The voluntary wheel group showed full preservation of PNN which also correlated with reduced hyperreflexia and better locomotor recovery. Furthermore, both voluntary wheel and treadmill running reduced hyperalgesia, but this finding was independent of lumbar PNN levels. In the brainstem sensory nuclei, SCI did not modify PNN whereas some activity-based therapies reduced them. The results of the present study highlight the impact of SCI on decreasing PNN at caudal segments of the spinal cord and the potential of physical activity-based therapies to reverse PNN disaggregation and to improve functional recovery. As modulating plasticity is crucial for restoring damaged neural circuits, regulating PNN by activity is an encouraging target to improve the outcome after injury.
Collapse
Affiliation(s)
- J Sánchez-Ventura
- Institute of Neurosciences, Department Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Bellaterra, Spain
| | - L Giménez-Llort
- Institute of Neurosciences, Department of Psychiatry and Forensic Medicine, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - C Penas
- Institute of Neurosciences, Department Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Bellaterra, Spain
| | - E Udina
- Institute of Neurosciences, Department Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Bellaterra, Spain.
| |
Collapse
|
38
|
Environmental regulation of the chloride transporter KCC2: switching inflammation off to switch the GABA on? Transl Psychiatry 2020; 10:349. [PMID: 33060559 PMCID: PMC7562743 DOI: 10.1038/s41398-020-01027-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 09/08/2020] [Accepted: 09/14/2020] [Indexed: 12/15/2022] Open
Abstract
Chloride homeostasis, the main determinant factor for the dynamic tuning of GABAergic inhibition during development, has emerged as a key element altered in a wide variety of brain disorders. Accordingly, developmental disorders such as schizophrenia, Autism Spectrum Disorder, Down syndrome, epilepsy, and tuberous sclerosis complex (TSC) have been associated with alterations in the expression of genes codifying for either of the two cotransporters involved in the excitatory-to-inhibitory GABA switch, KCC2 and NKCC1. These alterations can result from environmental insults, including prenatal stress and maternal separation which share, as common molecular denominator, the elevation of pro-inflammatory cytokines. In this review we report and systemize recent research articles indicating that different perinatal environmental perturbations affect the expression of chloride transporters, delaying the developmental switch of GABA signaling, and that inflammatory cytokines, in particular interleukin 1β, may represent a key causal factor for this phenomenon. Based on literature data, we provide therefore a unifying conceptual framework, linking environmental hits with the excitatory-to-inhibitory GABA switch in the context of brain developmental disorders.
Collapse
|
39
|
Caron G, Bilchak JN, Côté MP. Direct evidence for decreased presynaptic inhibition evoked by PBSt group I muscle afferents after chronic SCI and recovery with step-training in rats. J Physiol 2020; 598:4621-4642. [PMID: 32721039 DOI: 10.1113/jp280070] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 07/27/2020] [Indexed: 01/11/2023] Open
Abstract
KEY POINTS Presynaptic inhibition is modulated by supraspinal centres and primary afferents in order to filter sensory information, adjust spinal reflex excitability, and ensure smooth movement. After spinal cord injury (SCI), the supraspinal control of primary afferent depolarization (PAD) interneurons is disengaged, suggesting an increased role for sensory afferents. While increased H-reflex excitability in spastic individuals indicates a possible decrease in presynaptic inhibition, it remains unclear whether a decrease in sensory-evoked PAD contributes to this effect. We investigated whether the PAD evoked by hindlimb afferents contributes to the change in presynaptic inhibition of the H-reflex in a decerebrated rat preparation. We found that chronic SCI decreases presynaptic inhibition of the plantar H-reflex through a reduction in PAD evoked by posterior biceps-semitendinosus (PBSt) muscle group I afferents. We further found that step-training restored presynaptic inhibition of the plantar H-reflex evoked by PBSt, suggesting the presence of activity-dependent plasticity of PAD pathways activated by flexor muscle group I afferents. ABSTRACT Spinal cord injury (SCI) results in the disruption of supraspinal control of spinal networks and an increase in the relative influence of afferent feedback to sublesional neural networks, both of which contribute to enhancing spinal reflex excitability. Hyperreflexia occurs in ∼75% of individuals with a chronic SCI and critically hinders functional recovery and quality of life. It is suggested that it results from an increase in motoneuronal excitability and a decrease in presynaptic and postsynaptic inhibitory mechanisms. In contrast, locomotor training decreases hyperreflexia by restoring presynaptic inhibition. Primary afferent depolarization (PAD) is a powerful presynaptic inhibitory mechanism that selectively gates primary afferent transmission to spinal neurons to adjust reflex excitability and ensure smooth movement. However, the effect of chronic SCI and step-training on the reorganization of presynaptic inhibition evoked by hindlimb afferents, and the contribution of PAD has never been demonstrated. The objective of this study is to directly measure changes in presynaptic inhibition through dorsal root potentials (DRPs) and its association with plantar H-reflex inhibition. We provide direct evidence that H-reflex hyperexcitability is associated with a decrease in transmission of PAD pathways activated by posterior biceps-semitendinosus (PBSt) afferents after chronic SCI. More precisely, we illustrate that the pattern of inhibition evoked by PBSt group I muscle afferents onto both L4-DRPs and plantar H-reflexes evoked by the distal tibial nerve is impaired after chronic SCI. These changes are not observed in step-trained animals, suggesting a role for activity-dependent plasticity to regulate PAD pathways activated by flexor muscle group I afferents.
Collapse
Affiliation(s)
- Guillaume Caron
- Marion Murray Spinal Cord Research Center, Department of Neurobiology and Anatomy, Drexel University, Philadelphia, PA, 19129
| | - Jadwiga N Bilchak
- Marion Murray Spinal Cord Research Center, Department of Neurobiology and Anatomy, Drexel University, Philadelphia, PA, 19129
| | - Marie-Pascale Côté
- Marion Murray Spinal Cord Research Center, Department of Neurobiology and Anatomy, Drexel University, Philadelphia, PA, 19129
| |
Collapse
|
40
|
Grau JW, Baine RE, Bean PA, Davis JA, Fauss GN, Henwood MK, Hudson KE, Johnston DT, Tarbet MM, Strain MM. Learning to promote recovery after spinal cord injury. Exp Neurol 2020; 330:113334. [PMID: 32353465 PMCID: PMC7282951 DOI: 10.1016/j.expneurol.2020.113334] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 04/19/2020] [Accepted: 04/26/2020] [Indexed: 02/06/2023]
Abstract
The present review explores the concept of learning within the context of neurorehabilitation after spinal cord injury (SCI). The aim of physical therapy and neurorehabilitation is to bring about a lasting change in function-to encourage learning. Traditionally, it was assumed that the adult spinal cord is hardwired-immutable and incapable of learning. Research has shown that neurons within the lower (lumbosacral) spinal cord can support learning after communication with the brain has been disrupted by means of a thoracic transection. Noxious stimulation can sensitize nociceptive circuits within the spinal cord, engaging signal pathways analogous to those implicated in brain-dependent learning and memory. After a spinal contusion injury, pain input can fuel hemorrhage, increase the area of tissue loss (secondary injury), and undermine long-term recovery. Neurons within the spinal cord are sensitive to environmental relations. This learning has a metaplastic effect that counters neural over-excitation and promotes adaptive learning through an up-regulation of brain-derived neurotrophic factor (BDNF). Exposure to rhythmic stimulation, treadmill training, and cycling also enhances the expression of BDNF and counters the development of nociceptive sensitization. SCI appears to enable plastic potential within the spinal cord by down-regulating the Cl- co-transporter KCC2, which reduces GABAergic inhibition. This enables learning, but also fuels over-excitation and nociceptive sensitization. Pairing epidural stimulation with activation of motor pathways also promotes recovery after SCI. Stimulating motoneurons in response to activity within the motor cortex, or a targeted muscle, has a similar effect. It is suggested that a neurofunctionalist approach can foster the discovery of processes that impact spinal function and how they may be harnessed to foster recovery after SCI.
Collapse
Affiliation(s)
- James W Grau
- Behavioral and Cellular Neuroscience, Department of Psychology, Texas A&M University, College Station, TX 77843, USA.
| | - Rachel E Baine
- Behavioral and Cellular Neuroscience, Department of Psychology, Texas A&M University, College Station, TX 77843, USA
| | - Paris A Bean
- Behavioral and Cellular Neuroscience, Department of Psychology, Texas A&M University, College Station, TX 77843, USA
| | - Jacob A Davis
- Behavioral and Cellular Neuroscience, Department of Psychology, Texas A&M University, College Station, TX 77843, USA
| | - Gizelle N Fauss
- Behavioral and Cellular Neuroscience, Department of Psychology, Texas A&M University, College Station, TX 77843, USA
| | - Melissa K Henwood
- Behavioral and Cellular Neuroscience, Department of Psychology, Texas A&M University, College Station, TX 77843, USA
| | - Kelsey E Hudson
- Behavioral and Cellular Neuroscience, Department of Psychology, Texas A&M University, College Station, TX 77843, USA
| | - David T Johnston
- Behavioral and Cellular Neuroscience, Department of Psychology, Texas A&M University, College Station, TX 77843, USA
| | - Megan M Tarbet
- Behavioral and Cellular Neuroscience, Department of Psychology, Texas A&M University, College Station, TX 77843, USA
| | - Misty M Strain
- Battlefield Pain Research, U.S. Army Institute of Surgical Research, 3698 Chambers Pass, BHT-1, BSA Fort Sam Houston, TX 78234, USA
| |
Collapse
|
41
|
Bras H, Liabeuf S. Differential effects of spinal cord transection on glycinergic and GABAergic synaptic signaling in sub-lesional lumbar motoneurons. J Chem Neuroanat 2020; 113:101847. [PMID: 32653413 DOI: 10.1016/j.jchemneu.2020.101847] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 07/01/2020] [Accepted: 07/01/2020] [Indexed: 01/11/2023]
Abstract
This review takes stock on the impact of complete spinal cord transection (SCT) on the plasticity of inhibitory synaptic transmission on sub-lesional lumbar motoneurons (MNs), differentiating between studies in neonate and adult rats. After neonatal SCT, normal maturational up-regulation of glycine receptors was observed. On the other hand, the developmental downregulation of the GABAA receptors, as well as the up-regulation of the co-transporter KCC2 were prevented, but not the normal decrease of NKCC1. In adult SCT rats, glycinergic synaptic transmission, which is the major contributor to spinal MNs inhibition in adulthood, had normal control levels 2 months post-injury. On the other hand, the GABAergic transmission was altered through an up-regulation of the pre-signaling levels and a down-regulation in the density of post synaptic receptors. KCC2 membrane expression was down-regulated at all post-injury times tested (24h to 4 months), thereby depolarizing the Cl- equilibrium potential and reducing the strength of postsynaptic inhibition. The preservation of glycinergic pre- and post signaling is probably a key factor in the success of locomotor rehabilitation programs in adult SCT rats. However, these data highlight the need to develop strategies to restore KCC2 levels in lumbar MNs, to stabilize the excitation/inhibition balance, which is essential to the effective control of skeletal muscle activity.
Collapse
Affiliation(s)
- Hélène Bras
- Institut de Neurosciences de la Timone, UMR 7289, CNRS and Aix Marseille Université, Campus Santé Timone, 13385, Marseille, France.
| | - Sylvie Liabeuf
- Institut de Neurosciences de la Timone, UMR 7289, CNRS and Aix Marseille Université, Campus Santé Timone, 13385, Marseille, France
| |
Collapse
|
42
|
Horie M, Yoshioka N, Kusumi S, Sano H, Kurose M, Watanabe‐Iida I, Hossain I, Chiken S, Abe M, Yamamura K, Sakimura K, Nambu A, Shibata M, Takebayashi H. Disruption of
dystonin
in Schwann cells results in late‐onset neuropathy and sensory ataxia. Glia 2020; 68:2330-2344. [DOI: 10.1002/glia.23843] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 04/26/2020] [Accepted: 04/28/2020] [Indexed: 01/26/2023]
Affiliation(s)
- Masao Horie
- Division of Neurobiology and AnatomyGraduate School of Medical and Dental Sciences, Niigata University Niigata Japan
- Department of Morphological SciencesKagoshima University Kagoshima Japan
- Department of NursingNiigata College of Nursing Niigata Japan
| | - Nozomu Yoshioka
- Division of Neurobiology and AnatomyGraduate School of Medical and Dental Sciences, Niigata University Niigata Japan
| | - Satoshi Kusumi
- Department of Morphological SciencesKagoshima University Kagoshima Japan
| | - Hiromi Sano
- Division of System NeurophysiologyNational Institute for Physiological Sciences Okazaki Japan
- Department of Physiological SciencesSOKENDAI Okazaki Japan
| | - Masayuki Kurose
- Division of Oral PhysiologyGraduate School of Medical and Dental Sciences, Niigata University Niigata Japan
- Department of Physiology, School of Dentistry, Iwate Medical University Morioka Japan
| | - Izumi Watanabe‐Iida
- Department of Cellular NeurobiologyBrain Research Institute, Niigata University Niigata Japan
- Division of Oral Biochemistry, Graduate School of Medical and Dental Sciences, Niigata University Niigata Japan
| | - Ibrahim Hossain
- Division of Neurobiology and AnatomyGraduate School of Medical and Dental Sciences, Niigata University Niigata Japan
- Department of Biochemistry and Molecular BiologyJahangirnagar University Savar Dhaka Bangladesh
| | - Satomi Chiken
- Division of System NeurophysiologyNational Institute for Physiological Sciences Okazaki Japan
- Department of Physiological SciencesSOKENDAI Okazaki Japan
| | - Manabu Abe
- Department of Cellular NeurobiologyBrain Research Institute, Niigata University Niigata Japan
- Department of Animal Model DevelopmentBrain Research Institute, Niigata University Niigata Japan
| | - Kensuke Yamamura
- Division of Oral PhysiologyGraduate School of Medical and Dental Sciences, Niigata University Niigata Japan
| | - Kenji Sakimura
- Department of Cellular NeurobiologyBrain Research Institute, Niigata University Niigata Japan
- Department of Animal Model DevelopmentBrain Research Institute, Niigata University Niigata Japan
| | - Atsushi Nambu
- Division of System NeurophysiologyNational Institute for Physiological Sciences Okazaki Japan
- Department of Physiological SciencesSOKENDAI Okazaki Japan
| | - Masahiro Shibata
- Department of Morphological SciencesKagoshima University Kagoshima Japan
| | - Hirohide Takebayashi
- Division of Neurobiology and AnatomyGraduate School of Medical and Dental Sciences, Niigata University Niigata Japan
- Center for Coordination of Research FacilitiesNiigata University Niigata Japan
| |
Collapse
|
43
|
Beverungen H, Klaszky SC, Klaszky M, Côté MP. Rehabilitation Decreases Spasticity by Restoring Chloride Homeostasis through the Brain-Derived Neurotrophic Factor-KCC2 Pathway after Spinal Cord Injury. J Neurotrauma 2020; 37:846-859. [PMID: 31578924 PMCID: PMC7071070 DOI: 10.1089/neu.2019.6526] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Activity-based therapy is routinely integrated in rehabilitation programs to facilitate functional recovery after spinal cord injury (SCI). Among its beneficial effects is a reduction of hyperreflexia and spasticity, which affects ∼75% of the SCI population. Unlike current anti-spastic pharmacological treatments, rehabilitation attenuates spastic symptoms without causing an active depression in spinal excitability, thus avoiding further interference with motor recovery. Understanding how activity-based therapies contribute to decrease spasticity is critical to identifying new pharmacological targets and to optimize rehabilitation programs. It was recently demonstrated that a decrease in the expression of KCC2, a neuronal Cl- extruder, contributes to the development spasticity in SCI rats. Although exercise can decrease spinal hyperexcitability and increase KCC2 expression on lumbar motoneurons after SCI, a causal effect remains to be established. Activity-dependent processes include an increase in brain-derived neurotrophic factor (BDNF) expression. Interestingly, BDNF is a regulator of KCC2 but also a potent modulator of spinal excitability. Therefore, we hypothesized that after SCI, the activity-dependent increase in KCC2 expression: 1) functionally contributes to reduce hyperreflexia, and 2) is regulated by BDNF. SCI rats chronically received VU0240551 (KCC2 blocker) or TrkB-IgG (BDNF scavenger) during the daily rehabilitation sessions and the frequency-dependent depression of the H-reflex, a monitor of hyperreflexia, was recorded 4 weeks post-injury. Our results suggest that the activity-dependent increase in KCC2 functionally contributes to H-reflex recovery and critically depends on BDNF activity. This study provides a new perspective in understanding how exercise impacts hyperreflexia by identifying the biological basis of the recovery of function.
Collapse
Affiliation(s)
- Henrike Beverungen
- Department of Neurobiology and Anatomy, Marion Murray Spinal Cord Research Center, Drexel University College of Medicine, Philadelphia, Pennsylvania
| | - Samantha Choyke Klaszky
- Department of Neurobiology and Anatomy, Marion Murray Spinal Cord Research Center, Drexel University College of Medicine, Philadelphia, Pennsylvania
| | - Michael Klaszky
- Department of Neurobiology and Anatomy, Marion Murray Spinal Cord Research Center, Drexel University College of Medicine, Philadelphia, Pennsylvania
| | - Marie-Pascale Côté
- Department of Neurobiology and Anatomy, Marion Murray Spinal Cord Research Center, Drexel University College of Medicine, Philadelphia, Pennsylvania
| |
Collapse
|
44
|
Plantier V, Sanchez-Brualla I, Dingu N, Brocard C, Liabeuf S, Gackière F, Brocard F. Calpain fosters the hyperexcitability of motoneurons after spinal cord injury and leads to spasticity. eLife 2019; 8:e51404. [PMID: 31815668 PMCID: PMC6927741 DOI: 10.7554/elife.51404] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 12/08/2019] [Indexed: 12/12/2022] Open
Abstract
Up-regulation of the persistent sodium current (INaP) and down-regulation of the potassium/chloride extruder KCC2 lead to spasticity after spinal cord injury (SCI). We here identified calpain as the driver of the up- and down-regulation of INaP and KCC2, respectively, in neonatal rat lumbar motoneurons. Few days after SCI, neonatal rats developed behavioral signs of spasticity with the emergence of both hyperreflexia and abnormal involuntary muscle contractions on hindlimbs. At the same time, in vitro isolated lumbar spinal cords became hyperreflexive and displayed numerous spontaneous motor outputs. Calpain-I expression paralleled with a proteolysis of voltage-gated sodium (Nav) channels and KCC2. Acute inhibition of calpains reduced this proteolysis, restored the motoneuronal expression of Nav and KCC2, normalized INaP and KCC2 function, and curtailed spasticity. In sum, by up- and down-regulating INaP and KCC2, the calpain-mediated proteolysis of Nav and KCC2 drives the hyperexcitability of motoneurons which leads to spasticity after SCI.
Collapse
Affiliation(s)
- Vanessa Plantier
- Institut de Neurosciences de la Timone (UMR7289), Aix-Marseille Université and CNRSMarseilleFrance
| | - Irene Sanchez-Brualla
- Institut de Neurosciences de la Timone (UMR7289), Aix-Marseille Université and CNRSMarseilleFrance
| | - Nejada Dingu
- Institut de Neurosciences de la Timone (UMR7289), Aix-Marseille Université and CNRSMarseilleFrance
| | - Cécile Brocard
- Institut de Neurosciences de la Timone (UMR7289), Aix-Marseille Université and CNRSMarseilleFrance
| | - Sylvie Liabeuf
- Institut de Neurosciences de la Timone (UMR7289), Aix-Marseille Université and CNRSMarseilleFrance
| | - Florian Gackière
- Institut de Neurosciences de la Timone (UMR7289), Aix-Marseille Université and CNRSMarseilleFrance
| | - Frédéric Brocard
- Institut de Neurosciences de la Timone (UMR7289), Aix-Marseille Université and CNRSMarseilleFrance
| |
Collapse
|
45
|
Lizhnyak PN, Muldoon PP, Pilaka PP, Povlishock JT, Ottens AK. Traumatic Brain Injury Temporal Proteome Guides KCC2-Targeted Therapy. J Neurotrauma 2019; 36:3092-3102. [PMID: 31122143 PMCID: PMC6818491 DOI: 10.1089/neu.2019.6415] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Advancing therapeutics for traumatic brain injury (TBI) remains a challenge, necessitating testable targets with interventions appropriately timed to intercede on evolving secondary insults. Neuroproteomics provides a global molecular approach to deduce the complex post-translational processes that underlie secondary events after TBI. Yet method advancement has outpaced approaches to interrogate neuroproteomic complexity, in particular when addressing the well-recognized temporal evolution of TBI pathobiology. Presented is a detailed account of the temporal neuroproteomic response to mild-moderate rat controlled cortical impact within perilesioned somatosensory neocortex across the first two weeks after injury. Further, this investigation assessed use of artificial neural network and functional enrichment analyses to discretize the temporal response across some 2047 significantly impacted proteins. Results were efficiently narrowed onto ion transporters with phenotypic relevance to abnormal GABAergic transmission and a delayed decline amenable to intervention under managed care conditions. The prototypical target potassium/chloride co-transporter 2 (KCC2 or SLC12A5) was investigated further with the KCC2-selective modulator CLP290. Guided by post-translational processing revealed one-day after insult to precede KCC2 protein loss a day after, CLP290 was highly effective at restoring up to 70% of lost KCC2 localization, which was significantly correlated with recovery of sham-level function in assessed somatosensory behavioral tasks. The timing of administration was important, with no significant improvement observed if given earlier, one-hour after insult, or later when KCC2 protein decline begins. Results portend importance for a detailed post-translational characterization when devising TBI treatments, and support the therapeutic promise of KCC2-targeted CLP290 intervention for positive functional recovery after brain injury.
Collapse
Affiliation(s)
- Pavel N. Lizhnyak
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, Virginia
| | - Pretal P. Muldoon
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, Virginia
| | - Pallavi P. Pilaka
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, Virginia
| | - John T. Povlishock
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, Virginia
| | - Andrew K. Ottens
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, Virginia
| |
Collapse
|
46
|
Repeated transspinal stimulation decreases soleus H-reflex excitability and restores spinal inhibition in human spinal cord injury. PLoS One 2019; 14:e0223135. [PMID: 31557238 PMCID: PMC6762874 DOI: 10.1371/journal.pone.0223135] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 09/14/2019] [Indexed: 12/12/2022] Open
Abstract
Transcutaneous spinal cord or transspinal stimulation over the thoracolumbar enlargement, the spinal location of motoneurons innervating leg muscles, modulates neural circuits engaged in the control of movement. The extent to which daily sessions (e.g. repeated) of transspinal stimulation affects soleus H-reflex excitability in individuals with chronic spinal cord injury (SCI) remains largely unknown. In this study, we established the effects of repeated cathodal transspinal stimulation on soleus H-reflex excitability and spinal inhibition in individuals with and without chronic SCI. Ten SCI and 10 healthy control subjects received monophasic transspinal stimuli of 1-ms duration at 0.2 Hz at subthreshold and suprathreshold intensities of the right soleus transspinal evoked potential (TEP). SCI subjects received an average of 16 stimulation sessions, while healthy control subjects received an average of 10 stimulation sessions. Before and one or two days post intervention, we used the soleus H reflex to assess changes in motoneuron recruitment, homosynaptic depression following single tibial nerve stimuli delivered at 0.1, 0.125, 0.2, 0.33 and 1.0 Hz, and postactivation depression following paired tibial nerve stimuli at the interstimulus intervals of 60, 100, 300, and 500 ms. Soleus H-reflex excitability was decreased in both legs in motor incomplete and complete SCI but not in healthy control subjects. Soleus H-reflex homosynaptic and postactivation depression was present in motor incomplete and complete SCI but was of lesser strength to that observed in healthy control subjects. Repeated transspinal stimulation increased homosynaptic depression in all SCI subjects and remained unaltered in healthy controls. Postactivation depression remained unaltered in all subject groups. Lastly, transspinal stimulation decreased the severity of spasms and ankle clonus. The results indicate decreased reflex hyperexcitability and recovery of spinal inhibitory control in the injured human spinal cord with repeated transspinal stimulation. Transspinal stimulation is a noninvasive neuromodulation method for restoring spinally-mediated afferent reflex actions after SCI in humans.
Collapse
|
47
|
Mazzone GL, Nistri A. Modulation of extrasynaptic GABAergic receptor activity influences glutamate release and neuronal survival following excitotoxic damage to mouse spinal cord neurons. Neurochem Int 2019; 128:175-185. [PMID: 31051211 DOI: 10.1016/j.neuint.2019.04.018] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 04/08/2019] [Accepted: 04/30/2019] [Indexed: 12/14/2022]
Abstract
Excitotoxic levels of released glutamate trigger a cascade of deleterious cellular events leading to delayed neuronal death. This phenomenon implies extensive dysregulation in the balance between network excitation and inhibition. Our hypothesis was that enhancing network inhibition should prevent excitotoxicity and provide neuroprotection. To test this notion, we used mouse organotypic spinal slice cultures and explored if excitotoxicity caused by the potent glutamate analogue kainate was blocked by pharmacological increase in GABAA receptor activity. To this end we monitored (with a biosensor) real-time glutamate release following 1 h kainate application and quantified neuronal survival 24 h later. Glutamate release evoked by kainate was strongly decreased by the allosteric GABAA modulator midazolam (10 nM) or the GABA agonist THIP (10 μM), leading to neuroprotection. On the contrary, much higher glutamate release was induced by the GABA antagonist bicuculline (20 μM) that inhibits synaptic and extrasynaptic GABAA receptors. Gabazine (20 μM), an antagonist of synaptic GABAA receptors, had no effect on glutamate release or neuroprotection. No effect was observed with the glycine antagonist strychnine or the glycine agonist L-alanine. These findings indicate that enhancement of GABA receptor activity was an effective tool to counteract excitotoxic death in spinal networks. In view of the potent activity by THIP, preferentially acting on extrasynaptic GABAA receptors, the present data imply a significant role for extrasynaptic GABAA receptors in sparing spinal cord neurons from injury.
Collapse
Affiliation(s)
- Graciela L Mazzone
- Instituto de Investigaciones en Medicina Traslacional (IIMT), CONICET-Universidad Austral, Derqui-Pilar, Buenos Aires, Argentina.
| | - Andrea Nistri
- Neuroscience Dept., International School for Advanced Studies (SISSA), Trieste, Italy
| |
Collapse
|
48
|
Lee-Hotta S, Uchiyama Y, Kametaka S. Role of the BDNF-TrkB pathway in KCC2 regulation and rehabilitation following neuronal injury: A mini review. Neurochem Int 2019; 128:32-38. [PMID: 30986502 DOI: 10.1016/j.neuint.2019.04.003] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 03/04/2019] [Accepted: 04/08/2019] [Indexed: 02/08/2023]
Abstract
In most mature neurons, low levels of intracellular Cl- concentrations ([Cl-]i) are maintained by channels and transporters, particularly the K+-Cl- cotransporter 2 (KCC2), which is the only Cl- extruder in most neurons. Recent studies have implicated KCC2 expression in the molecular mechanisms underlying neuronal disorders, such as spasticity, epilepsy and neuropathic pain. Alterations in KCC2 expression have been associated with brain-derived neurotrophic factor (BDNF) and its receptor tropomyosin-related kinase B (TrkB). The present review summarizes recent progress regarding the roles of Cl- regulators in immature and mature neurons. Moreover, we focus on the role of KCC2 regulation via the BDNF-TrkB pathway in spinal cord injury and rehabilitation, as prior studies have shown that the BDNF-TrkB pathway can affect both the pathological development and functional amelioration of spinal cord injuries. Evidence suggests that rehabilitation using active exercise and mechanical stimulation can attenuate spasticity and neuropathic pain in animal models, likely due to the upregulation of KCC2 expression via the BDNF-TrkB pathway. Moreover, research suggests that such rehabilitation efforts may recover KCC2 expression without the use of exogenous BDNF.
Collapse
Affiliation(s)
- Sachiko Lee-Hotta
- Department of Rehabilitation Sciences, Graduate School of Medicine, Nagoya University, 1-1-20, Daiko-minami Higashi-ku, Nagoya-shi, Aichi, 461-8673, Japan.
| | - Yasushi Uchiyama
- Department of Rehabilitation Sciences, Graduate School of Medicine, Nagoya University, 1-1-20, Daiko-minami Higashi-ku, Nagoya-shi, Aichi, 461-8673, Japan.
| | - Satoshi Kametaka
- Department of Rehabilitation Sciences, Graduate School of Medicine, Nagoya University, 1-1-20, Daiko-minami Higashi-ku, Nagoya-shi, Aichi, 461-8673, Japan.
| |
Collapse
|
49
|
Intramuscular Injection of Adenoassociated Virus Encoding Human Neurotrophic Factor 3 and Exercise Intervention Contribute to Reduce Spasms after Spinal Cord Injury. Neural Plast 2019; 2019:3017678. [PMID: 30984254 PMCID: PMC6432737 DOI: 10.1155/2019/3017678] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 10/12/2018] [Accepted: 10/23/2018] [Indexed: 01/08/2023] Open
Abstract
Limb spasms are phenomena of hyperreflexia that occur after spinal cord injury. Currently, the clinical treatment is less than ideal. Our goal is to develop a combination therapy based on individualized medicine to reduce spasticity after spinal cord injury. In this study, rats received a severe contusive injury at the T9 segment of the spinal cord, followed by gene therapy with adenoassociated virus encoding human neurotrophic factor 3 (AAV-NT3) and a 2-week exercise program starting at 4 weeks after injury. We quantified the frequency of spasms during a swimming test at 4 and 6 weeks after injury and confirmed the results of the swimming test by measuring the H-reflex of the plantar muscle. We obtained weekly hind limb exercise scores to assess the effect of the interventions in hind limb motor function improvement. Then, we used immunofluorescence to observe the immunoreactivity of spinal motor neurons, synaptophysin, cholinergic interneurons, and GABAergic interneurons. We also measured the expression of KCC2 in the spinal cord by western blot. We found that AAV-NT3 gene therapy, exercise, and combination therapy all attenuated the frequency of spasms in the swimming test conducted at 6 weeks after spinal cord injury and increased rate-dependent depression of H-reflex. Combination therapy was significantly superior to AAV-NT3 alone in protecting motor neurons. Recovery of KCC2 expression was significantly greater in rats treated with combination therapy than in the exercise group. Combination therapy was also significantly superior to individual therapies in remodeling spinal cord neurons. Our study shows that the combination of AAV-NT3 gene therapy and exercise can alleviate muscle spasm after spinal cord injury by altering the excitability of spinal interneurons and motor neurons. However, combination therapy did not show a significant additive effect, which needs to be improved by adjusting the combined strategy.
Collapse
|
50
|
Wilkinson CM, Fedor BA, Aziz JR, Nadeau CA, Brar PS, Clark JJA, Colbourne F. Failure of bumetanide to improve outcome after intracerebral hemorrhage in rat. PLoS One 2019; 14:e0210660. [PMID: 30629699 PMCID: PMC6328169 DOI: 10.1371/journal.pone.0210660] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 12/28/2018] [Indexed: 11/19/2022] Open
Abstract
After intracerebral hemorrhage (ICH), brain edema commonly occurs and can cause death. Along with edema, there are significant alterations in the concentrations of key ions such as sodium, potassium, and chloride, which are essential to brain function. NKCC1, a cation-chloride cotransporter, is upregulated after brain damage, such as traumatic injury and ischemic stroke. NKCC1 brings sodium and chloride into the cell, possibly worsening ion dyshomeostasis. Bumetanide, a specific NKCC1 antagonist, blocks the transport of chloride into cells, and thus should attenuate the increases in chloride, which should lessen brain edema and improve neuronal functioning post-ICH, as with other injuries. We used the collagenase model of ICH to test whether bumetanide treatment for three days (vs. vehicle) would improve outcome. We gave bumetanide beginning at two hours or seven days post-ICH and measured behavioural outcome, edema, and brain ion content after treatment. There was some evidence for a minor reduction in edema after early dosing, but this did not improve behaviour or lessen injury. Contrary to our hypothesis, bumetanide did not normalize ion concentrations after late dosing. Bumetanide did not improve behavioural outcome or affect lesion volume. After ICH, bumetanide is safe to use in rats but does not improve functional outcome in the majority of animals.
Collapse
Affiliation(s)
| | - Brittany A. Fedor
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Jasmine R. Aziz
- Department of Psychology, University of Alberta, Edmonton, Alberta, Canada
| | - Colby A. Nadeau
- Department of Psychology, University of Alberta, Edmonton, Alberta, Canada
| | - Paul S. Brar
- Department of Psychology, University of Alberta, Edmonton, Alberta, Canada
| | - Julia J. A. Clark
- Department of Psychology, University of Alberta, Edmonton, Alberta, Canada
| | - Frederick Colbourne
- Department of Psychology, University of Alberta, Edmonton, Alberta, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|