1
|
Escarrat V, Reato D, Blivet G, Touchon J, Rougon G, Bos R, Debarbieux F. Dorsoventral photobiomodulation therapy safely reduces inflammation and sensorimotor deficits in a mouse model of multiple sclerosis. J Neuroinflammation 2024; 21:321. [PMID: 39696356 DOI: 10.1186/s12974-024-03294-2] [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: 09/02/2024] [Accepted: 11/11/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Non-invasive photobiomodulation therapy (PBMT), employing specific infrared light wavelengths to stimulate biological tissues, has recently gained attention for its application to treat neurological disorders. Here, we aimed to uncover the cellular targets of PBMT and assess its potential as a therapeutic intervention for multiple sclerosis (MS). METHODS We applied daily dorsoventral PBMT in an experimental autoimmune encephalomyelitis (EAE) mouse model, which recapitulates key features of MS, and revealed a strong positive impact of PBMT on the sensorimotor deficits. To understand the cellular mechanisms underlying these striking effects, we used state-of-the-art tools and methods ranging from two-photon longitudinal imaging of triple fluorescent reporter mice to histological investigations and patch-clamp electrophysiological recordings. RESULTS We found that PBMT induced anti-inflammatory and neuroprotective effects in the dorsal spinal cord. PBMT prevented peripheral immune cell infiltration, glial reactivity, as well as the EAE-induced hyperexcitability of spinal interneurons, both in dorsal and ventral areas, which likely underlies the behavioral effects of the treatment. Thus, aside from confirming the safety of PBMT in healthy mice, our preclinical investigation suggests that PBMT exerts a systemic and beneficial effect on the physiopathology of EAE, primarily resulting in the modulation of the inflammatory processes. CONCLUSION PBMT may therefore represent a new valuable therapeutic option to treat MS symptoms.
Collapse
Affiliation(s)
- Vincent Escarrat
- Aix Marseille Univ, CNRS, INT, Inst. Neurosci. Timone, Marseille, France
- Aix Marseille Univ, CNRS, CERIMED, Marseille, France
- REGEnLIFE, Paris, France
| | - Davide Reato
- Aix Marseille Univ, CNRS, INT, Inst. Neurosci. Timone, Marseille, France
- Département BEL, Mines Saint-Etienne, Centre CMP, 13541, Gardanne, France
| | | | | | - Geneviève Rougon
- Aix Marseille Univ, CNRS, INT, Inst. Neurosci. Timone, Marseille, France
| | - Rémi Bos
- Aix Marseille Univ, CNRS, INT, Inst. Neurosci. Timone, Marseille, France.
| | - Franck Debarbieux
- Aix Marseille Univ, CNRS, INT, Inst. Neurosci. Timone, Marseille, France.
- Aix Marseille Univ, CNRS, CERIMED, Marseille, France.
- Institut Universitaire de France (IUF), Paris, France.
| |
Collapse
|
2
|
Aghili SH, Manavi MA, Panji M, Farhang Ranjbar M, Abrishami R, Dehpour AR. Mirtazapine Improves Locomotor Activity and Attenuates Neuropathic Pain Following Spinal Cord Injury in Rats via Neuroinflammation Modulation. Neurochem Res 2024; 49:3326-3341. [PMID: 39271550 DOI: 10.1007/s11064-024-04240-7] [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: 04/01/2024] [Revised: 05/16/2024] [Accepted: 09/03/2024] [Indexed: 09/15/2024]
Abstract
Neuroinflammation-related locomotor deficits and neuropathic pain are expected outcomes of spinal cord injury (SCI). The atypical antidepressant mirtazapine has exhibited potential neuroprotective and anti-inflammatory effects. This research aims to investigate the impacts of mirtazapine on post-SCI neuropathic pain and locomotor recovery, with a particular focus on neuroinflammation. The study utilized 30 male Wistar rats divided into five groups: Sham, SCI with vehicle treatment, and SCI administered with mirtazapine (3, 10, and 30 mg/kg/day, ip, for one week). Locomotor activity was assessed using the Basso, Beattie, and Bresnahan (BBB) scale. Mechanical, thermal, and cold allodynia were assessed using von-Frey filaments, tail flick latency, and the acetone test, respectively. ELISA was utilized to measure cytokines, while Western blotting was used to determine TRPV1 channel, 5-HT2A receptor, NLRP3, and iNOS expression. Histopathological analyses were also examined, including hematoxylin and eosin (H&E) and Luxol fast blue (LFB) staining. Mirtazapine (10 and 30 mg/kg/day) significantly improved locomotor recovery according to BBB score. It attenuated mechanical, thermal, and cold allodynia post-SCI. Moreover, it decreased pro-inflammatory cytokines TNF-α, IL-1β, IL-6, and IL-18, while increasing anti-inflammatory cytokine IL-4 and IL-10. Furthermore, it downregulated iNOS, NLRP3, and TRPV1 expression and upregulated the 5-HT2A receptor. H&E and LFB staining further revealed attenuated tissue damage and decreased demyelination. Our findings suggest that mirtazapine can alleviate neuropathic pain and reinforce locomotor recovery post-SCI by modulating neuroinflammatory responses, NLRP3, iNOS, TRPV1 channel, and 5-HT2A receptor expression.
Collapse
Affiliation(s)
- Seyed Hadi Aghili
- Research Center for Trauma in Police Operations, Directorate of Health, Rescue & Treatment, Police Headquarter, Tehran, Iran
- Neurosurgery Department, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
- Department of Neurosurgery, Valiasr Hospital, Tehran, Iran
| | - Mohammad Amin Manavi
- Experimental Medicine Research Center, Tehran University of Medical Sciences, P.O. Box 13145-784, Tehran, Iran
| | - Mohammad Panji
- Research Center for Life, Health Sciences & Biotechnology of the Police, Directorate of Health, Rescue & Treatment, Police Headquarters, Tehran, Iran
| | - Mehri Farhang Ranjbar
- Department of Support and Services Management, Institute of Management and Organizational Resources, Policing Sciences and Social Studies Research Institute, Tehran, Iran
| | - Ramin Abrishami
- Research Center for Trauma in Police Operations, Directorate of Health, Rescue & Treatment, Police Headquarter, Tehran, Iran.
- Department of Clinical Pharmacy, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Ahmad Reza Dehpour
- Experimental Medicine Research Center, Tehran University of Medical Sciences, P.O. Box 13145-784, Tehran, Iran.
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
3
|
Capilla-López J, Hernández RG, Carrero-Rojas G, Calvo PM, Alvarez FJ, de la Cruz RR, Pastor AM. VEGF, but Not BDNF, Prevents the Downregulation of KCC2 Induced by Axotomy in Extraocular Motoneurons. Int J Mol Sci 2024; 25:9942. [PMID: 39337430 PMCID: PMC11432591 DOI: 10.3390/ijms25189942] [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: 08/14/2024] [Revised: 09/09/2024] [Accepted: 09/11/2024] [Indexed: 09/30/2024] Open
Abstract
The potassium-chloride cotransporter KCC2 is the main extruder of Cl- in neurons. It plays a fundamental role in the activity of the inhibitory neurotransmitters (GABA and glycine) since low levels of KCC2 promote intracellular Cl- accumulation, leading to the depolarizing activity of GABA and glycine. The downregulation of this cotransporter occurs in neurological disorders characterized by hyperexcitability, such as epilepsy, neuropathic pain, and spasticity. KCC2 is also downregulated after axotomy. If muscle reinnervation is allowed, the KCC2 levels recover in motoneurons. Therefore, we argued that target-derived neurotrophic factors might be involved in the regulation of KCC2 expression. For this purpose, we performed the axotomy of extraocular motoneurons via the monocular enucleation of adult rats, and a pellet containing either VEGF or BDNF was chronically implanted in the orbit. Double confocal immunofluorescence of choline acetyl-transferase (ChAT) and KCC2 was carried out in the brainstem sections. Axotomy led to a KCC2 decrease in the neuropil and somata of extraocular motoneurons, peaking at 15 days post-lesion, with the exception of the abducens motoneuron somata. VEGF administration prevented the axotomy-induced KCC2 downregulation. By contrast, BDNF either maintained or reduced the KCC2 levels following axotomy, suggesting that BDNF is involved in the axotomy-induced KCC2 downregulation in extraocular motoneurons. The finding that VEGF prevents KCC2 decrease opens up new possibilities for the treatment of neurological disorders coursing with neuronal hyperactivity due to KCC2 downregulation.
Collapse
Affiliation(s)
- Jaime Capilla-López
- Departamento de Fisiología, Facultad de Biología, Universidad de Sevilla, 41012 Sevilla, Spain
| | - Rosendo G Hernández
- Departamento de Fisiología, Facultad de Biología, Universidad de Sevilla, 41012 Sevilla, Spain
| | - Génova Carrero-Rojas
- Center for Anatomy and Cell Biology, Division of Anatomy, Medical University Vienna, 1090 Vienna, Austria
| | - Paula M Calvo
- Departamento de Fisiología, Facultad de Biología, Universidad de Sevilla, 41012 Sevilla, Spain
- Department of Cell Biology, Emory University, Atlanta, GA 30322, USA
| | | | - Rosa R de la Cruz
- Departamento de Fisiología, Facultad de Biología, Universidad de Sevilla, 41012 Sevilla, Spain
| | - Angel M Pastor
- Departamento de Fisiología, Facultad de Biología, Universidad de Sevilla, 41012 Sevilla, Spain
| |
Collapse
|
4
|
Zou Z, Fan W, Liu H, Liu Q, He H, Huang F. The roles of 5-HT in orofacial pain. Oral Dis 2024; 30:3838-3849. [PMID: 38622872 DOI: 10.1111/odi.14960] [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: 12/13/2023] [Revised: 03/10/2024] [Accepted: 04/02/2024] [Indexed: 04/17/2024]
Abstract
OBJECTIVES Acute and chronic orofacial pain are very common and remain a vexing health problem that has a negative effect on the quality of life. Serotonin (5-HydroxyTryptamine, 5-HT) is a kind of monoamine neurotransmitter that is involved in many physiological and pathological processes. However, its role in orofacial pain remains inconclusive. Therefore, this review aims to summarize the recent advances in understanding the effect exerted by 5-HT on the modulation of orofacial pain. SUBJECTS AND METHODS An extensive search was conducted on PubMed and Web of Science for pertinent studies focusing on the effects of 5-HT on the modulation of orofacial pain. RESULTS In this review, we concisely review how 5-HT mediates orofacial pain, how 5-HT is regulated and how we can translate these findings into clinical applications for the prevention and/or treatment of orofacial pain. CONCLUSIONS 5-HT plays a key role in the modulation of orofacial pain, implying that 5-HT modulators may serve as effective treatment for orofacial pain. However, further research on the precise mechanisms underlying the modulation of orofacial pain is still warranted.
Collapse
Affiliation(s)
- Zhishan Zou
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
- Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Wenguo Fan
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
- Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Haotian Liu
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Qing Liu
- Paediatric Dentistry and Orthodontics, Faculty of Dentistry, The University of Hong Kong, Hong Kong, SAR, China
| | - Hongwen He
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
- Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Fang Huang
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
- Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
5
|
Ionescu MI, Grigoras IF, Ionescu RB, Chitimus DM, Haret RM, Ianosi B, Ceanga M, Zagrean AM. Oxytocin Exhibits Neuroprotective Effects on Hippocampal Cultures under Severe Oxygen-Glucose Deprivation Conditions. Curr Issues Mol Biol 2024; 46:6223-6236. [PMID: 38921042 PMCID: PMC11202210 DOI: 10.3390/cimb46060371] [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: 05/19/2024] [Revised: 06/12/2024] [Accepted: 06/16/2024] [Indexed: 06/27/2024] Open
Abstract
Perinatal asphyxia (PA) and hypoxic-ischemic encephalopathy can result in severe, long-lasting neurological deficits. In vitro models, such as oxygen-glucose deprivation (OGD), are used experimentally to investigate neuronal response to metabolic stress. However, multiple variables can affect the severity level of OGD/PA and may confound any measured treatment effect. Oxytocin (OXT) has emerged as a potential neuroprotective agent against the deleterious effects of PA. Previous studies have demonstrated OXT's potential to enhance neuronal survival in immature hippocampal cultures exposed to OGD, possibly by modulating gamma-aminobutyric acid-A receptor activity. Moreover, OXT's precise impact on developing hippocampal neurons under different severities of OGD/PA remains uncertain. In this study, we investigated the effects of OXT (0.1 µM and 1 µM) on 7-day-old primary rat hippocampal cultures subjected to 2 h OGD/sham normoxic conditions. Cell culture viability was determined using the resazurin assay. Our results indicate that the efficacy of 1 µM OXT treatment varied according to the severity of the OGD-induced lesion, exhibiting a protective effect (p = 0.022) only when cellular viability dropped below 49.41% in non-treated OGD cultures compared to normoxic ones. Furthermore, administration of 0.1 µM OXT did not yield significant effects, irrespective of lesion severity (p > 0.05). These findings suggest that 1 µM OXT treatment during OGD confers neuroprotection exclusively in severe lesions in hippocampal neurons after 7 days in vitro. Further research is warranted to elucidate the mechanisms involved in OXT-mediated neuroprotection.
Collapse
Affiliation(s)
- Mara Ioana Ionescu
- Department of Functional Sciences, Division of Physiology II-Neuroscience, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (M.I.I.); (I.-F.G.); (R.-B.I.); (D.M.C.); (R.M.H.); (B.I.)
| | - Ioana-Florentina Grigoras
- Department of Functional Sciences, Division of Physiology II-Neuroscience, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (M.I.I.); (I.-F.G.); (R.-B.I.); (D.M.C.); (R.M.H.); (B.I.)
- Wellcome Centre for Integrative Neuroimaging, Functional MRI of the Brain, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
| | - Rosana-Bristena Ionescu
- Department of Functional Sciences, Division of Physiology II-Neuroscience, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (M.I.I.); (I.-F.G.); (R.-B.I.); (D.M.C.); (R.M.H.); (B.I.)
- Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 1TN, UK
- NIHR Biomedical Research Centre, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Diana Maria Chitimus
- Department of Functional Sciences, Division of Physiology II-Neuroscience, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (M.I.I.); (I.-F.G.); (R.-B.I.); (D.M.C.); (R.M.H.); (B.I.)
| | - Robert Mihai Haret
- Department of Functional Sciences, Division of Physiology II-Neuroscience, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (M.I.I.); (I.-F.G.); (R.-B.I.); (D.M.C.); (R.M.H.); (B.I.)
- Department of Ophthalmology, University Medical Center Gottingen, 37075 Gottingen, Germany
| | - Bogdan Ianosi
- Department of Functional Sciences, Division of Physiology II-Neuroscience, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (M.I.I.); (I.-F.G.); (R.-B.I.); (D.M.C.); (R.M.H.); (B.I.)
- Department of Neurology, Stroke Unit, Neuromed Campus, Kepler University Hospital, 4020 Linz, Austria
| | - Mihai Ceanga
- Department of Functional Sciences, Division of Physiology II-Neuroscience, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (M.I.I.); (I.-F.G.); (R.-B.I.); (D.M.C.); (R.M.H.); (B.I.)
- Section of Translational Neuroimmunology, Department of Neurology, Jena University Hospital, 07747 Jena, Germany
| | - Ana-Maria Zagrean
- Department of Functional Sciences, Division of Physiology II-Neuroscience, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (M.I.I.); (I.-F.G.); (R.-B.I.); (D.M.C.); (R.M.H.); (B.I.)
| |
Collapse
|
6
|
Parameshwarappa V, Siponen MI, Watabe I, Karkaba A, Galazyuk A, Noreña AJ. Noise-induced hearing loss alters potassium-chloride cotransporter KCC2 and GABA inhibition in the auditory centers. Sci Rep 2024; 14:10689. [PMID: 38724641 PMCID: PMC11082187 DOI: 10.1038/s41598-024-60858-1] [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: 09/26/2023] [Accepted: 04/29/2024] [Indexed: 05/12/2024] Open
Abstract
Homeostatic plasticity, the ability of neurons to maintain their averaged activity constant around a set point value, is thought to account for the central hyperactivity after hearing loss. Here, we investigated the putative role of GABAergic neurotransmission in this mechanism after a noise-induced hearing loss larger than 50 dB in high frequencies in guinea pigs. The effect of GABAergic inhibition is linked to the normal functioning of K + -Cl- co-transporter isoform 2 (KCC2) which maintains a low intracellular concentration of chloride. The expression of membrane KCC2 were investigated before and after noise trauma in the ventral and dorsal cochlear nucleus (VCN and DCN, respectively) and in the inferior colliculus (IC). Moreover, the effect of gabazine (GBZ), a GABA antagonist, was also studied on the neural activity in IC. We show that KCC2 is downregulated in VCN, DCN and IC 3 days after noise trauma, and in DCN and IC 30 days after the trauma. As expected, GBZ application in the IC of control animals resulted in an increase of spontaneous and stimulus-evoked activity. In the noise exposed animals, on the other hand, GBZ application decreased the stimulus-evoked activity in IC neurons. The functional implications of these central changes are discussed.
Collapse
Affiliation(s)
- V Parameshwarappa
- Laboratory of Cognitive Neurosciences, Centre National de la Recherche Scientifique, Aix-Marseille University, 3 Place Victor Hugo, 13003, Marseille, France
| | - M I Siponen
- Laboratory of Cognitive Neurosciences, Centre National de la Recherche Scientifique, Aix-Marseille University, 3 Place Victor Hugo, 13003, Marseille, France
| | - I Watabe
- Laboratory of Cognitive Neurosciences, Centre National de la Recherche Scientifique, Aix-Marseille University, 3 Place Victor Hugo, 13003, Marseille, France
| | - A Karkaba
- Laboratory of Cognitive Neurosciences, Centre National de la Recherche Scientifique, Aix-Marseille University, 3 Place Victor Hugo, 13003, Marseille, France
| | - A Galazyuk
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University, Rootstown, OH, USA
| | - A J Noreña
- Laboratory of Cognitive Neurosciences, Centre National de la Recherche Scientifique, Aix-Marseille University, 3 Place Victor Hugo, 13003, Marseille, France.
| |
Collapse
|
7
|
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
|
8
|
Zhu H, Dalvi U, Cazenave W, Cattaert D, Branchereau P. Excitatory action of low frequency depolarizing GABA/glycine synaptic inputs is prevalent in prenatal spinal SOD1 G93A motoneurons. J Physiol 2024; 602:913-932. [PMID: 38345477 DOI: 10.1113/jp285105] [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: 06/01/2023] [Accepted: 01/24/2024] [Indexed: 03/09/2024] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal adult-onset neurodegenerative disease characterized by progressive motor neuron degeneration and muscle paralysis. Recent evidence suggests the dysfunction of inhibitory signalling in ALS motor neurons. We have shown that embryonic day (E)17.5 spinal motoneurons (MNs) of the SOD1G93A mouse model of ALS exhibit an altered chloride homeostasis. At this prenatal stage, inhibition of spinal motoneurons (MNs) is mediated by depolarizing GABAergic/glycinergic postsynaptic potentials (dGPSPs). Here, using an ex vivo preparation and patch clamp recording from MNs with a chloride equilibrium set below spike threshold, we report that low input resistance (Rin ) E17.5 MNs from the SOD1G93A ALS mouse model do not correctly integrate dGPSPs evoked by electrical stimulations of GABA/glycine inputs at different frequencies. Indeed, firing activity of most wild-type (WT) MNs with low Rin was inhibited by incoming dGPSPs, whereas low Rin SOD1G93A MNs were excited or exhibited a dual response (excited by low frequency dGPSPs and inhibited by high frequency dGPSPs). Simulation highlighted the importance of the GABA/glycine input density and showed that pure excitation could be obtained in SOD-like MNs by moving GABA/glycine input away from the cell body to dendrites. This was in agreement with confocal imaging showing a lack of peri-somatic inhibitory terminals in SOD1G93A MNs compared to WT littermates. Putative fast ALS-vulnerable MNs with low Rin are therefore lacking functional inhibition at the near-term prenatal stage. KEY POINTS: We analysed the integration of GABAergic/glycinergic synaptic events by embryonic spinal motoneurons (MNs) in a mouse model of the amyotrophic lateral sclerosis (ALS) neurodegenerative disease. We found that GABAergic/glycinergic synaptic events do not properly inhibit ALS MNs with low input resistance, most probably corresponding to future vulnerable MNs. We used a neuron model to highlight the importance of the GABA/glycine terminal location and density in the integration of the GABAergic/glycinergic synaptic events. Confocal imaging showed a lack of GABA/glycine terminals on the cell body of ALS MNs. The present study suggests that putative ALS vulnerable MNs with low Rin lack functional inhibition at the near-term stage.
Collapse
Affiliation(s)
- Hongmei Zhu
- University Bordeaux, CNRS, INCIA, UMR 5287, Bordeaux, France
| | - Urvashi Dalvi
- University Bordeaux, CNRS, INCIA, UMR 5287, Bordeaux, France
| | | | - Daniel Cattaert
- University Bordeaux, CNRS, INCIA, UMR 5287, Bordeaux, France
| | | |
Collapse
|
9
|
Romero ME, McElhenney SJ, Yu J. Trapping a non-cognate nucleotide upon initial binding for replication fidelity control in SARS-CoV-2 RNA dependent RNA polymerase. Phys Chem Chem Phys 2024; 26:1792-1808. [PMID: 38168789 DOI: 10.1039/d3cp04410f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
The RNA dependent RNA polymerase (RdRp) in SARS-CoV-2 is a highly conserved enzyme responsible for viral genome replication/transcription. To understand how the viral RdRp achieves fidelity control during such processes, here we computationally investigate the natural non-cognate vs. cognate nucleotide addition and selectivity during viral RdRp elongation. We focus on the nucleotide substrate initial binding (RdRp active site open) to the prechemical insertion (active site closed) of the RdRp. The current studies were first carried out using microsecond ensemble equilibrium all-atom molecular dynamics (MD) simulations. Due to the slow conformational changes (from open to closed) during nucleotide insertion and selection, enhanced or umbrella sampling methods have been further employed to calculate the free energy profiles of the nucleotide insertion. Our studies find notable stability of noncognate dATP and GTP upon initial binding in the active-site open state. The results indicate that while natural cognate ATP and Remdesivir drug analogue (RDV-TP) are biased toward stabilization in the closed state to facilitate insertion, the natural non-cognate dATP and GTP can be well trapped in off-path initial binding configurations and prevented from insertion so that to be further rejected. The current work thus presents the intrinsic nucleotide selectivity of SARS-CoV-2 RdRp for natural substrate fidelity control, which should be considered in antiviral drug design.
Collapse
Affiliation(s)
- Moises E Romero
- Department of Chemistry, University of California, Irvine, CA 92697, USA
| | | | - Jin Yu
- Department of Physics and Astronomy, Department of Chemistry, NSF-Simmons Center for Multi-scale Cell Fate Research, University of California, Irvine, CA 92697, USA.
| |
Collapse
|
10
|
Delestrée N, Semizoglou E, Pagiazitis JG, Vukojicic A, Drobac E, Paushkin V, Mentis GZ. Serotonergic dysfunction impairs locomotor coordination in spinal muscular atrophy. Brain 2023; 146:4574-4593. [PMID: 37678880 PMCID: PMC10629775 DOI: 10.1093/brain/awad221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 05/12/2023] [Accepted: 06/11/2023] [Indexed: 09/09/2023] Open
Abstract
Neuromodulation by serotonin regulates the activity of neuronal networks responsible for a wide variety of essential behaviours. Serotonin (or 5-HT) typically activates metabotropic G protein-coupled receptors, which in turn initiate second messenger signalling cascades and induce short and long-lasting behavioural effects. Serotonin is intricately involved in the production of locomotor activity and gait control for different motor behaviours. Although dysfunction of serotonergic neurotransmission has been associated with mood disorders and spasticity after spinal cord injury, whether and to what extent such dysregulation is implicated in movement disorders has not been firmly established. Here, we investigated whether serotonergic neuromodulation is affected in spinal muscular atrophy (SMA), a neurodegenerative disease caused by ubiquitous deficiency of the SMN protein. The hallmarks of SMA are death of spinal motor neurons, muscle atrophy and impaired motor control, both in human patients and mouse models of disease. We used a severe mouse model of SMA, that closely recapitulates the severe symptoms exhibited by type I SMA patients, the most common and most severe form of the disease. Together, with mouse genetics, optogenetics, physiology, morphology and behavioural analysis, we report severe dysfunction of serotonergic neurotransmission in the spinal cord of SMA mice, both at early and late stages of the disease. This dysfunction is followed by reduction of 5-HT synapses on vulnerable motor neurons. We demonstrate that motor neurons innervating axial and trunk musculature are preferentially affected, suggesting a possible cause for the proximo-distal progression of disease, and raising the possibility that it may underlie scoliosis in SMA patients. We also demonstrate that the 5-HT dysfunction is caused by SMN deficiency in serotonergic neurons in the raphe nuclei of the brainstem. The behavioural significance of the dysfunction in serotonergic neuromodulation is underlined by inter-limb discoordination in SMA mice, which is ameliorated when selective restoration of SMN in 5-HT neurons is achieved by genetic means. Our study uncovers an unexpected dysfunction of serotonergic neuromodulation in SMA and indicates that, if normal function is to be restored under disease conditions, 5-HT neuromodulation should be a key target for therapeutic approaches.
Collapse
Affiliation(s)
- Nicolas Delestrée
- Center for Motor Neuron Biology and Disease, Columbia University, New York, NY 10032, USA
- Department of Neurology, Columbia University, New York, NY 10032, USA
| | - Evangelia Semizoglou
- Center for Motor Neuron Biology and Disease, Columbia University, New York, NY 10032, USA
- Department of Neurology, Columbia University, New York, NY 10032, USA
| | - John G Pagiazitis
- Center for Motor Neuron Biology and Disease, Columbia University, New York, NY 10032, USA
- Department of Neurology, Columbia University, New York, NY 10032, USA
| | - Aleksandra Vukojicic
- Center for Motor Neuron Biology and Disease, Columbia University, New York, NY 10032, USA
- Department of Neurology, Columbia University, New York, NY 10032, USA
| | - Estelle Drobac
- Center for Motor Neuron Biology and Disease, Columbia University, New York, NY 10032, USA
- Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
| | - Vasilissa Paushkin
- Center for Motor Neuron Biology and Disease, Columbia University, New York, NY 10032, USA
- Department of Neurology, Columbia University, New York, NY 10032, USA
| | - George Z Mentis
- Center for Motor Neuron Biology and Disease, Columbia University, New York, NY 10032, USA
- Department of Neurology, Columbia University, New York, NY 10032, USA
- Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
| |
Collapse
|
11
|
Calvo PM, de la Cruz RR, Pastor AM, Alvarez FJ. Preservation of KCC2 expression in axotomized abducens motoneurons and its enhancement by VEGF. Brain Struct Funct 2023; 228:967-984. [PMID: 37005931 PMCID: PMC10428176 DOI: 10.1007/s00429-023-02635-w] [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: 11/08/2022] [Accepted: 03/23/2023] [Indexed: 04/04/2023]
Abstract
The potassium chloride cotransporter 2 (KCC2) is the main Cl- extruder in neurons. Any alteration in KCC2 levels leads to changes in Cl- homeostasis and, consequently, in the polarity and amplitude of inhibitory synaptic potentials mediated by GABA or glycine. Axotomy downregulates KCC2 in many different motoneurons and it is suspected that interruption of muscle-derived factors maintaining motoneuron KCC2 expression is in part responsible. In here, we demonstrate that KCC2 is expressed in all oculomotor nuclei of cat and rat, but while trochlear and oculomotor motoneurons downregulate KCC2 after axotomy, expression is unaltered in abducens motoneurons. Exogenous application of vascular endothelial growth factor (VEGF), a neurotrophic factor expressed in muscle, upregulated KCC2 in axotomized abducens motoneurons above control levels. In parallel, a physiological study using cats chronically implanted with electrodes for recording abducens motoneurons in awake animals, demonstrated that inhibitory inputs related to off-fixations and off-directed saccades in VEGF-treated axotomized abducens motoneurons were significantly higher than in control, but eye-related excitatory signals in the on direction were unchanged. This is the first report of lack of KCC2 regulation in a motoneuron type after injury, proposing a role for VEGF in KCC2 regulation and demonstrating the link between KCC2 and synaptic inhibition in awake, behaving animals.
Collapse
Affiliation(s)
- Paula M Calvo
- Departamento de Fisiología, Facultad de Biología, Universidad de Sevilla, 41012, Seville, Spain
- Department of Cell Biology, Emory University, Atlanta, GA, 30322, USA
| | - Rosa R de la Cruz
- Departamento de Fisiología, Facultad de Biología, Universidad de Sevilla, 41012, Seville, Spain
| | - Angel M Pastor
- Departamento de Fisiología, Facultad de Biología, Universidad de Sevilla, 41012, Seville, Spain
| | | |
Collapse
|
12
|
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
|
13
|
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
|
14
|
Hudson KE, Grau JW. Ionic Plasticity: Common Mechanistic Underpinnings of Pathology in Spinal Cord Injury and the Brain. Cells 2022; 11:2910. [PMID: 36139484 PMCID: PMC9496934 DOI: 10.3390/cells11182910] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 09/12/2022] [Accepted: 09/14/2022] [Indexed: 11/16/2022] Open
Abstract
The neurotransmitter GABA is normally characterized as having an inhibitory effect on neural activity in the adult central nervous system (CNS), which quells over-excitation and limits neural plasticity. Spinal cord injury (SCI) can bring about a modification that weakens the inhibitory effect of GABA in the central gray caudal to injury. This change is linked to the downregulation of the potassium/chloride cotransporter (KCC2) and the consequent rise in intracellular Cl- in the postsynaptic neuron. As the intracellular concentration increases, the inward flow of Cl- through an ionotropic GABA-A receptor is reduced, which decreases its hyperpolarizing (inhibitory) effect, a modulatory effect known as ionic plasticity. The loss of GABA-dependent inhibition enables a state of over-excitation within the spinal cord that fosters aberrant motor activity (spasticity) and chronic pain. A downregulation of KCC2 also contributes to the development of a number of brain-dependent pathologies linked to states of neural over-excitation, including epilepsy, addiction, and developmental disorders, along with other diseases such as hypertension, asthma, and irritable bowel syndrome. Pharmacological treatments that target ionic plasticity have been shown to bring therapeutic benefits.
Collapse
Affiliation(s)
- Kelsey E. Hudson
- Neuroscience, Texas A&M University, College Station, TX 77843, USA
| | - James W. Grau
- Psychological & Brain Sciences, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
15
|
Microglial Dynamics Modulate Vestibular Compensation in a Rodent Model of Vestibulopathy and Condition the Expression of Plasticity Mechanisms in the Deafferented Vestibular Nuclei. Cells 2022; 11:cells11172693. [PMID: 36078101 PMCID: PMC9454928 DOI: 10.3390/cells11172693] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Revised: 08/17/2022] [Accepted: 08/24/2022] [Indexed: 11/17/2022] Open
Abstract
Unilateral vestibular loss (UVL) induces a vestibular syndrome composed of posturo-locomotor, oculomotor, vegetative, and perceptivo-cognitive symptoms. With time, these functional deficits progressively disappear due to a phenomenon called vestibular compensation, known to be supported by the expression in the deafferented vestibular nuclei (VNs) of various adaptative plasticity mechanisms. UVL is known to induce a neuroinflammatory response within the VNs, thought to be caused by the structural alteration of primary vestibular afferents. The acute inflammatory response, expressed in the deafferented VNs was recently proven to be crucial for the expression of the endogenous plasticity supporting functional recovery. Neuroinflammation is supported by reactive microglial cells, known to have various phenotypes with adverse effects on brain tissue. Here, we used markers of pro-inflammatory and anti-inflammatory phenotypes of reactive microglia to study microglial dynamics following a unilateral vestibular neurectomy (UVN) in the adult rat. In addition, to highlight the role of acute inflammation in vestibular compensation and its underlying mechanisms, we enhanced the inflammatory state of the deafferented VNs using systemic injections of lipopolysaccharide (LPS) during the acute phase after a UVN. We observed that the UVN induced the expression of both M1 proinflammatory and M2 anti-inflammatory microglial phenotypes in the deafferented VNs. The acute LPS treatment exacerbated the inflammatory reaction and increased the M1 phenotype while decreasing M2 expression. These effects were associated with impaired postlesional plasticity in the deafferented VNs and exacerbated functional deficits. These results highlight the importance of a homeostatic inflammatory level in the expression of the adaptative plasticity mechanisms underlying vestibular compensation. Understanding the rules that govern neuroinflammation would provide therapeutic leads in neuropathologies associated with these processes.
Collapse
|
16
|
Yuan S, Wang N, Yao Y, Liu J, Gao M, Mo H, Zhang S, Su R. Role of 5-HT 2A receptor in modulating glutamatergic activity in the ventrolateral orbital cortex: implication in trigeminal neuralgia. Neuroscience 2022; 502:107-116. [PMID: 36038038 DOI: 10.1016/j.neuroscience.2022.08.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 08/02/2022] [Accepted: 08/23/2022] [Indexed: 11/16/2022]
Abstract
5-HT2A receptors (5-HT2ARs) are widely expressed in the central nervous system, including in the ventrolateral orbital cortex (VLO). The VLO is an important cortical component for pain processing. Brain 5-HT2ARs are implicated in both pro- and anti- nociceptive functions. However, the roles of 5-HT2ARs in the VLO in trigeminal neuralgia and neuronal synaptic function remain to be understood. We used chronic constriction injury of infraorbital nerve (IoN-CCI) model and shRNA mediated gene knockdown in mice to investigate the role of 5-HT2ARs in the VLO in trigeminal neuralgia. We found that knockdown of 5-HT2ARs in the VLO aggravated spontaneous pain and mechanical allodynia in mice after IoN-CCI. At the synaptic level, decreasing 5-HT2AR expression by shRNA or inhibition of 5-HT2AR activity by its antagonist ketanserin decreased the frequency and amplitude of spontaneous excitatory postsynaptic currents (sEPSCs) of the neurons in the VLO, whereas 5-HT2AR partial agonist 2,5-Dimethoxy-4-iodoamphetamine (DOI) enhanced sEPSCs of the neurons in the VLO. In summary, 5-HT2ARs in the VLO modulate the trigeminal pain by regulating neuronal glutamatergic activity.
Collapse
Affiliation(s)
- Shanshan Yuan
- Beijing Institute of Pharmacology and Toxicology, Beijing, China; School of Stomatology, Jiamusi University, Jiamusi, China
| | - Na Wang
- Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Yunxia Yao
- Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Junhong Liu
- Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Mingwei Gao
- Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Hongbing Mo
- School of Stomatology, Jiamusi University, Jiamusi, China
| | - Shuzhuo Zhang
- Beijing Institute of Pharmacology and Toxicology, Beijing, China.
| | - Ruibin Su
- Beijing Institute of Pharmacology and Toxicology, Beijing, China
| |
Collapse
|
17
|
Molecular Mechanisms of Epilepsy: The Role of the Chloride Transporter KCC2. J Mol Neurosci 2022; 72:1500-1515. [PMID: 35819636 DOI: 10.1007/s12031-022-02041-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 06/07/2022] [Indexed: 10/17/2022]
Abstract
Epilepsy is a neurological disease characterized by abnormal or synchronous brain activity causing seizures, which may produce convulsions, minor physical signs, or a combination of symptoms. These disorders affect approximately 65 million people worldwide, from all ages and genders. Seizures apart, epileptic patients present a high risk to develop neuropsychological comorbidities such as cognitive deficits, emotional disturbance, and psychiatric disorders, which severely impair quality of life. Currently, the treatment for epilepsy includes the administration of drugs or surgery, but about 30% of the patients treated with antiepileptic drugs develop time-dependent pharmacoresistence. Therefore, further investigation about epilepsy and its causes is needed to find new pharmacological targets and innovative therapeutic strategies. Pharmacoresistance is associated to changes in neuronal plasticity and alterations of GABAA receptor-mediated neurotransmission. The downregulation of GABA inhibitory activity may arise from a positive shift in GABAA receptor reversal potential, due to an alteration in chloride homeostasis. In this paper, we review the contribution of K+-Cl--cotransporter (KCC2) to the alterations in the Cl- gradient observed in epileptic condition, and how these alterations are coupled to the increase in the excitability.
Collapse
|
18
|
Hui KK, Chater TE, Goda Y, Tanaka M. How Staying Negative Is Good for the (Adult) Brain: Maintaining Chloride Homeostasis and the GABA-Shift in Neurological Disorders. Front Mol Neurosci 2022; 15:893111. [PMID: 35875665 PMCID: PMC9305173 DOI: 10.3389/fnmol.2022.893111] [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: 03/10/2022] [Accepted: 06/10/2022] [Indexed: 01/27/2023] Open
Abstract
Excitatory-inhibitory (E-I) imbalance has been shown to contribute to the pathogenesis of a wide range of neurodevelopmental disorders including autism spectrum disorders, epilepsy, and schizophrenia. GABA neurotransmission, the principal inhibitory signal in the mature brain, is critically coupled to proper regulation of chloride homeostasis. During brain maturation, changes in the transport of chloride ions across neuronal cell membranes act to gradually change the majority of GABA signaling from excitatory to inhibitory for neuronal activation, and dysregulation of this GABA-shift likely contributes to multiple neurodevelopmental abnormalities that are associated with circuit dysfunction. Whilst traditionally viewed as a phenomenon which occurs during brain development, recent evidence suggests that this GABA-shift may also be involved in neuropsychiatric disorders due to the "dematuration" of affected neurons. In this review, we will discuss the cell signaling and regulatory mechanisms underlying the GABA-shift phenomenon in the context of the latest findings in the field, in particular the role of chloride cotransporters NKCC1 and KCC2, and furthermore how these regulatory processes are altered in neurodevelopmental and neuropsychiatric disorders. We will also explore the interactions between GABAergic interneurons and other cell types in the developing brain that may influence the GABA-shift. Finally, with a greater understanding of how the GABA-shift is altered in pathological conditions, we will briefly outline recent progress on targeting NKCC1 and KCC2 as a therapeutic strategy against neurodevelopmental and neuropsychiatric disorders associated with improper chloride homeostasis and GABA-shift abnormalities.
Collapse
Affiliation(s)
- Kelvin K. Hui
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, United States
- Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Thomas E. Chater
- Laboratory for Synaptic Plasticity and Connectivity, RIKEN Center for Brain Science, Wako, Japan
| | - Yukiko Goda
- Laboratory for Synaptic Plasticity and Connectivity, RIKEN Center for Brain Science, Wako, Japan
- Synapse Biology Unit, Okinawa Institute for Science and Technology Graduate University, Onna, Japan
| | - Motomasa Tanaka
- Laboratory for Protein Conformation Diseases, RIKEN Center for Brain Science, Wako, Japan
| |
Collapse
|
19
|
Talifu Z, Qin C, Xin Z, Chen Y, Liu J, Dangol S, Ma X, Gong H, Pei Z, Yu Y, Li J, Du L. The Overexpression of Insulin-Like Growth Factor-1 and Neurotrophin-3 Promote Functional Recovery and Alleviate Spasticity After Spinal Cord Injury. Front Neurosci 2022; 16:863793. [PMID: 35573286 PMCID: PMC9099063 DOI: 10.3389/fnins.2022.863793] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 03/30/2022] [Indexed: 11/22/2022] Open
Abstract
Objective This study was conducted to investigate the effects of the exogenous overexpression of nerve growth factors NT-3 and IGF-1 on the recovery of nerve function after spinal cord injury (SCI) and identify the potential mechanism involved. Methods Sixty-four female SD rats were randomly divided into four groups: an SCI group, an adeno-associated viral (AAV)-RFP and AAV-GFP injection group, an AAV-IGF-1 and AAV-NT-3 injection group, and a Sham group. After grouping, the rats were subjected to a 10-week electrophysiological and behavioral evaluation to comprehensively evaluate the effects of the intervention on motor function, spasticity, mechanical pain, and thermal pain. Ten weeks later, samples were taken for immunofluorescence (IF) staining and Western blot (WB) detection, focusing on the expression of KCC2, 5-HT2A, and 5-HT2C receptors in motor neurons and the spinal cord. Results Electrophysiological and behavioral data indicated that the AAV-IGF-1 and AAV-NT-3 groups showed better recovery of motor function (P < 0.05 from D14 compared with the AAV-RFP + AAV-GFP group; P < 0.05 from D42 compared with SCI group) and less spasticity (4-10 weeks, at 5 Hz all P < 0.05 compared with SCI group and AAV- RFP + AAV-GFP group) but with a trend for more pain sensitivity. Compared with the SCI group, the von Frey value result of the AAV-IGF-1 and AAV-NT-3 groups showed a lower pain threshold (P < 0.05 at 4-8 weeks), and shorter thermal pain threshold (P < 0.05 at 8-10 weeks). IF staining further suggested that compared with the SCI group, the overexpression of NT-3 and IGF-1 in the SCI-R + G group led to increased levels of KCC2 (p < 0.05), 5-HT2A (p < 0.05), and 5-HT2C (p < 0.001) in motor neurons. WB results showed that compared with the SCI group, the SCI-R + G group exhibited higher expression levels of CHAT (p < 0.01), 5-HT2A (p < 0.05), and 5-HT2C (p < 0.05) proteins in the L2-L6 lumbar enlargement. Conclusion Data analysis showed that the overexpression of NT-3 and IGF-1 may improve motor function after SCI and alleviate spasms in a rat model; however, these animals were more sensitive to mechanical pain and thermal pain. These behavioral changes may be related to increased numbers of KCC2, 5-HT2A, and 5-HT2C receptors in the spinal cord tissue. The results of this study may provide a new theoretical basis for the clinical treatment of SCI.
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
| | - Chuan Qin
- Department of Urology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Zhang Xin
- School of Sports Medicine and Rehabilitation, Beijing Sport University, Beijing, China
| | - Yixin Chen
- 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
- Department of Rehabilitation Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Jiayi Liu
- 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
| | - Subarna Dangol
- 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
| | - Xiaodong Ma
- 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
| | - 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
| | - Zhisheng Pei
- 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
| | - 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
| | - 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
| | - Liangjie Du
- 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
| |
Collapse
|
20
|
Yuan XC, Wang YY, Tian LX, Yan XJ, Guo YX, Zhao YL, Baba SS, Jia H, Wang HS, Li M, Huo FQ. Spinal 5-HT2A Receptor is Involved in Electroacupuncture Inhibition of Chronic Pain. Mol Pain 2022; 18:17448069221087583. [PMID: 35240891 PMCID: PMC9006364 DOI: 10.1177/17448069221087583] [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] [Indexed: 11/20/2022] Open
Abstract
Knee osteoarthritis (KOA) is a highly prevalent, chronic joint disorder, and it is a typical disease which can develop chronic pain. Our previous study has proved that endocannabinoid (2-AG)-CB1R-GABA-5-HT pathway is involved in electroacupuncture (EA) mediated inhibition of chronic pain. However, it is still unclear which among the 5-HT receptor subtype is involved in EA evoked 5-HT mediated inhibition of chronic pain in the dorsal spinal cord. 5-HT2A is a G protein-coupled receptor and it is involved in 5-HT descending pain modulation system. We found that EA treatment at frequency of 2 Hz +1 mA significantly increased the expression of 5-HT2A receptor in the dorsal spinal cord and intrathecal injection of 5-HT2A receptor antagonist or agonist reversed or mimicked the analgesic effect of EA in each case respectively. Intrathecal injection of a selective GABAA receptor antagonist Bicuculline also reversed the EA effect on pain hypersensitivity. Additionally, EA treatment reversed the reduced expression of GABAA receptor and KCC2 in the dorsal spinal cord of KOA mice. Furthermore, we demonstrated that intrathecal 5-HT2A receptor antagonist/agonist reversed or mimicked the effect of EA up-regulate of KCC2 expression, respectively. Similarly, intrathecal injection of PLC and PKC inhibitors prevented both anti-allodynic effect and up-regulation of KCC2 expression by EA treatment. Our data suggest that EA treatment up-regulated KCC2 expression through activating 5-HT2A-Gq-PLC-PKC pathway and enhanced the inhibitory function of GABAA receptor, thereby inhibiting chronic pain in a mouse model of KOA.
Collapse
Affiliation(s)
| | | | - Li-Xia Tian
- Department of Physiology and Pathophysiology
| | | | - Yi-Xiao Guo
- Department of Physiology and Pathophysiology
| | | | | | | | | | - Man Li
- Department of Neurobiology
| | - Fu-Quan Huo
- Department of Physiology and Pathophysiology
| |
Collapse
|
21
|
Kimmey BA, Wittenberg RE, Croicu A, Shadani N, Ostroumov A, Dani JA. The serotonin 2A receptor agonist TCB-2 attenuates heavy alcohol drinking and alcohol-induced midbrain inhibitory plasticity. Addict Biol 2022; 27:e13147. [PMID: 35229942 PMCID: PMC8896307 DOI: 10.1111/adb.13147] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 11/26/2021] [Accepted: 01/10/2022] [Indexed: 12/21/2022]
Abstract
Disruption of neuronal chloride ion (Cl- ) homeostasis has been linked to several pathological conditions, including substance use disorder, yet targeted pharmacotherapies are lacking. In this study, we explored the potential of serotonin 2A receptor (5-HT2A R) agonism to reduce alcohol consumption in male wild-type C57Bl/6J mice and to ameliorate alcohol-induced inhibitory plasticity in the midbrain. We found that administration of the putative 5-HT2A R agonist TCB-2 attenuated alcohol consumption and preference but did not alter water or saccharin consumption. We hypothesized that the selective behavioural effects of TCB-2 on alcohol drinking were due, at least in part, to effects of the agonist on ventral tegmental area (VTA) neurocircuitry. Alcohol consumption impairs Cl- transport in VTA GABA neurons, which acts as a molecular adaptation leading to increased alcohol self-administration. Using ex vivo electrophysiological recordings, we found that exposure to either intermittent volitional alcohol drinking or an acute alcohol injection diminished homeostatic Cl- transport in VTA GABA neurons. Critically, in vivo TCB-2 administration normalized Cl- transport in the VTA after alcohol exposure. Thus, we show a potent effect of alcohol consumption on VTA inhibitory circuitry, in the form of dysregulated Cl- homeostasis that is reversible with agonism of 5-HT2A Rs. Our results provide insight into the potential therapeutic action of 5-HT2A R agonists for alcohol abuse.
Collapse
Affiliation(s)
| | | | | | | | - Alexey Ostroumov
- Co-corresponding authors: Alexey Ostroumov, Ph.D., Georgetown University, Department of Pharmacology and Physiology, New Research Building, Room W226, 3970 Reservoir Road, N.W., Washington D.C. 20057, USA, Phone: (832) 641-5562, , John A. Dani, Ph.D., University of Pennsylvania, Department of Neuroscience, Clinical Research Building, Room 211, 415 Curie Boulevard, Philadelphia, P.A. 19104, USA, Phone: (215) 898-8498,
| | - John A. Dani
- Co-corresponding authors: Alexey Ostroumov, Ph.D., Georgetown University, Department of Pharmacology and Physiology, New Research Building, Room W226, 3970 Reservoir Road, N.W., Washington D.C. 20057, USA, Phone: (832) 641-5562, , John A. Dani, Ph.D., University of Pennsylvania, Department of Neuroscience, Clinical Research Building, Room 211, 415 Curie Boulevard, Philadelphia, P.A. 19104, USA, Phone: (215) 898-8498,
| |
Collapse
|
22
|
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
|
23
|
Cherubini E, Di Cristo G, Avoli M. Dysregulation of GABAergic Signaling in Neurodevelomental Disorders: Targeting Cation-Chloride Co-transporters to Re-establish a Proper E/I Balance. Front Cell Neurosci 2022; 15:813441. [PMID: 35069119 PMCID: PMC8766311 DOI: 10.3389/fncel.2021.813441] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 11/30/2021] [Indexed: 01/01/2023] Open
Abstract
The construction of the brain relies on a series of well-defined genetically and experience- or activity -dependent mechanisms which allow to adapt to the external environment. Disruption of these processes leads to neurological and psychiatric disorders, which in many cases are manifest already early in postnatal life. GABA, the main inhibitory neurotransmitter in the adult brain is one of the major players in the early assembly and formation of neuronal circuits. In the prenatal and immediate postnatal period GABA, acting on GABAA receptors, depolarizes and excites targeted cells via an outwardly directed flux of chloride. In this way it activates NMDA receptors and voltage-dependent calcium channels contributing, through intracellular calcium rise, to shape neuronal activity and to establish, through the formation of new synapses and elimination of others, adult neuronal circuits. The direction of GABAA-mediated neurotransmission (depolarizing or hyperpolarizing) depends on the intracellular levels of chloride [Cl−]i, which in turn are maintained by the activity of the cation-chloride importer and exporter KCC2 and NKCC1, respectively. Thus, the premature hyperpolarizing action of GABA or its persistent depolarizing effect beyond the postnatal period, leads to behavioral deficits associated with morphological alterations and an excitatory (E)/inhibitory (I) imbalance in selective brain areas. The aim of this review is to summarize recent data concerning the functional role of GABAergic transmission in building up and refining neuronal circuits early in development and its dysfunction in neurodevelopmental disorders such as Autism Spectrum Disorders (ASDs), schizophrenia and epilepsy. In particular, we focus on novel information concerning the mechanisms by which alterations in cation-chloride co-transporters (CCC) generate behavioral and cognitive impairment in these diseases. We discuss also the possibility to re-establish a proper GABAA-mediated neurotransmission and excitatory (E)/inhibitory (I) balance within selective brain areas acting on CCC.
Collapse
Affiliation(s)
- Enrico Cherubini
- European Brain Research Institute (EBRI)-Rita Levi-Montalcini, Roma, Italy
- *Correspondence: Enrico Cherubini
| | - Graziella Di Cristo
- Neurosciences Department, Université de Montréal and CHU Sainte-Justine Research Center, Montreal, QC, Canada
| | - Massimo Avoli
- Montreal Neurological Institute-Hospital and Departments of Neurology and Neurosurgery and of Physiology, McGill University, Montreal, QC, Canada
| |
Collapse
|
24
|
Branchereau P, Cattaert D. Chloride Homeostasis in Developing Motoneurons. ADVANCES IN NEUROBIOLOGY 2022; 28:45-61. [PMID: 36066820 DOI: 10.1007/978-3-031-07167-6_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Maturation of GABA/Glycine chloride-mediated synaptic inhibitions is crucial for the establishment of a balance between excitation and inhibition. GABA and glycine are excitatory neurotransmitters on immature neurons that exhibit elevated [Cl-]i. Later in development [Cl-]i drops leading to the occurrence of inhibitory synaptic activity. This ontogenic change is closely correlated to a differential expression of two cation-chloride cotransporters that are the Cl- channel K+/Cl- co-transporter type 2 (KCC2) that extrudes Cl- ions and the Na+-K+-2Cl- cotransporter NKCC1 that accumulates Cl- ions. The classical scheme built from studies performed on cortical and hippocampal networks proposes that immature neurons display high [Cl-]i because NKCC1 is overexpressed compared to KCC2 and that the co-transporters ratio reverses in mature neurons, lowering [Cl-]i. In this chapter, we will see that this classical scheme is not true in motoneurons (MNs) and that an early alteration of the chloride homeostasis may be involved in pathological conditions.
Collapse
Affiliation(s)
- Pascal Branchereau
- Institut de Neurosciences Cognitives et Intégratives d'Aquitaine (INCIA), Univ. Bordeaux, UMR 5287, CNRS, Bordeaux, France.
| | - Daniel Cattaert
- Institut de Neurosciences Cognitives et Intégratives d'Aquitaine (INCIA), Univ. Bordeaux, UMR 5287, CNRS, Bordeaux, France
| |
Collapse
|
25
|
Asraf H, Bogdanovic M, Gottesman N, Sekler I, Aizenman E, Hershfinkel M. SNAP23 regulates KCC2 membrane insertion and activity following mZnR/GPR39 activation in hippocampal neurons. iScience 2022; 25:103751. [PMID: 35118363 PMCID: PMC8800107 DOI: 10.1016/j.isci.2022.103751] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 11/25/2021] [Accepted: 01/06/2022] [Indexed: 11/23/2022] Open
Abstract
Modulation of the neuronal K+/Cl− cotransporter 2 (KCC2) activity, which mediates Cl− export, is critical to neuronal function. Here, we demonstrate that KCC2 interacts with the SNARE protein synaptosome-associated protein 23, SNAP23, an essential component of membrane insertion machinery. Using KCC2 truncated mutants, we show that KCC2 C-terminal domain is essential for membrane targeting and SNAP23-dependent upregulation of KCC2 activity triggered by activation of the Zn2+-sensitive receptor mZnR/GPR39 in HEK293 cells. Expression of SNAP23 phosphorylation-insensitive mutants or inhibition of its upstream activator IκB kinase (IKK) prevents mZnR/GPR39 upregulation of KCC2 activity in mouse hippocampal neurons. We further find that SNAP23 interacts with Syntaxin 1A and KCC2, and that all three proteins exhibit increased membrane insertion following mZnR/GPR39 activation in neurons. Our results elucidate a G-protein-coupled receptor-dependent pathway for regulation of KCC activity, mediated via interaction with SNARE proteins. Neuronal K+/Cl− cotransporter 2 (KCC2) is regulated via interaction with SNAP23 Zn2+ enhances interaction and membrane insertion of SNAP23, Syntaxin 1A, and KCC2 Zn2+-dependent mZnR/GPR39 regulation of KCC2 requires SNAP23 phosphorylation Epithelial KCC3 regulation by ZnR/GPR39 also requires SNAP23
Collapse
|
26
|
Lim WM, Chin EWM, Tang BL, Chen T, Goh ELK. WNK3 Maintains the GABAergic Inhibitory Tone, Synaptic Excitation and Neuronal Excitability via Regulation of KCC2 Cotransporter in Mature Neurons. Front Mol Neurosci 2021; 14:762142. [PMID: 34858138 PMCID: PMC8631424 DOI: 10.3389/fnmol.2021.762142] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Accepted: 10/14/2021] [Indexed: 11/13/2022] Open
Abstract
The activation of chloride (Cl−)permeable gamma (γ)-aminobutyric acid type A(GABAA) receptors induces synaptic inhibition in mature and excitation in immature neurons. This developmental “switch” in GABA function controlled by its polarity depends on the postnatal decrease in intraneuronal Cl− concentration mediated by KCC2, a member of cation-chloride cotransporters (CCCs). The serine-threonine kinase WNK3 (With No Lysine [K]), is a potent regulator of all CCCs and is expressed in neurons. Here, we characterized the functions of WNK3 and its role in GABAergic signaling in cultured embryonic day 18 (E18) hippocampal neurons. We observed a decrease in WNK3 expression as neurons mature. Knocking down of WNK3 significantly hyperpolarized EGABA in mature neurons (DIV13–15) but had no effect on immature neurons (DIV6–8). This hyperpolarized EGABA in WNK3-deficient neurons was not due to the total expression of NKCC1 and KCC2, that remained unchanged. However, there was a reduction in phosphorylated KCC2 at the membrane, suggesting an increase in KCC2 chloride export activity. Furthermore, hyperpolarized EGABA observed in WNK3-deficient neurons can be reversed by the KCC2 inhibitor, VU024055, thus indicating that WNK3 acts through KCC2 to influence EGABA. Notably, WNK3 knockdown resulted in morphological changes in mature but not immature neurons. Electrophysiological characterization of WNK3-deficient mature neurons revealed reduced capacitances but increased intrinsic excitability and synaptic excitation. Hence, our study demonstrates that WNK3 maintains the “adult” GABAergic inhibitory tone in neurons and plays a role in the morphological development of neurons and excitability.
Collapse
Affiliation(s)
- Wee Meng Lim
- Neuroscience Academic Clinical Programme, Duke-NUS Medical School, Singapore, Singapore.,NUS Graduate School for Integrative Sciences and Engineering, Singapore, Singapore
| | - Eunice W M Chin
- Neuroscience Academic Clinical Programme, Duke-NUS Medical School, Singapore, Singapore.,NUS Graduate School for Integrative Sciences and Engineering, Singapore, Singapore.,Neuroscience and Mental Health Faculty, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Bor Luen Tang
- NUS Graduate School for Integrative Sciences and Engineering, Singapore, Singapore.,Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Tingting Chen
- School of Pharmacy, Nantong University, Nantong, China
| | - Eyleen L K Goh
- Neuroscience Academic Clinical Programme, Duke-NUS Medical School, Singapore, Singapore.,Neuroscience and Mental Health Faculty, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| |
Collapse
|
27
|
The Alteration of Chloride Homeostasis/GABAergic Signaling in Brain Disorders: Could Oxidative Stress Play a Role? Antioxidants (Basel) 2021; 10:antiox10081316. [PMID: 34439564 PMCID: PMC8389245 DOI: 10.3390/antiox10081316] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 08/18/2021] [Accepted: 08/19/2021] [Indexed: 12/22/2022] Open
Abstract
In neuronal precursors and immature neurons, the depolarizing (excitatory) effect of γ-Aminobutyric acid (GABA) signaling is associated with elevated [Cl−]i; as brain cells mature, a developmental switch occurs, leading to the decrease of [Cl−]i and to the hyperpolarizing (inhibitory) effect of GABAergic signaling. [Cl−]i is controlled by two chloride co-transporters: NKCC1, which causes Cl− to accumulate into the cells, and KCC2, which extrudes it. The ontogenetic upregulation of the latter determines the above-outlined switch; however, many other factors contribute to the correct [Cl−]i in mature neurons. The dysregulation of chloride homeostasis is involved in seizure generation and has been associated with schizophrenia, Down’s Syndrome, Autism Spectrum Disorder, and other neurodevelopmental disorders. Recently, much effort has been put into developing new drugs intended to inhibit NKCC1 activity, while no attention has been paid to the origin of [Cl−]i dysregulation. Our study examines the pathophysiology of Cl− homeostasis and focuses on the impact of oxidative stress (OS) and inflammation on the activity of Cl− co-transporters, highlighting the relevance of OS in numerous brain abnormalities and diseases. This hypothesis supports the importance of primary prevention during pregnancy. It also integrates the therapeutic framework addressed to restore normal GABAergic signaling by counteracting the alteration in chloride homeostasis in central nervous system (CNS) cells, aiming at limiting the use of drugs that potentially pose a health risk.
Collapse
|
28
|
El Mahmoudi N, Rastoldo G, Marouane E, Péricat D, Watabe I, Tonetto A, Hautefort C, Chabbert C, Sargolini F, Tighilet B. Breaking a dogma: acute anti-inflammatory treatment alters both post-lesional functional recovery and endogenous adaptive plasticity mechanisms in a rodent model of acute peripheral vestibulopathy. J Neuroinflammation 2021; 18:183. [PMID: 34419105 PMCID: PMC8380392 DOI: 10.1186/s12974-021-02222-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 07/19/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Due to their anti-inflammatory action, corticosteroids are the reference treatment for brain injuries and many inflammatory diseases. However, the benefits of acute corticotherapy are now being questioned, particularly in the case of acute peripheral vestibulopathies (APV), characterized by a vestibular syndrome composed of sustained spinning vertigo, spontaneous ocular nystagmus and oscillopsia, perceptual-cognitive, posturo-locomotor, and vegetative disorders. We assessed the effectiveness of acute corticotherapy, and the functional role of acute inflammation observed after sudden unilateral vestibular loss. METHODS We used the rodent model of unilateral vestibular neurectomy, mimicking the syndrome observed in patients with APV. We treated the animals during the acute phase of the vestibular syndrome, either with placebo or methylprednisolone, an anti-inflammatory corticosteroid. At the cellular level, impacts of methylprednisolone on endogenous plasticity mechanisms were assessed through analysis of cell proliferation and survival, glial reactions, neuron's membrane excitability, and stress marker. At the behavioral level, vestibular and posturo-locomotor functions' recovery were assessed with appropriate qualitative and quantitative evaluations. RESULTS We observed that acute treatment with methylprednisolone significantly decreases glial reactions, cell proliferation and survival. In addition, stress and excitability markers were significantly impacted by the treatment. Besides, vestibular syndrome's intensity was enhanced, and vestibular compensation delayed under acute methylprednisolone treatment. CONCLUSIONS We show here, for the first time, that acute anti-inflammatory treatment alters the expression of the adaptive plasticity mechanisms in the deafferented vestibular nuclei and generates enhanced and prolonged vestibular and postural deficits. These results strongly suggest a beneficial role for acute endogenous neuroinflammation in vestibular compensation. They open the way to a change in dogma for the treatment and therapeutic management of vestibular patients.
Collapse
Affiliation(s)
- Nada El Mahmoudi
- Aix-Marseille Université-CNRS, Laboratoire de Neurosciences Cognitives, LNC UMR 7291, Centre Saint Charles, Case C; 3 Place Victor Hugo, 13331, Marseille Cedex 03, France
- Centre Saint-Charles, Aix-Marseille Université CNRS, Case C; 3 Place Victor Hugo, 13331, Marseille Cedex 03, France
| | - Guillaume Rastoldo
- Aix-Marseille Université-CNRS, Laboratoire de Neurosciences Cognitives, LNC UMR 7291, Centre Saint Charles, Case C; 3 Place Victor Hugo, 13331, Marseille Cedex 03, France
- Centre Saint-Charles, Aix-Marseille Université CNRS, Case C; 3 Place Victor Hugo, 13331, Marseille Cedex 03, France
| | - Emna Marouane
- Aix-Marseille Université-CNRS, Laboratoire de Neurosciences Cognitives, LNC UMR 7291, Centre Saint Charles, Case C; 3 Place Victor Hugo, 13331, Marseille Cedex 03, France
- Centre Saint-Charles, Aix-Marseille Université CNRS, Case C; 3 Place Victor Hugo, 13331, Marseille Cedex 03, France
| | - David Péricat
- Institut de Pharmacologie Et de Biologie Structurale, Université de Toulouse Paul Sabatier-CNRS, Toulouse, France
| | - Isabelle Watabe
- Aix-Marseille Université-CNRS, Laboratoire de Neurosciences Cognitives, LNC UMR 7291, Centre Saint Charles, Case C; 3 Place Victor Hugo, 13331, Marseille Cedex 03, France
- Centre Saint-Charles, Aix-Marseille Université CNRS, Case C; 3 Place Victor Hugo, 13331, Marseille Cedex 03, France
| | - Alain Tonetto
- Centrale Marseille, FSCM (FR 1739), PRATIM, Aix Marseille Université-CNRS, 13397, Marseille, France
| | - Charlotte Hautefort
- Department of Head and Neck Surgery, Lariboisière University Hospital, Paris, France
| | - Christian Chabbert
- Aix-Marseille Université-CNRS, Laboratoire de Neurosciences Cognitives, LNC UMR 7291, Centre Saint Charles, Case C; 3 Place Victor Hugo, 13331, Marseille Cedex 03, France
- Centre Saint-Charles, Aix-Marseille Université CNRS, Case C; 3 Place Victor Hugo, 13331, Marseille Cedex 03, France
- GDR Physiopathologie Vestibulaire-Unité GDR2074 CNRS, Marseille, France
| | - Francesca Sargolini
- Aix-Marseille Université-CNRS, Laboratoire de Neurosciences Cognitives, LNC UMR 7291, Centre Saint Charles, Case C; 3 Place Victor Hugo, 13331, Marseille Cedex 03, France
| | - Brahim Tighilet
- Aix-Marseille Université-CNRS, Laboratoire de Neurosciences Cognitives, LNC UMR 7291, Centre Saint Charles, Case C; 3 Place Victor Hugo, 13331, Marseille Cedex 03, France.
- GDR Physiopathologie Vestibulaire-Unité GDR2074 CNRS, Marseille, France.
| |
Collapse
|
29
|
Worthington A, Kalteniece A, Ferdousi M, D'Onofrio L, Dhage S, Azmi S, Adamson C, Hamdy S, Malik RA, Calcutt NA, Marshall AG. Spinal Inhibitory Dysfunction in Patients With Painful or Painless Diabetic Neuropathy. Diabetes Care 2021; 44:1835-1841. [PMID: 34385346 DOI: 10.2337/dc20-2797] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 05/13/2021] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Impaired rate-dependent depression of the Hoffman reflex (HRDD) is a marker of spinal inhibitory dysfunction and has previously been associated with painful neuropathy in a proof-of-concept study in patients with type 1 diabetes. We have now undertaken an assessment of HRDD in patients with type 1 or type 2 diabetes. RESEARCH DESIGN AND METHODS A total of 148 participants, including 34 healthy control subjects, 42 patients with painful diabetic neuropathy, and 62 patients with diabetic neuropathy without pain, underwent an assessment of HRDD and a detailed assessment of peripheral neuropathy, including nerve conduction studies, corneal confocal microscopy, and thermal threshold testing. RESULTS Compared with healthy control subjects (P < 0.001) and patients without pain (P < 0.001), we found that HRDD is impaired in patients with type 1 or type 2 diabetes with neuropathic pain. These impairments are unrelated to diabetes type and the presence or severity of neuropathy. In contrast, patients without neuropathic pain (P < 0.05) exhibited enhanced HRDD compared with control subjects. CONCLUSIONS We suggest that loss or impairment of HRDD may help to identify a subpopulation of patients with painful diabetic neuropathy mediated by impaired spinal inhibitory systems who may respond optimally to therapies that target spinal or supraspinal mechanisms. Enhanced RDD in patients without pain may reflect engagement of spinal pain-suppressing mechanisms.
Collapse
Affiliation(s)
- Anne Worthington
- Division of Diabetes, Endocrinology and Gastroenterology, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, U.K
| | - Alise Kalteniece
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, U.K
| | - Maryam Ferdousi
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, U.K
| | - Luca D'Onofrio
- Department of Experimental Medicine, Sapienza University, Rome, Italy
| | - Shaishav Dhage
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, U.K
| | - Shazli Azmi
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, U.K.,Diabetes Centre, Manchester University NHS Foundation Trust, Manchester, U.K
| | - Clare Adamson
- Diabetes Centre, Manchester University NHS Foundation Trust, Manchester, U.K
| | - Shaheen Hamdy
- Centre for Gastrointestinal Sciences, Division of Diabetes and Endocrinology, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, U.K
| | - Rayaz A Malik
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, U.K.,Weill Cornell Medicine-Qatar, Research Division, Qatar Foundation, Education City
| | - Nigel A Calcutt
- Department of Pathology, University of California, San Diego, La Jolla, CA
| | - Andrew G Marshall
- Division of Neuroscience and Experimental Psychology, Faculty of Medical and Human Sciences, University of Manchester, Manchester, U.K. .,Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, U.K
| |
Collapse
|
30
|
Bouali-Benazzouz R, Landry M, Benazzouz A, Fossat P. Neuropathic pain modeling: Focus on synaptic and ion channel mechanisms. Prog Neurobiol 2021; 201:102030. [PMID: 33711402 DOI: 10.1016/j.pneurobio.2021.102030] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Accepted: 02/22/2021] [Indexed: 12/28/2022]
Abstract
Animal models of pain consist of modeling a pain-like state and measuring the consequent behavior. The first animal models of neuropathic pain (NP) were developed in rodents with a total lesion of the sciatic nerve. Later, other models targeting central or peripheral branches of nerves were developed to identify novel mechanisms that contribute to persistent pain conditions in NP. Objective assessment of pain in these different animal models represents a significant challenge for pre-clinical research. Multiple behavioral approaches are used to investigate and to validate pain phenotypes including withdrawal reflex to evoked stimuli, vocalizations, spontaneous pain, but also emotional and affective behaviors. Furthermore, animal models were very useful in investigating the mechanisms of NP. This review will focus on a detailed description of rodent models of NP and provide an overview of the assessment of the sensory and emotional components of pain. A detailed inventory will be made to examine spinal mechanisms involved in NP-induced hyperexcitability and underlying the current pharmacological approaches used in clinics with the possibility to present new avenues for future treatment. The success of pre-clinical studies in this area of research depends on the choice of the relevant model and the appropriate test based on the objectives of the study.
Collapse
Affiliation(s)
- Rabia Bouali-Benazzouz
- Université de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France.
| | - Marc Landry
- Université de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France
| | - Abdelhamid Benazzouz
- Université de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France
| | - Pascal Fossat
- Université de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France
| |
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
|
Sergeeva EG, Rosenberg PA, Benowitz LI. Non-Cell-Autonomous Regulation of Optic Nerve Regeneration by Amacrine Cells. Front Cell Neurosci 2021; 15:666798. [PMID: 33935656 PMCID: PMC8085350 DOI: 10.3389/fncel.2021.666798] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 03/19/2021] [Indexed: 11/13/2022] Open
Abstract
Visual information is conveyed from the eye to the brain through the axons of retinal ganglion cells (RGCs) that course through the optic nerve and synapse onto neurons in multiple subcortical visual relay areas. RGCs cannot regenerate their axons once they are damaged, similar to most mature neurons in the central nervous system (CNS), and soon undergo cell death. These phenomena of neurodegeneration and regenerative failure are widely viewed as being determined by cell-intrinsic mechanisms within RGCs or to be influenced by the extracellular environment, including glial or inflammatory cells. However, a new concept is emerging that the death or survival of RGCs and their ability to regenerate axons are also influenced by the complex circuitry of the retina and that the activation of a multicellular signaling cascade involving changes in inhibitory interneurons - the amacrine cells (AC) - contributes to the fate of RGCs. Here, we review our current understanding of the role that interneurons play in cell survival and axon regeneration after optic nerve injury.
Collapse
Affiliation(s)
- Elena G. Sergeeva
- Department of Neurology, Boston Children’s Hospital, Boston, MA, United States
- Kirby Center for Neuroscience, Boston Children’s Hospital, Boston, MA, United States
- Department of Neurology, Harvard Medical School, Boston, MA, United States
| | - Paul A. Rosenberg
- Department of Neurology, Boston Children’s Hospital, Boston, MA, United States
- Kirby Center for Neuroscience, Boston Children’s Hospital, Boston, MA, United States
- Department of Neurology, Harvard Medical School, Boston, MA, United States
| | - Larry I. Benowitz
- Kirby Center for Neuroscience, Boston Children’s Hospital, Boston, MA, United States
- Laboratories for Neuroscience Research in Neurosurgery, Boston Children’s Hospital, Boston, MA, United States
- Department of Neurosurgery, Boston Children’s Hospital, Boston, MA, United States
- Department of Neurosurgery, Harvard Medical School, Boston, MA, United States
- Department of Ophthalmology, Harvard Medical School, Boston, MA, United States
| |
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
|
Tsuda S, Golam M, Hou J, Nelson R, Bernavil P, Richardson K, Wang KKW, Thompson F, Bose P. Altered monoaminergic levels, spasticity, and balance disability following repetitive blast-induced traumatic brain injury in rats. Brain Res 2020; 1747:147060. [PMID: 32828734 PMCID: PMC10424094 DOI: 10.1016/j.brainres.2020.147060] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 07/28/2020] [Accepted: 08/13/2020] [Indexed: 12/14/2022]
Abstract
Spasticity and balance disability are major complications following traumatic brain injury (TBI). Although monoaminergic inputs provide critical adaptive neuromodulations to the motor system, data are not available regarding the levels of monoamines in the brain regions related to motor functions following repetitive blast TBI (bTBI). The objective of this study was to determine if mild, repetitive bTBI results in spasticity/balance deficits and if these are correlated with altered levels of norepinephrine, dopamine, and serotonin in the brain regions related to the motor system. Repetitive bTBI was induced by a blast overpressure wave in male rats on days 1, 4, and 7. Following bTBI, physiological/behavioral tests were conducted and tissues in the central motor system (i.e., motor cortex, locus coeruleus, vestibular nuclei, and lumbar spinal cord) were collected for electrochemical detection of norepinephrine, dopamine, and serotonin by high-performance liquid chromatography. The results showed that norepinephrine was significantly increased in the locus coeruleus and decreased in the vestibular nuclei, while dopamine was significantly decreased in the vestibular nuclei. On the other hand, serotonin was significantly increased in the motor cortex and the lumbar spinal cord. Because these monoamines play important roles in regulating the excitability of neurons, these results suggest that mild, repetitive bTBI-induced dysregulation of monoaminergic inputs in the central motor system could contribute to spasticity and balance disability. This is the first study to report altered levels of multiple monoamines in the central motor system following acute mild, repetitive bTBI.
Collapse
Affiliation(s)
- Shigeharu Tsuda
- Brain Rehabilitation Research Center, Malcom Randall VA Medical Center, North Florida/South Georgia Veterans Health System, 1601 SW Archer Rd, Gainesville 32608-1197, USA; Department of Anesthesiology, College of Medicine, University of Florida, 1600 SW Archer Rd m509, Gainesville, FL 32610-0254, USA
| | - Mustafa Golam
- Brain Rehabilitation Research Center, Malcom Randall VA Medical Center, North Florida/South Georgia Veterans Health System, 1601 SW Archer Rd, Gainesville 32608-1197, USA; Department of Physiological Sciences, University of Florida, 1333 Center Dr, Gainesville, FL 32603, USA
| | - Jiamei Hou
- Brain Rehabilitation Research Center, Malcom Randall VA Medical Center, North Florida/South Georgia Veterans Health System, 1601 SW Archer Rd, Gainesville 32608-1197, USA; Department of Anesthesiology, College of Medicine, University of Florida, 1600 SW Archer Rd m509, Gainesville, FL 32610-0254, USA
| | - Rachel Nelson
- Brain Rehabilitation Research Center, Malcom Randall VA Medical Center, North Florida/South Georgia Veterans Health System, 1601 SW Archer Rd, Gainesville 32608-1197, USA
| | - Phillip Bernavil
- Brain Rehabilitation Research Center, Malcom Randall VA Medical Center, North Florida/South Georgia Veterans Health System, 1601 SW Archer Rd, Gainesville 32608-1197, USA
| | - Kenneth Richardson
- Brain Rehabilitation Research Center, Malcom Randall VA Medical Center, North Florida/South Georgia Veterans Health System, 1601 SW Archer Rd, Gainesville 32608-1197, USA
| | - Kevin K W Wang
- Department of Emergency Medicine, University of Florida, 1329 SW 16th Street, Suite 5270, Gainesville, FL 32610, USA
| | - Floyd Thompson
- Brain Rehabilitation Research Center, Malcom Randall VA Medical Center, North Florida/South Georgia Veterans Health System, 1601 SW Archer Rd, Gainesville 32608-1197, USA; Department of Physiological Sciences, University of Florida, 1333 Center Dr, Gainesville, FL 32603, USA; Department of Neuroscience, University of Florida, 1149 Newell Dr, Gainesville, FL 32610, USA
| | - Prodip Bose
- Brain Rehabilitation Research Center, Malcom Randall VA Medical Center, North Florida/South Georgia Veterans Health System, 1601 SW Archer Rd, Gainesville 32608-1197, USA; Department of Anesthesiology, College of Medicine, University of Florida, 1600 SW Archer Rd m509, Gainesville, FL 32610-0254, USA; Department of Neurology, University of Florida, 1149 Newell Dr, Gainesville, FL 32611, USA.
| |
Collapse
|
36
|
Zarepour L, Gharaylou Z, Hadjighassem M, Shafaghi L, Majedi H, Behzad E, Hosseindoost S, Ramezani F, Nasirinezhad F. Preliminary study of analgesic effect of bumetanide on neuropathic pain in patients with spinal cord injury. J Clin Neurosci 2020; 81:477-484. [PMID: 33222966 DOI: 10.1016/j.jocn.2020.10.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 09/07/2020] [Accepted: 10/03/2020] [Indexed: 02/01/2023]
Abstract
BACKGROUND/OBJECTIVE The current study evaluated the analgesic effects of bumetanide as an adjunctive treatment in managing neuropathic pain following spinal cord injury. The peripheral expression level of Na-K-Cl-cotransporter-1 (NKCC1) and K-Cl-cotransporter-2 (KCC2) genes in polymorphonuclear lymphocytes (PMLs) assessed as a possible biomarker indicating central underlying mechanisms. METHODS This open-label, single-arm, pilot trial of bumetanide (2 mg/day) is an add-on treatment conducted in 14 SCI patients for 19 weeks. The whole duration consisted of three phases: pre-treatment (1 month), titration (3 weeks), and active treatment (4 months). Ultimately, nine patients completed the study. The primary outcome variables were the endpoint pain score measured by the numeric rating scale (NRS), and the short-form McGill Pain Questionnaire. Secondary endpoints included the Short-Form Health Survey that measures the quality of life. Blood samples were collected and used for determining the expression of NKCC1 and KCC2 genes in transcription and translation levels. RESULTS Bumetanide treatment significantly reduced average pain intensity according to the NRS and the short form of the McGill Pain Questionnaire scores. The baseline expression of KCC2 protein was low between groups and increased significantly following treatment (P < 0.05). Through the current study, pain improvement accompanied by the more significant mean change from the baseline for the overall quality of life. CONCLUSION These data might be a piece of preliminary evidence for the analgesic effect of bumetanide on neuropathic pain and could support the potential role of the upregulation of KCC2 protein and involvement of GABAergic disinhibition in producing neuropathic pain.
Collapse
Affiliation(s)
- Leila Zarepour
- Department of Physiology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zeinab Gharaylou
- Multiple Sclerosis Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahmoudreza Hadjighassem
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran; Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran.
| | - Lida Shafaghi
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hossein Majedi
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Ebrahim Behzad
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Saereh Hosseindoost
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Ramezani
- Department of Physiology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Farinaz Nasirinezhad
- Department of Physiology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
37
|
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
|
38
|
Tan AQ, Barth S, Trumbower RD. Acute intermittent hypoxia as a potential adjuvant to improve walking following spinal cord injury: evidence, challenges, and future directions. CURRENT PHYSICAL MEDICINE AND REHABILITATION REPORTS 2020; 8:188-198. [PMID: 33738145 DOI: 10.1007/s40141-020-00270-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Purpose of Review The reacquisition and preservation of walking ability are highly valued goals in spinal cord injury (SCI) rehabilitation. Recurrent episodes of breathing low oxygen (i.e., acute intermittent hypoxia, AIH) is a potential therapy to promote walking recovery after incomplete SCI via endogenous mechanisms of neuroplasticity. Here, we report on the progress of AIH, alone or paired with other treatments, on walking recovery in persons with incomplete SCI. We evaluate the evidence of AIH as a therapy ready for clinical and home use and the real and perceived challenges that may interfere with this possibility. Recent Findings Repetitive AIH is a safe and an efficacious treatment to enhance strength, walking speed and endurance, as well as, dynamic balance in persons with chronic, incomplete SCI. Summary The potential for AIH as a treatment for SCI remains high, but further research is necessary to understand treatment targets and effectiveness in a large cohort of persons with SCI.
Collapse
Affiliation(s)
- Andrew Quesada Tan
- Department of Physical Medicine and Rehabilitation, Harvard Medical School, Boston, MA. USA
- Spaulding Rehabilitation Hospital, Boston MA
| | - Stella Barth
- Spaulding Rehabilitation Hospital, Boston MA
- Harvard University, Cambridge MA
| | - Randy D Trumbower
- Department of Physical Medicine and Rehabilitation, Harvard Medical School, Boston, MA. USA
- Spaulding Rehabilitation Hospital, Boston MA
| |
Collapse
|
39
|
Bonnet M, Alluin O, Trimaille T, Gigmes D, Marqueste T, Decherchi P. Delayed Injection of a Physically Cross-Linked PNIPAAm- g-PEG Hydrogel in Rat Contused Spinal Cord Improves Functional Recovery. ACS OMEGA 2020; 5:10247-10259. [PMID: 32426581 PMCID: PMC7226861 DOI: 10.1021/acsomega.9b03611] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Accepted: 04/14/2020] [Indexed: 06/11/2023]
Abstract
Spinal cord injury is a main health issue, leading to multiple functional deficits with major consequences such as motor and sensitive impairment below the lesion. To date, all repair strategies remain ineffective. In line with the experiments showing that implanted hydrogels, immunologically inert biomaterials, from natural or synthetic origins, are promising tools and in order to reduce functional deficits, to increase locomotor recovery, and to reduce spasticity, we injected into the lesion area, 1 week after a severe T10 spinal cord contusion, a thermoresponsive physically cross-linked poly(N-isopropylacrylamide)-poly(ethylene glycol) copolymer hydrogel. The effect of postinjury intensive rehabilitation training was also studied. A group of male Sprague-Dawley rats receiving the hydrogel was enrolled in an 8 week program of physical activity (15 min/day, 5 days/week) in order to verify if the combination of a treadmill step-training and hydrogel could lead to better outcomes. The data obtained were compared to those obtained in animals with a spinal lesion alone receiving a saline injection with or without performing the same program of physical activity. Furthermore, in order to verify the biocompatibility of our designed biomaterial, an inflammatory reaction (interleukin-1β, interleukin-6, and tumor necrosis factor-α) was examined 15 days post-hydrogel injection. Functional recovery (postural and locomotor activities and sensorimotor coordination) was assessed from the day of injection, once a week, for 9 weeks. Finally, 9 weeks postinjection, the spinal reflexivity (rate-dependent depression of the H-reflex) was measured. The results indicate that the hydrogel did not induce an additional inflammation. Furthermore, we observed the same significant locomotor improvements in hydrogel-injected animals as in trained saline-injected animals. However, the combination of hydrogel with exercise did not show higher recovery compared to that evaluated by the two strategies independently. Finally, the H-reflex depression recovery was found to be induced by the hydrogel and, albeit to a lesser degree, exercise. However, no recovery was observed when the two strategies were combined. Our results highlight the effectiveness of our copolymer and its high therapeutic potential to preserve/repair the spinal cord after lesion.
Collapse
Affiliation(s)
- Maxime Bonnet
- Aix
Marseille Univ, CNRS, ISM, UMR 7287, Institut des Sciences du Mouvement:
Etienne-Jules MAREY, Equipe, Plasticité des Systèmes
Nerveux et Musculaire, (PSNM), Parc Scientifique et Technologique
de Luminy, Faculté des Sciences du Sport de Marseille, CC910—163 Avenue de Luminy, F-13288 Marseille Cedex 09, France
| | - Olivier Alluin
- Aix
Marseille Univ, CNRS, ISM, UMR 7287, Institut des Sciences du Mouvement:
Etienne-Jules MAREY, Equipe, Plasticité des Systèmes
Nerveux et Musculaire, (PSNM), Parc Scientifique et Technologique
de Luminy, Faculté des Sciences du Sport de Marseille, CC910—163 Avenue de Luminy, F-13288 Marseille Cedex 09, France
| | - Thomas Trimaille
- Aix
Marseille Univ, CNRS, ICR, UMR 7273, Institut de Chimie Radicalaire,
Equipe, Chimie Radicalaire Organique et Polymères de Spécialité,
(CROPS), Case 562—Avenue
Escadrille Normandie-Niemen, F-13397 Marseille Cedex 20, France
| | - Didier Gigmes
- Aix
Marseille Univ, CNRS, ICR, UMR 7273, Institut de Chimie Radicalaire,
Equipe, Chimie Radicalaire Organique et Polymères de Spécialité,
(CROPS), Case 562—Avenue
Escadrille Normandie-Niemen, F-13397 Marseille Cedex 20, France
| | - Tanguy Marqueste
- Aix
Marseille Univ, CNRS, ISM, UMR 7287, Institut des Sciences du Mouvement:
Etienne-Jules MAREY, Equipe, Plasticité des Systèmes
Nerveux et Musculaire, (PSNM), Parc Scientifique et Technologique
de Luminy, Faculté des Sciences du Sport de Marseille, CC910—163 Avenue de Luminy, F-13288 Marseille Cedex 09, France
| | - Patrick Decherchi
- Aix
Marseille Univ, CNRS, ISM, UMR 7287, Institut des Sciences du Mouvement:
Etienne-Jules MAREY, Equipe, Plasticité des Systèmes
Nerveux et Musculaire, (PSNM), Parc Scientifique et Technologique
de Luminy, Faculté des Sciences du Sport de Marseille, CC910—163 Avenue de Luminy, F-13288 Marseille Cedex 09, France
| |
Collapse
|
40
|
Duy PQ, He M, He Z, Kahle KT. Preclinical insights into therapeutic targeting of KCC2 for disorders of neuronal hyperexcitability. Expert Opin Ther Targets 2020; 24:629-637. [PMID: 32336175 DOI: 10.1080/14728222.2020.1762174] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
INTRODUCTION Epilepsy is a common neurological disorder of neuronal hyperexcitability that begets recurrent and unprovoked seizures. The lack of a truly satisfactory pharmacotherapy for epilepsy highlights the clinical urgency for the discovery of new drug targets. To that end, targeting the electroneutral K+/Cl- cotransporter KCC2 has emerged as a novel therapeutic strategy for the treatment of epilepsy. AREAS COVERED We summarize the roles of KCC2 in the maintenance of synaptic inhibition and the evidence linking KCC2 dysfunction to epileptogenesis. We also discuss preclinical proof-of-principle studies that demonstrate that augmentation of KCC2 function can reduce seizure activity. Moreover, potential strategies to modulate KCC2 activity for therapeutic benefit are highlighted. EXPERT OPINION Although KCC2 is a promising drug target, questions remain before clinical translation. It is unclear whether increasing KCC2 activity can reverse epileptogenesis, the ultimate curative goal for epilepsy therapy that extends beyond seizure reduction. Furthermore, the potential adverse effects associated with increased KCC2 function have not been studied. Continued investigations into the neurobiology of KCC2 will help to translate promising preclinical insights into viable therapeutic avenues that leverage fundamental properties of KCC2 to treat medically intractable epilepsy and other disorders of failed synaptic inhibition with attendant neuronal hyperexcitability.
Collapse
Affiliation(s)
- Phan Q Duy
- Department of Neurosurgery, Yale University School of Medicine , New Haven, CT, USA.,Medical Scientist Training Program, Yale University School of Medicine , New Haven, CT, USA
| | - Miao He
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, and Department of Neurology, Harvard Medical School , Boston, MA, USA
| | - Zhigang He
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, and Department of Neurology, Harvard Medical School , Boston, MA, USA
| | - Kristopher T Kahle
- Department of Neurosurgery, Yale University School of Medicine , New Haven, CT, USA.,Department of Genetics, Yale University School of Medicine , New Haven, CT, USA.,Departments of Pediatrics and Cellular & Molecular Physiology, Yale University School of Medicine , New Haven, CT, USA.,Yale-Rockefeller NIH Centers for Mendelian Genomics, Yale University , New Haven, CT, USA.,Yale Stem Cell Center, Yale School of Medicine , New Haven, CT, USA
| |
Collapse
|
41
|
Alteration of glycinergic receptor expression in lumbar spinal motoneurons is involved in the mechanisms underlying spasticity after spinal cord injury. J Chem Neuroanat 2020; 106:101787. [PMID: 32339654 DOI: 10.1016/j.jchemneu.2020.101787] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 03/19/2020] [Accepted: 03/19/2020] [Indexed: 02/06/2023]
Abstract
Spasticity is a disabling motor disorder affecting 70% of people with brain and spinal cord injury. The rate-dependent depression (RDD) of the H reflex is the only electrophysiological measurement correlated with the degree of spasticity assessed clinically in spastic patients. Several lines of evidence suggest that the mechanism underlying the H reflex RDD depends on the strength of synaptic inhibition through GABAA (GABAAR) and glycine receptors (GlyR). In adult rats with spinal cord transection (SCT), we studied the time course of the expression of GABAAR and GlyR at the membrane of retrogradely identified Gastrocnemius and Tibialis anterior motoneurons (MNs) 3, 8 and 16 weeks after injury, and measured the RDD of the H reflex at similar post lesion times. Three weeks after SCT, a significant decrease in the expression of GABAA and GlyR was observed compared to intact rats, and the H-reflex RDD was much less pronounced than in controls. Eight weeks after SCT, GlyR values returned to normal. Simultaneously, we observed a tendency to recover normal RDD of the H reflex at higher frequencies. We tested whether an anti-inflammatory treatment using methylprednisolone performed immediately after SCT could prevent alterations in GABAA/glycine receptors and/or the development of spasticity observed 3 weeks after injury. This treatment restored control levels of GlyR but not the expression of GABAAR, and it completely prevented the attenuation of RDD. These data strongly suggest that alteration of glycinergic inhibition of lumbar MNs is involved in the mechanisms underlying spasticity after SCI.
Collapse
|
42
|
Sun J, Li R, Li X, Chen L, Liang Y, Zhang Q, Sun R, Hu H, Shao X, Fang J. Electroacupuncture therapy for change of pain in classical trigeminal neuralgia. Medicine (Baltimore) 2020; 99:e19710. [PMID: 32311955 PMCID: PMC7440061 DOI: 10.1097/md.0000000000019710] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
INTRODUCTION Classical trigeminal neuralgia (CTN) is a kind of trigeminal neuralgia which is due to neurovascular compression. The common neurological treatment CTN drug called carbamazepine is the main measure, although it usually has side effects and a high-rate of relapse. As a critical alternative therapy, electroacupuncture (EA) has been shown to benefit for neuropathic pain. The aims of this study are to observe the therapeutic effect and safety of EA for CTN, to evaluate whether EA has the advantage over carbamazepine in the analgesia of CTN. Furthermore, we would to establish a standardized, effective, and convenient therapy program of EA. METHODS AND ANALYSIS One hundred twenty patients diagnosed with CTN will be randomized for a 4-week intervention. The interventions will be different according to the four groups (EA + carbamazepine group, sham EA + carbamazepine group, EA + placebo group and sham EA + placebo group). EA therapy will be performed in specific acupoints with a dilute wave (2/100 Hz) for 60 minutes. Carbamazepine tablets will be taken orally with 0.1 g each time, thrice daily. Sham EA and placebo intervention will not receive EA and drug treatment. The main outcomes are the change from baseline intensity of pain at 6 months (pain evaluation by visual analogue score) and the change from baseline brief introduction of 2-week pain to evaluate pain comprehensively. The data management and statistical analysis will be conducted by third party statisticians. Incidence of adverse events will be investigated. ETHICS AND DISSEMINATION Ethics approval was obtained from the Clinical Trial Ethics Committee of The Third Affiliated Hospital of Zhejiang Chinese Medical University (NO. ZSLL-KY-2017-033) and Jiaxing Hospital of Traditional Chinese Medicine (NO. 2018-JZLK-002). The results will be disseminated by presentation at peer-reviewed journals.
Collapse
Affiliation(s)
- Jing Sun
- The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province
- The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Rongrong Li
- The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province
| | - Xiaoyu Li
- The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province
| | - Lifang Chen
- The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Yi Liang
- The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province
| | - Qifei Zhang
- The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province
| | - Ruohan Sun
- The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province
| | - Hantong Hu
- The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Xiaomei Shao
- The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province
- The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Jianqiao Fang
- The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province
| |
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
|
Yang A, Wang H, Zuo X, Yang J. Potassium Chloride Cotransporter 2 Inhibits Neuropathic Pain and Future Development of Neurodegeneration. J Alzheimers Dis 2020; 74:875-881. [PMID: 32144993 DOI: 10.3233/jad-200027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Persistent neuropathic pain (NP) causes future development of neurodegenerative diseases, e.g., Alzheimer' disease, and thus needs to be optimally treated. Surgically-induced neuropathic pain (SNPP) is a persistent pain that occurs in nearly half of the individuals after common operations. Here, we showed that specific activation of 5-hydroxytryptamine (5-HT) type 2A receptors by systemic administration of TCB-2 [(4-bromo-3,6-dimethoxybenzocyclobuten-1-yl) methylamine hydrobromide] improved the function of potassium chloride cotransporter 2 (KCC2), resulting in reduction in neuropathic pain after chronic constriction injury (CCI), a rat model that mimics SNPP. Moreover, TCB-2 administration attenuated both mechanical and thermal hyperalgesia, likely through augmentation of dorsal horn KCC2 levels, since this effect was abolished by intrathecal provision of dihydroindenyl oxy alkanoic acid (DIOA), which blocked the effects of KCC2. Furthermore, TCB-2-mediated re-activation of KCC2 likely reduces future development of neurodegeneration in rats. Together, our data support further studies on the possibility of using this strategy to reduce postoperative pain and future neurodegenerative disorders in clinic.
Collapse
Affiliation(s)
- Aimin Yang
- Department of Anesthesiology, Pain and Perioperative Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hongwei Wang
- Department of Anesthesiology, Pain and Perioperative Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaoxiao Zuo
- Department of Radiation Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jianjun Yang
- Department of Anesthesiology, Pain and Perioperative Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
45
|
Lorenzo LE, Godin AG, Ferrini F, Bachand K, Plasencia-Fernandez I, Labrecque S, Girard AA, Boudreau D, Kianicka I, Gagnon M, Doyon N, Ribeiro-da-Silva A, De Koninck Y. Enhancing neuronal chloride extrusion rescues α2/α3 GABA A-mediated analgesia in neuropathic pain. Nat Commun 2020; 11:869. [PMID: 32054836 PMCID: PMC7018745 DOI: 10.1038/s41467-019-14154-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 12/16/2019] [Indexed: 02/06/2023] Open
Abstract
Spinal disinhibition has been hypothesized to underlie pain hypersensitivity in neuropathic pain. Apparently contradictory mechanisms have been reported, raising questions on the best target to produce analgesia. Here, we show that nerve injury is associated with a reduction in the number of inhibitory synapses in the spinal dorsal horn. Paradoxically, this is accompanied by a BDNF-TrkB-mediated upregulation of synaptic GABAARs and by an α1-to-α2GABAAR subunit switch, providing a mechanistic rationale for the analgesic action of the α2,3GABAAR benzodiazepine-site ligand L838,417 after nerve injury. Yet, we demonstrate that impaired Cl- extrusion underlies the failure of L838,417 to induce analgesia at high doses due to a resulting collapse in Cl- gradient, dramatically limiting the benzodiazepine therapeutic window. In turn, enhancing KCC2 activity not only potentiated L838,417-induced analgesia, it rescued its analgesic potential at high doses, revealing a novel strategy for analgesia in pathological pain, by combined targeting of the appropriate GABAAR-subtypes and restoring Cl- homeostasis.
Collapse
Affiliation(s)
- Louis-Etienne Lorenzo
- CERVO Brain Research Centre, Quebec Mental Health Institute, Québec, QC, Canada
- Department of Pharmacology & Therapeutics, McGill University, Montreal, QC, Canada
| | - Antoine G Godin
- CERVO Brain Research Centre, Quebec Mental Health Institute, Québec, QC, Canada
- Department of Psychiatry & Neuroscience, Université Laval, Québec, QC, Canada
- Graduate program in Neuroscience, Université Laval, Québec, QC, Canada
| | - Francesco Ferrini
- CERVO Brain Research Centre, Quebec Mental Health Institute, Québec, QC, Canada
- Department of Psychiatry & Neuroscience, Université Laval, Québec, QC, Canada
- Graduate program in Neuroscience, Université Laval, Québec, QC, Canada
- Department of Veterinary Sciences, University of Turin, Turin, Italy
| | - Karine Bachand
- CERVO Brain Research Centre, Quebec Mental Health Institute, Québec, QC, Canada
| | - Isabel Plasencia-Fernandez
- CERVO Brain Research Centre, Quebec Mental Health Institute, Québec, QC, Canada
- Graduate program in Neuroscience, Université Laval, Québec, QC, Canada
| | - Simon Labrecque
- CERVO Brain Research Centre, Quebec Mental Health Institute, Québec, QC, Canada
| | - Alexandre A Girard
- CERVO Brain Research Centre, Quebec Mental Health Institute, Québec, QC, Canada
- Ecole Polytechnique, IP Paris, Palaiseau, France
| | - Dominic Boudreau
- CERVO Brain Research Centre, Quebec Mental Health Institute, Québec, QC, Canada
- Graduate program in Neuroscience, Université Laval, Québec, QC, Canada
| | - Irenej Kianicka
- Chlorion Pharma, Laval, Québec, QC, Canada
- Laurent Pharmaceuticals Inc., Montreal, QC, Canada
| | - Martin Gagnon
- CERVO Brain Research Centre, Quebec Mental Health Institute, Québec, QC, Canada
- Centre for Innovation, University of Otago, Dunedin, New Zealand
| | - Nicolas Doyon
- CERVO Brain Research Centre, Quebec Mental Health Institute, Québec, QC, Canada
- Finite Element Interdisciplinary Research Group (GIREF), Université Laval, Québec, QC, Canada
| | - Alfredo Ribeiro-da-Silva
- Department of Pharmacology & Therapeutics, McGill University, Montreal, QC, Canada
- Department of Anatomy & Cell Biology, McGill University, Montreal, QC, Canada
- Alan Edwards Centre for Research on Pain, McGill University, Montreal, QC, Canada
| | - Yves De Koninck
- CERVO Brain Research Centre, Quebec Mental Health Institute, Québec, QC, Canada.
- Department of Pharmacology & Therapeutics, McGill University, Montreal, QC, Canada.
- Department of Psychiatry & Neuroscience, Université Laval, Québec, QC, Canada.
- Graduate program in Neuroscience, Université Laval, Québec, QC, Canada.
- Alan Edwards Centre for Research on Pain, McGill University, Montreal, QC, Canada.
| |
Collapse
|
46
|
Martin E, Cazenave W, Allain AE, Cattaert D, Branchereau P. Implication of 5-HT in the Dysregulation of Chloride Homeostasis in Prenatal Spinal Motoneurons from the G93A Mouse Model of Amyotrophic Lateral Sclerosis. Int J Mol Sci 2020; 21:E1107. [PMID: 32046135 PMCID: PMC7039234 DOI: 10.3390/ijms21031107] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Revised: 02/04/2020] [Accepted: 02/05/2020] [Indexed: 12/14/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease characterized by progressive motor neuron degeneration and muscle paralysis. The early presymptomatic onset of abnormal processes is indicative of cumulative defects that ultimately lead to a late manifestation of clinical symptoms. It remains of paramount importance to identify the primary defects that underlie this condition and to determine how these deficits lead to a cycle of deterioration. We recently demonstrated that prenatal E17.5 lumbar spinal motoneurons (MNs) from SOD1G93A mice exhibit a KCC2-related alteration in chloride homeostasis, i.e., the EGABAAR is more depolarized than in WT littermates. Here, using immunohistochemistry, we found that the SOD1G93A lumbar spinal cord is less enriched with 5-HT descending fibres than the WT lumbar spinal cord. High-performance liquid chromatography confirmed the lower level of the monoamine 5-HT in the SOD1G93A spinal cord compared to the WT spinal cord. Using ex vivo perforated patch-clamp recordings of lumbar MNs coupled with pharmacology, we demonstrated that 5-HT strongly hyperpolarizes the EGABAAR by interacting with KCC2. Therefore, the deregulation of the interplay between 5-HT and KCC2 may explain the alteration in chloride homeostasis detected in prenatal SOD1G93A MNs. In conclusion, 5-HT and KCC2 are two likely key factors in the presymptomatic phase of ALS, particular in familial ALS involving the SOD1G93A mutation.
Collapse
Affiliation(s)
| | | | | | | | - Pascal Branchereau
- University of Bordeaux, CNRS, INCIA, UMR 5287, F-33000 Bordeaux, France; (E.M.); (W.C.); (A.-E.A.); (D.C.)
| |
Collapse
|
47
|
Auer T, Schreppel P, Erker T, Schwarzer C. Impaired chloride homeostasis in epilepsy: Molecular basis, impact on treatment, and current treatment approaches. Pharmacol Ther 2020; 205:107422. [DOI: 10.1016/j.pharmthera.2019.107422] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 10/07/2019] [Indexed: 12/14/2022]
|
48
|
The Role of Descending Pain Modulation in Chronic Primary Pain: Potential Application of Drugs Targeting Serotonergic System. Neural Plast 2019; 2019:1389296. [PMID: 31933624 PMCID: PMC6942873 DOI: 10.1155/2019/1389296] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 11/02/2019] [Accepted: 11/27/2019] [Indexed: 11/24/2022] Open
Abstract
Chronic primary pain (CPP) is a group of diseases with long-term pain and functional disorders but without structural or specific tissue pathologies. CPP is becoming a serious health problem in clinical practice due to the unknown cause of intractable pain and high cost of health care yet has not been satisfactorily addressed. During the past decades, a significant role for the descending pain modulation and alterations due to specific diseases of CPP has been emphasized. It has been widely established that central sensitization and alterations in neuroplasticity induced by the enhancement of descending pain facilitation and/or the impairment of descending pain inhibition can explain many chronic pain states including CPP. The descending serotonergic neurons in the raphe nuclei target receptors along the descending pain circuits and exert either pro- or antinociceptive effects in different pain conditions. In this review, we summarize the possible underlying descending pain regulation mechanisms in CPP and the role of serotonin, thus providing evidence for potential application of analgesic medications based on the serotonergic system in CPP patients.
Collapse
|
49
|
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
|
50
|
5-HT 2A receptor activation normalizes stress-induced dysregulation of GABAergic signaling in the ventral tegmental area. Proc Natl Acad Sci U S A 2019; 116:27028-27034. [PMID: 31806759 DOI: 10.1073/pnas.1911446116] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Stress is known to alter GABAergic signaling in the ventral tegmental area (VTA), and this inhibitory plasticity is associated with increased alcohol self-administration. In humans, serotonin 2A receptor (5-HT2AR) agonists can treat stress- and alcohol-related disorders, but the neural substrates are ill-defined. Thus, we reasoned that 5-HT2AR pharmacotherapies may ameliorate the stress-induced dysregulated inhibitory VTA circuitry that contributes to subsequent alcohol abuse. We found that acute stress exposure in mice compromised GABA-mediated inhibition of VTA GABA neurons corresponding with increased ethanol-induced GABAergic transmission. This stress-induced inhibitory plasticity was reversible by applying the 5-HT2AR agonist TCB-2 ex vivo via functional enhancement of the potassium-chloride cotransporter KCC2. The signaling pathway linking 5-HT2AR activation and normalization of KCC2 function was dependent on protein kinase C signaling and phosphorylation of KCC2 at serine 940 (S940), as mutation of S940 to alanine prevented restoration of chloride transport function by TCB-2. Through positive modulation of KCC2, TCB-2 also reduced elevated ethanol-induced GABAergic signaling after stress exposure that has previously been linked to increased ethanol consumption. Collectively, these findings provide mechanistic insights into the therapeutic action of 5-HT2AR agonists at the neuronal and circuit levels of brain reward circuitry.
Collapse
|