1
|
Lu MN, Wang D, Ye CJ, Yan GJ, Song JF, Shi XY, Li SS, Liu LN, Zhang HX, Dong XH, Hu T, Wang XY, Xiyang YB. Navβ2 Intracellular Fragments Contribute to Aβ1-42-Induced Cognitive Impairment and Synaptic Deficit Through Transcriptional Suppression of BDNF. Mol Neurobiol 2024:10.1007/s12035-024-04317-y. [PMID: 38965172 DOI: 10.1007/s12035-024-04317-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Accepted: 06/17/2024] [Indexed: 07/06/2024]
Abstract
A pathological hallmark of Alzheimer's disease (AD) is the region-specific accumulation of the amyloid-beta protein (Aβ), which triggers aberrant neuronal excitability, synaptic impairment, and progressive cognitive decline. Previous works have demonstrated that Aβ pathology induced aberrant elevation in the levels and excessive enzymatic hydrolysis of voltage-gated sodium channel type 2 beta subunit (Navβ2) in the brain of AD models, accompanied by alteration in excitability of hippocampal neurons, synaptic deficits, and subsequently, cognitive dysfunction. However, the mechanism is unclear. In this research, by employing cell models treated with toxic Aβ1-42 and AD mice, the possible effects and potential mechanisms induced by Navβ2. The results reveal that Aβ1-42 induces remarkable increases in Navβ2 intracellular domain (Navβ2-ICD) and decreases in both BDNF exons and protein levels, as well as phosphorylated tropomyosin-related kinase B (pTrkB) expression in cells and mice, coupled with cognitive impairments, synaptic deficits, and aberrant neuronal excitability. Administration with exogenous Navβ2-ICD further enhances these effects induced by Aβ1-42, while interfering the generation of Navβ2-ICD and/or complementing BDNF neutralize the Navβ2-ICD-conducted effects. Luciferase reporter assay verifies that Navβ2-ICD regulates BDNF transcription and expression by targeting its promoter. Collectively, our findings partially elucidate that abnormal enzymatic hydrolysis of Navβ2 induced by Aβ1-42-associated AD pathology leads to intracellular Navβ2-ICD overload, which may responsible to abnormal neuronal excitability, synaptic deficit, and cognition dysfunction, through its transcriptional suppression on BDNF. Therefore, this work supplies novel evidences that Navβ2 plays crucial roles in the occurrence and progression of cognitive impairment of AD by transcriptional regulatory activity of its cleaved ICD.
Collapse
Affiliation(s)
- Min-Nan Lu
- Institute of Neuroscience, Kunming Medical University, Kunming, 650500, Yunnan, China
- Science and Technology Achievement Incubation Center, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Dan Wang
- Institute of Neuroscience, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Chen-Jun Ye
- Institute of Neuroscience, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Guo-Ji Yan
- Institute of Neuroscience, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Jing-Feng Song
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Xin-Ying Shi
- Institute of Neuroscience, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Shan-Shan Li
- Experimental Teaching Center, Basic Medical College, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Li-Na Liu
- Institute of Neuroscience, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Hui-Xiang Zhang
- Institute of Neuroscience, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Xiao-Han Dong
- Institute of Neuroscience, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Tao Hu
- Department of Laboratory Medicine, The Third People's Hospital of Yunnan Province, Kunming, 650000, Yunnan, China
| | - Xu-Yang Wang
- Department of Neurosurgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Yan-Bin Xiyang
- Institute of Neuroscience, Kunming Medical University, Kunming, 650500, Yunnan, China.
| |
Collapse
|
2
|
Reduced Expression of Voltage-Gated Sodium Channel Beta 2 Restores Neuronal Injury and Improves Cognitive Dysfunction Induced by A β1-42. Neural Plast 2022; 2022:3995227. [PMID: 36406589 PMCID: PMC9671742 DOI: 10.1155/2022/3995227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 10/05/2022] [Accepted: 10/18/2022] [Indexed: 11/12/2022] Open
Abstract
Voltage-gated sodium channel beta 2 (Nav2.2 or Navβ2, coded by SCN2B mRNA), a gene involved in maintaining normal physiological functions of the prefrontal cortex and hippocampus, might be associated with prefrontal cortex aging and memory decline. This study investigated the effects of Navβ2 in amyloid-β 1-42- (Aβ1-42-) induced neural injury model and the potential underlying molecular mechanism. The results showed that Navβ2 knockdown restored neuronal viability of Aβ1-42-induced injury in neurons; increased the contents of brain-derived neurotrophic factor (BDNF), enzyme neprilysin (NEP) protein, and NEP enzyme activity; and effectively altered the proportions of the amyloid precursor protein (APP) metabolites including Aβ42, sAPPα, and sAPPβ, thus ameliorating cognitive dysfunction. This may be achieved through regulating NEP transcription and APP metabolism, accelerating Aβ degradation, alleviating neuronal impairment, and regulating BDNF-related signal pathways to repair neuronal synaptic efficiency. This study provides novel evidence indicating that Navβ2 plays crucial roles in the repair of neuronal injury induced by Aβ1-42 both in vivo and in vitro.
Collapse
|
3
|
Sen S, Lagas S, Roy A, Kumar H. Cytoskeleton saga: Its regulation in normal physiology and modulation in neurodegenerative disorders. Eur J Pharmacol 2022; 925:175001. [PMID: 35525310 DOI: 10.1016/j.ejphar.2022.175001] [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: 02/02/2022] [Revised: 03/31/2022] [Accepted: 04/29/2022] [Indexed: 11/25/2022]
Abstract
Cells are fundamental units of life. To ensure the maintenance of homeostasis, integrity of structural and functional counterparts is needed to be essentially balanced. The cytoskeleton plays a vital role in regulating the cellular morphology, signalling and other factors involved in pathological conditions. Microtubules, actin (microfilaments), intermediate filaments (IF) and their interactions are required for these activities. Various proteins associated with these components are primary requirements for directing their functions. Disruption of this organization due to faulty genetics, oxidative stress or impaired transport mechanisms are the major causes of dysregulated signalling cascades leading to various pathological conditions like Alzheimer's (AD), Parkinson's (PD), Huntington's disease (HD) or amyotrophic lateral sclerosis (ALS), hereditary spastic paraplegia (HSP) or any traumatic injury like spinal cord injury (SCI). Novel or conventional therapeutic approaches may be specific or non-specific, targeting either three basic components of the cytoskeleton or various cascades that serve as a cue to numerous pathways like ROCK signalling or the GSK-3β pathway. An enormous number of drugs have been redirected for modulating the cytoskeletal dynamics and thereby may pave the way for inhibiting the progression of these diseases and their complications.
Collapse
Affiliation(s)
- Santimoy Sen
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat, India
| | - Sheetal Lagas
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat, India
| | - Abhishek Roy
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat, India
| | - Hemant Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat, India.
| |
Collapse
|
4
|
Boscia F, Elkjaer ML, Illes Z, Kukley M. Altered Expression of Ion Channels in White Matter Lesions of Progressive Multiple Sclerosis: What Do We Know About Their Function? Front Cell Neurosci 2021; 15:685703. [PMID: 34276310 PMCID: PMC8282214 DOI: 10.3389/fncel.2021.685703] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Accepted: 05/23/2021] [Indexed: 12/19/2022] Open
Abstract
Despite significant advances in our understanding of the pathophysiology of multiple sclerosis (MS), knowledge about contribution of individual ion channels to axonal impairment and remyelination failure in progressive MS remains incomplete. Ion channel families play a fundamental role in maintaining white matter (WM) integrity and in regulating WM activities in axons, interstitial neurons, glia, and vascular cells. Recently, transcriptomic studies have considerably increased insight into the gene expression changes that occur in diverse WM lesions and the gene expression fingerprint of specific WM cells associated with secondary progressive MS. Here, we review the ion channel genes encoding K+, Ca2+, Na+, and Cl- channels; ryanodine receptors; TRP channels; and others that are significantly and uniquely dysregulated in active, chronic active, inactive, remyelinating WM lesions, and normal-appearing WM of secondary progressive MS brain, based on recently published bulk and single-nuclei RNA-sequencing datasets. We discuss the current state of knowledge about the corresponding ion channels and their implication in the MS brain or in experimental models of MS. This comprehensive review suggests that the intense upregulation of voltage-gated Na+ channel genes in WM lesions with ongoing tissue damage may reflect the imbalance of Na+ homeostasis that is observed in progressive MS brain, while the upregulation of a large number of voltage-gated K+ channel genes may be linked to a protective response to limit neuronal excitability. In addition, the altered chloride homeostasis, revealed by the significant downregulation of voltage-gated Cl- channels in MS lesions, may contribute to an altered inhibitory neurotransmission and increased excitability.
Collapse
Affiliation(s)
- Francesca Boscia
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, School of Medicine, University of Naples "Federico II", Naples, Italy
| | - Maria Louise Elkjaer
- Neurology Research Unit, Department of Clinical Research, University of Southern Denmark, Odense, Denmark.,Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Zsolt Illes
- Neurology Research Unit, Department of Clinical Research, University of Southern Denmark, Odense, Denmark.,Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark.,Department of Neurology, Odense University Hospital, Odense, Denmark
| | - Maria Kukley
- Achucarro Basque Center for Neuroscience, Leioa, Spain.,Ikerbasque Basque Foundation for Science, Bilbao, Spain
| |
Collapse
|
5
|
Tan YX, Hong Y, Jiang S, Lu MN, Li S, Chen B, Zhang L, Hu T, Mao R, Mei R, Xiyang YB. MicroRNA‑449a regulates the progression of brain aging by targeting SCN2B in SAMP8 mice. Int J Mol Med 2020; 45:1091-1102. [PMID: 32124967 PMCID: PMC7053848 DOI: 10.3892/ijmm.2020.4502] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 01/29/2020] [Indexed: 12/23/2022] Open
Abstract
Our previous study demonstrated that the expression of sodium channel voltage-gated beta 2 (SCN2B) increased with aging in senescence-accelerated mouse prone 8 (SAMP8) mice, and was identified to be associated with a decline in learning and memory, while the underlying mechanism is unclear. In the present study, multiple differentially expressed miRNAs, which may be involved in the process of aging by regulating target genes, were identified in the prefrontal cortex and hippocampus of SAMP8 mice though miRNA microarray analysis. Using bioinformatics prediction, SCN2B was identified to be one of the potential target genes of miR-449a, which was downregulated in the hippocampus. Previous studies demonstrated that miR-449a is involved in the occurrence and progression of aging by regulating a variety of target genes. Therefore, it was hypothesized that miR-449a may be involved in the process of brain aging by targeting SCN2B. To verify this hypothesis, the following experiments were conducted: A reverse transcription-quantitative polymerase chain reaction assay revealed that the expression level of miR-449a was significantly decreased in the prefrontal cortex and hippocampus of 12-month old SAMP8 mice; a dual-luciferase reporter assay verified that miR-449a regulated SCN2B expression by binding to the 3′-UTR 'seed region'; an anti-Ago co-immunoprecipitation combined with Affymetrix micro-array analyses demonstrated that the target mRNA highly enriched with Ago-miRNPs was confirmed to be SCN2B. Finally, overexpression of miR-449a or inhibition of SCN2B promoted the extension of hippocampal neurons in vitro. The results of the present study suggested that miR-449a was downregulated in the prefrontal cortex and hippocampus of SAMP8 mice and may regulate the process of brain aging by targeting SCN2B.
Collapse
Affiliation(s)
- Ya-Xin Tan
- Institute of Neuroscience, Basic Medical College, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Ying Hong
- Department of Laboratory Medicine, The Third People's Hospital of Yunnan Province, Kunming, Yunnan 650011, P.R. China
| | - Shui Jiang
- Department of Laboratory Medicine, The Third People's Hospital of Yunnan Province, Kunming, Yunnan 650011, P.R. China
| | - Min-Nan Lu
- Science and Technology Achievement Incubation Center, Kunming, Yunnan 650500, P.R. China
| | - Shan Li
- Institute of Neuroscience, Basic Medical College, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Bo Chen
- Science and Technology Achievement Incubation Center, Kunming, Yunnan 650500, P.R. China
| | - Li Zhang
- Editorial Department of Journal of Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Tao Hu
- Department of Laboratory Medicine, The Third People's Hospital of Yunnan Province, Kunming, Yunnan 650011, P.R. China
| | - Rui Mao
- School of Stomatology, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Rong Mei
- Department of Neurology, The First People's Hospital of Yunnan Province, Kunming, Yunnan 650032, P.R. China
| | - Yan-Bin Xiyang
- Institute of Neuroscience, Basic Medical College, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| |
Collapse
|
6
|
Alrashdi B, Dawod B, Schampel A, Tacke S, Kuerten S, Marshall JS, Côté PD. Nav1.6 promotes inflammation and neuronal degeneration in a mouse model of multiple sclerosis. J Neuroinflammation 2019; 16:215. [PMID: 31722722 PMCID: PMC6852902 DOI: 10.1186/s12974-019-1622-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 10/22/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND In multiple sclerosis (MS) and in the experimental autoimmune encephalomyelitis (EAE) model of MS, the Nav1.6 voltage-gated sodium (Nav) channel isoform has been implicated as a primary contributor to axonal degeneration. Following demyelination Nav1.6, which is normally co-localized with the Na+/Ca2+ exchanger (NCX) at the nodes of Ranvier, associates with β-APP, a marker of neural injury. The persistent influx of sodium through Nav1.6 is believed to reverse the function of NCX, resulting in an increased influx of damaging Ca2+ ions. However, direct evidence for the role of Nav1.6 in axonal degeneration is lacking. METHODS In mice floxed for Scn8a, the gene that encodes the α subunit of Nav1.6, subjected to EAE we examined the effect of eliminating Nav1.6 from retinal ganglion cells (RGC) in one eye using an AAV vector harboring Cre and GFP, while using the contralateral either injected with AAV vector harboring GFP alone or non-targeted eye as control. RESULTS In retinas, the expression of Rbpms, a marker for retinal ganglion cells, was found to be inversely correlated to the expression of Scn8a. Furthermore, the gene expression of the pro-inflammatory cytokines Il6 (IL-6) and Ifng (IFN-γ), and of the reactive gliosis marker Gfap (GFAP) were found to be reduced in targeted retinas. Optic nerves from targeted eyes were shown to have reduced macrophage infiltration and improved axonal health. CONCLUSION Taken together, our results are consistent with Nav1.6 promoting inflammation and contributing to axonal degeneration following demyelination.
Collapse
Affiliation(s)
- Barakat Alrashdi
- Department of Biology, Dalhousie University, Halifax, NS, B3H 4R2, Canada.,Department of Biology, Al-Jouf University, Sakaka, Saudi Arabia
| | - Bassel Dawod
- Department of Pathology, Dalhousie University, Halifax, NS, B3H 4R2, Canada
| | - Andrea Schampel
- Institute of Anatomy and Cell Biology Friedrich Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Sabine Tacke
- Institute of Anatomy and Cell Biology Friedrich Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Stefanie Kuerten
- Institute of Anatomy and Cell Biology Friedrich Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Jean S Marshall
- Department of Pathology, Dalhousie University, Halifax, NS, B3H 4R2, Canada.,Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, B3H 4R2, Canada
| | - Patrice D Côté
- Department of Biology, Dalhousie University, Halifax, NS, B3H 4R2, Canada. .,Department of Ophthalmology and Visual Sciences, Dalhousie University, Halifax, NS, B3H 4R2, Canada.
| |
Collapse
|
7
|
Hu T, Li SS, Lu MN, Zhang L, Chen B, Mao R, Mei R, Tan YX, Li S, Xiyang YB. Neuroprotection induced by Navβ2‑knockdown in APP/PS1 transgenic neurons is associated with NEP regulation. Mol Med Rep 2019; 20:2002-2011. [PMID: 31257483 DOI: 10.3892/mmr.2019.10406] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 05/29/2019] [Indexed: 11/06/2022] Open
Abstract
Voltage‑gated sodium channel β2 (Navβ2), as an unconventional substrate of β‑site amyloid precursor protein cleaving enzyme 1, is involved in regulating the neuronal surface expression of sodium channels. A previous study demonstrated that knockdown of Navβ2 protected neurons and induced spatial cognition improvement by partially reducing pathological amyloidogenic processing of amyloid precursor protein (APP) in aged APP/presenilin 1 (PS1) transgenic mice. The present study aimed to investigate whether Navβ2 knockdown altered APP metabolism via regulation of the Aβ‑degrading enzyme neprilysin (NEP). APPswe/PS1ΔE9 mice (APP/PS1 transgenic mice with a C57BL/6J genetic background) carrying a Navβ2‑knockdown mutation (APP/PS1/Navβ2‑kd) or without Navβ2 knockdown (APP/PS1) were used for cell culture and further analysis. The present results demonstrated that in APP/PS1 mouse‑derived neurons, Navβ2 knockdown partially reversed the reduction in pathological APP cleavage, and the recovery of neurite extension and neuron area. Additionally, Navβ2 knockdown increased NEP activity and levels, and the levels of intracellular domain fragment binding to the NEP promoter. The present findings suggested that knockdown of Navβ2 reversed the APP/PS1 mutation‑induced deficiency in amyloid β degradation by regulating NEP.
Collapse
Affiliation(s)
- Tao Hu
- Institute of Neuroscience, Basic Medical College, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Shan-Shan Li
- Basic Medical College, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Min-Nan Lu
- Experiment Center for Medical Science Research, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Li Zhang
- Editorial Department of Journal of Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Bo Chen
- Experiment Center for Medical Science Research, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Rui Mao
- School of Stomatology, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Rong Mei
- Department of Neurology, The First People's Hospital of Yunnan Province, Kunming, Yunnan 650032, P.R. China
| | - Ya-Xin Tan
- Institute of Neuroscience, Basic Medical College, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Shan Li
- Institute of Neuroscience, Basic Medical College, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Yan-Bin Xiyang
- Institute of Neuroscience, Basic Medical College, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| |
Collapse
|
8
|
Lan M, Tang X, Zhang J, Yao Z. Insights in pathogenesis of multiple sclerosis: nitric oxide may induce mitochondrial dysfunction of oligodendrocytes. Rev Neurosci 2018; 29:39-53. [PMID: 28822986 DOI: 10.1515/revneuro-2017-0033] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 06/15/2017] [Indexed: 01/01/2023]
Abstract
Demyelinating diseases, such as multiple sclerosis (MS), are kinds of common diseases in the central nervous system (CNS), and originated from myelin loss and axonal damage. Oligodendrocyte dysfunction is the direct reason of demyelinating lesions in the CNS. Nitric oxide (NO) plays an important role in the pathological process of demyelinating diseases. Although the neurotoxicity of NO is more likely mediated by peroxynitrite rather than NO itself, NO can impair oligodendrocyte energy metabolism through mediating the damaging of mitochondrial DNA, mitochondrial membrane and mitochondrial respiratory chain complexes. In the progression of MS, NO can mainly mediate demyelination, axonal degeneration and cell death. Hence, in this review, we extensively discuss endangerments of NO in oligodendrocytes (OLs), which is suggested to be the main mediator in demyelinating diseases, e.g. MS. We hypothesize that NO takes part in MS through impairing the function of monocarboxylate transporter 1, especially causing axonal degeneration. Then, it further provides a new insight that NO for OLs may be a reliable therapeutic target to ameliorate the course of demyelinating diseases.
Collapse
Affiliation(s)
- Minghong Lan
- Department of Physiology, Third Military Medical University, Chongqing 400038, China
| | - Xiaoyi Tang
- Department of Physiology, Third Military Medical University, Chongqing 400038, China
| | - Jie Zhang
- Department of Physiology, Third Military Medical University, Chongqing 400038, China
| | - Zhongxiang Yao
- Department of Physiology, Third Military Medical University, Chongqing 400038, China
| |
Collapse
|
9
|
Guarnieri FC, Bellani S, Yekhlef L, Bergamaschi A, Finardi A, Fesce R, Pozzi D, Monzani E, Fornasiero EF, Matteoli M, Martino G, Furlan R, Taverna S, Muzio L, Valtorta F. Synapsin I deletion reduces neuronal damage and ameliorates clinical progression of experimental autoimmune encephalomyelitis. Brain Behav Immun 2018; 68:197-210. [PMID: 29066310 DOI: 10.1016/j.bbi.2017.10.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 10/10/2017] [Accepted: 10/20/2017] [Indexed: 11/20/2022] Open
Abstract
The classical view of multiple sclerosis (MS) pathogenesis states that inflammation-mediated demyelination is responsible for neuronal damage and loss. However, recent findings show that impairment of neuronal functions and demyelination can be independent events, suggesting the coexistence of other pathogenic mechanisms. Due to the inflammatory milieu, subtle alterations in synaptic function occur, which are probably at the basis of the early cognitive decline that often precedes the neurodegenerative phases in MS patients. In particular, it has been reported that inflammation enhances excitatory synaptic transmission while it decreases GABAergic transmission in vitro and ex vivo. This evidence points to the idea that an excitation/inhibition imbalance occurs in the inflamed MS brain, even though the exact molecular mechanisms leading to this synaptic dysfunction are as yet not completely clear. Along this line, we observed that acute treatment of primary hippocampal neurons in culture with pro-inflammatory cytokines leads to an increased phosphorylation of synapsin I (SynI) by ERK1/2 kinase and to an increase in the frequency of spontaneous synaptic vesicle release events, which is prevented by SynI deletion. In vivo, the ablation of SynI expression is protective in terms of disease progression and neuronal damage in the experimental autoimmune encephalomyelitis mouse model of MS. Our results point to a possible key role in MS pathogenesis of the neuronal protein SynI, a regulator of excitation/inhibition balance in neuronal networks.
Collapse
Affiliation(s)
- Fabrizia C Guarnieri
- Division of Neuroscience, San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy; Vita-Salute San Raffaele University, Via Olgettina 58, 20132 Milan, Italy
| | - Serena Bellani
- Division of Neuroscience, San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy
| | - Latefa Yekhlef
- Division of Neuroscience, San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy
| | - Andrea Bergamaschi
- Division of Neuroscience, San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy
| | - Annamaria Finardi
- Division of Neuroscience, San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy
| | - Riccardo Fesce
- Centre of Neuroscience and DISTA, University of Insubria, Via Ravasi 2, 21100 Varese, Italy
| | - Davide Pozzi
- Humanitas Clinical and Research Centre, Via Manzoni 113, 20089 Rozzano, Milan, Italy
| | - Elena Monzani
- Division of Neuroscience, San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy
| | - Eugenio F Fornasiero
- Division of Neuroscience, San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy
| | - Michela Matteoli
- Humanitas Clinical and Research Centre, Via Manzoni 113, 20089 Rozzano, Milan, Italy; CNR Institute of Neuroscience, via Vanvitelli 32, 20129 Milan, Italy
| | - Gianvito Martino
- Division of Neuroscience, San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy; Vita-Salute San Raffaele University, Via Olgettina 58, 20132 Milan, Italy
| | - Roberto Furlan
- Division of Neuroscience, San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy
| | - Stefano Taverna
- Division of Neuroscience, San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy
| | - Luca Muzio
- Division of Neuroscience, San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy
| | - Flavia Valtorta
- Division of Neuroscience, San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy; Vita-Salute San Raffaele University, Via Olgettina 58, 20132 Milan, Italy.
| |
Collapse
|
10
|
Wang J, Ou SW, Wang YJ. Distribution and function of voltage-gated sodium channels in the nervous system. Channels (Austin) 2017; 11:534-554. [PMID: 28922053 DOI: 10.1080/19336950.2017.1380758] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Voltage-gated sodium channels (VGSCs) are the basic ion channels for neuronal excitability, which are crucial for the resting potential and the generation and propagation of action potentials in neurons. To date, at least nine distinct sodium channel isoforms have been detected in the nervous system. Recent studies have identified that voltage-gated sodium channels not only play an essential role in the normal electrophysiological activities of neurons but also have a close relationship with neurological diseases. In this study, the latest research findings regarding the structure, type, distribution, and function of VGSCs in the nervous system and their relationship to neurological diseases, such as epilepsy, neuropathic pain, brain tumors, neural trauma, and multiple sclerosis, are reviewed in detail.
Collapse
Affiliation(s)
- Jun Wang
- a Department of Neurosurgery , The First Hospital of China Medical University , Shenyang , P.R. China
| | - Shao-Wu Ou
- a Department of Neurosurgery , The First Hospital of China Medical University , Shenyang , P.R. China
| | - Yun-Jie Wang
- a Department of Neurosurgery , The First Hospital of China Medical University , Shenyang , P.R. China
| |
Collapse
|
11
|
Hu T, Xiao Z, Mao R, Chen B, Lu MN, Tong J, Mei R, Li SS, Xiao ZC, Zhang LF, Xiyang YB. Navβ2 knockdown improves cognition in APP/PS1 mice by partially inhibiting seizures and APP amyloid processing. Oncotarget 2017; 8:99284-99295. [PMID: 29245901 PMCID: PMC5725092 DOI: 10.18632/oncotarget.21849] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2017] [Accepted: 10/02/2017] [Indexed: 11/25/2022] Open
Abstract
Voltage-gated sodium channels beta 2 (Navβ2, encoded by SCN2B) is a substrate of β-site amyloid precursor protein cleaving enzyme 1 (BACE1) and regulates cell surface expression of channels in neurons. Previous studies reported enhanced Navβ2 processing by BACE1 in Alzheimer’s disease (AD) model and patients. We investigated whether changes in Navβ2 expression affect neuronal seizure and amyloid precursor protein (APP) processing in an AD mouse model. Our study used eight-month-old APP/presenilin 1 (PS1) mice and transgenic Navβ2 knockdown [by 61% vs. wild type (WT)] APP/PS1 mice (APP/PS1/Navβ2-kd), with age-matched WT and Navβ2 knockdown (Navβ2-kd) mice as controls. We found that Navβ2 knockdown in APP/PS1 mice partially reversed the abnormal Navβ2 cleavage and the changes in intracellular and total Nav1.1α expression. It also restored sodium currents density in hippocampal neurons and neuronal activity, as indicated by EEG tracing; improved Morris water maze performance; and shifted APP amyloidogenic metabolism towards non-amyloidogenic processing. There were no differences in these indicators between WT and Navβ2-kd mice. These results suggest Navβ2 knockdown may be a promising strategy for treating AD.
Collapse
Affiliation(s)
- Tao Hu
- Institute of Neuroscience, Basic Medical College, Kunming Medical University, Kunming, Yunnan, PR China.,Department of Laboratory Medicine, The Third People's Hospital of Yunnan Province, Kunming, Yunnan, PR China
| | - Zhangang Xiao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, PR China.,Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, PR China
| | - Rui Mao
- School of Stomatology, Kunming Medical University, Kunming, Yunnan, PR China
| | - Bo Chen
- Experiment Center for Medical Science Research, Kunming Medical University, Kunming, Yunnan, PR China
| | - Min-Nan Lu
- Experiment Center for Medical Science Research, Kunming Medical University, Kunming, Yunnan, PR China
| | - Jun Tong
- Physical Education Department, Kunming Medical University, Kunming, Yunnan, PR China
| | - Rong Mei
- Department of Neurology, The First People's Hospital of Yunnan Province, Kunming, Yunnan, PR China
| | - Shan-Shan Li
- Basic Medical College, Kunming Medical University, Kunming, Yunnan, PR China
| | - Zhi-Cheng Xiao
- Institute of Molecular and Clinical Medicine, Kunming Medical University, Kunming, Yunnan, PR China.,Monash Immunology and Stem Cell Laboratories (MISCL), Monash University, Clayton, VIC, Australia
| | - Lian-Feng Zhang
- Key Laboratory of Human Diseases Comparative Medicine, Ministry of Health, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences(CAMS) & Comparative Medicine Centre, Peking Union Medical College (PUMC), Beijing, China
| | - Yan-Bin Xiyang
- Institute of Neuroscience, Basic Medical College, Kunming Medical University, Kunming, Yunnan, PR China
| |
Collapse
|
12
|
Hull JM, Isom LL. Voltage-gated sodium channel β subunits: The power outside the pore in brain development and disease. Neuropharmacology 2017; 132:43-57. [PMID: 28927993 DOI: 10.1016/j.neuropharm.2017.09.018] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 07/19/2017] [Accepted: 09/11/2017] [Indexed: 12/19/2022]
Abstract
Voltage gated sodium channels (VGSCs) were first identified in terms of their role in the upstroke of the action potential. The underlying proteins were later identified as saxitoxin and scorpion toxin receptors consisting of α and β subunits. We now know that VGSCs are heterotrimeric complexes consisting of a single pore forming α subunit joined by two β subunits; a noncovalently linked β1 or β3 and a covalently linked β2 or β4 subunit. VGSC α subunits contain all the machinery necessary for channel cell surface expression, ion conduction, voltage sensing, gating, and inactivation, in one central, polytopic, transmembrane protein. VGSC β subunits are more than simple accessories to α subunits. In the more than two decades since the original cloning of β1, our knowledge of their roles in physiology and pathophysiology has expanded immensely. VGSC β subunits are multifunctional. They confer unique gating mechanisms, regulate cellular excitability, affect brain development, confer distinct channel pharmacology, and have functions that are independent of the α subunits. The vast array of functions of these proteins stems from their special station in the channelome: being the only known constituents that are cell adhesion and intra/extracellular signaling molecules in addition to being part of channel complexes. This functional trifecta and how it goes awry demonstrates the power outside the pore in ion channel signaling complexes, broadening the term channelopathy beyond defects in ion conduction. This article is part of the Special Issue entitled 'Channelopathies.'
Collapse
Affiliation(s)
- Jacob M Hull
- Neuroscience Program and Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI 48109, United States
| | - Lori L Isom
- Neuroscience Program and Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI 48109, United States.
| |
Collapse
|
13
|
Sodium Channel β2 Subunits Prevent Action Potential Propagation Failures at Axonal Branch Points. J Neurosci 2017; 37:9519-9533. [PMID: 28871036 DOI: 10.1523/jneurosci.0891-17.2017] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2017] [Revised: 08/21/2017] [Accepted: 08/22/2017] [Indexed: 12/19/2022] Open
Abstract
Neurotransmitter release depends on voltage-gated Na+ channels (Navs) to propagate an action potential (AP) successfully from the axon hillock to a synaptic terminal. Unmyelinated sections of axon are very diverse structures encompassing branch points and numerous presynaptic terminals with undefined molecular partners of Na+ channels. Using optical recordings of Ca2+ and membrane voltage, we demonstrate here that Na+ channel β2 subunits (Navβ2s) are required to prevent AP propagation failures across the axonal arborization of cultured rat hippocampal neurons (mixed male and female). When Navβ2 expression was reduced, we identified two specific phenotypes: (1) membrane excitability and AP-evoked Ca2+ entry were impaired at synapses and (2) AP propagation was severely compromised with >40% of axonal branches no longer responding to AP-stimulation. We went on to show that a great deal of electrical signaling heterogeneity exists in AP waveforms across the axonal arborization independent of axon morphology. Therefore, Navβ2 is a critical regulator of axonal excitability and synaptic function in unmyelinated axons.SIGNIFICANCE STATEMENT Voltage-gated Ca2+ channels are fulcrums of neurotransmission that convert electrical inputs into chemical outputs in the form of vesicle fusion at synaptic terminals. However, the role of the electrical signal, the presynaptic action potential (AP), in modulating synaptic transmission is less clear. What is the fidelity of a propagating AP waveform in the axon and what molecules shape it throughout the axonal arborization? Our work identifies several new features of AP propagation in unmyelinated axons: (1) branches of a single axonal arborization have variable AP waveforms independent of morphology, (2) Na+ channel β2 subunits modulate AP-evoked Ca2+-influx, and (3) β2 subunits maintain successful AP propagation across the axonal arbor. These findings are relevant to understanding the flow of excitation in the brain.
Collapse
|
14
|
Sharma P, Wright DK, Johnston LA, Powell KL, Wlodek ME, Shultz SR, O'Brien TJ, Gilby KL. Differences in white matter structure between seizure prone (FAST) and seizure resistant (SLOW) rat strains. Neurobiol Dis 2017; 104:33-40. [DOI: 10.1016/j.nbd.2017.04.022] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Revised: 03/20/2017] [Accepted: 04/27/2017] [Indexed: 02/09/2023] Open
|
15
|
Zostawa J, Adamczyk J, Sowa P, Adamczyk-Sowa M. The influence of sodium on pathophysiology of multiple sclerosis. Neurol Sci 2017; 38:389-398. [PMID: 28078565 PMCID: PMC5331099 DOI: 10.1007/s10072-016-2802-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2016] [Accepted: 12/19/2016] [Indexed: 01/06/2023]
Abstract
Multiple sclerosis (MS) is a chronic, inflammatory, autoimmune disease of the central nervous system, and is an important cause of disability in young adults. In genetically susceptible individuals, several environmental factors may play a partial role in the pathogenesis of MS. Some studies suggests that high-salt diet (>5 g/day) may contribute to the MS and other autoimmune disease development through the induction of pathogenic Th17 cells and pro-inflammatory cytokines in both humans and mice. However, the precise mechanisms of pro-inflammatory effect of sodium chloride intake are not yet explained. The purpose of this review was to discuss the present state of knowledge on the potential role of environmental and dietary factors, particularly sodium chloride on the development and course of MS.
Collapse
Affiliation(s)
- Jacek Zostawa
- Department of Neurology in Zabrze, Medical University of Silesia, ul. 3-go Maja 13-15, 41-800, Zabrze, Poland
| | - Jowita Adamczyk
- Department of Neurology in Zabrze, Medical University of Silesia, ul. 3-go Maja 13-15, 41-800, Zabrze, Poland.
| | - Paweł Sowa
- Department of Otorhinolaryngology and Oncological Laryngology, Medical University of Silesia, ul. C. Skłodowskiej 10, 41-800, Zabrze, Poland
| | - Monika Adamczyk-Sowa
- Department of Neurology in Zabrze, Medical University of Silesia, ul. 3-go Maja 13-15, 41-800, Zabrze, Poland
| |
Collapse
|
16
|
Abstract
Voltage-gated sodium channels are protein complexes comprised of one pore forming α subunit and two, non-pore forming, β subunits. The voltage-gated sodium channel β subunits were originally identified to function as auxiliary subunits, which modulate the gating, kinetics, and localization of the ion channel pore. Since that time, the five β subunits have been shown to play crucial roles as multifunctional signaling molecules involved in cell adhesion, cell migration, neuronal pathfinding, fasciculation, and neurite outgrowth. Here, we provide an overview of the evidence implicating the β subunits in their conducting and non-conducting roles. Mutations in the β subunit genes (SCN1B-SCN4B) have been linked to a variety of diseases. These include cancer, epilepsy, cardiac arrhythmias, sudden infant death syndrome/sudden unexpected death in epilepsy, neuropathic pain, and multiple neurodegenerative disorders. β subunits thus provide novel therapeutic targets for future drug discovery.
Collapse
Affiliation(s)
- Alexandra A Bouza
- Department of Pharmacology, University of Michigan Medical School, 2200 MSRBIII, 1150 W. Medical Center Dr., Ann Arbor, MI, 48109-5632, USA
| | - Lori L Isom
- Department of Pharmacology, University of Michigan Medical School, 2301 MSRB III, 1150 W. Medical Center Dr., Ann Arbor, MI, 48109-5632, USA.
| |
Collapse
|
17
|
Shimada Y, Sato T, Yajima T, Fujita M, Hashimoto N, Shoji N, Sasano T, Ichikawa H. SCN2B in the Rat Trigeminal Ganglion and Trigeminal Sensory Nuclei. Cell Mol Neurobiol 2016; 36:1399-1408. [PMID: 26852328 DOI: 10.1007/s10571-016-0340-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 01/28/2016] [Indexed: 12/21/2022]
Abstract
The beta-2 subunit of the mammalian brain voltage-gated sodium channel (SCN2B) was examined in the rat trigeminal ganglion (TG) and trigeminal sensory nuclei. In the TG, 42.6 % of sensory neurons were immunoreactive (IR) for SCN2B. These neurons had various cell body sizes. In facial skins and oral mucosae, corpuscular nerve endings contained SCN2B-immunoreactivity. SCN2B-IR nerve fibers formed nerve plexuses beneath taste buds in the tongue and incisive papilla. However, SCN2B-IR free nerve endings were rare in cutaneous and mucosal epithelia. Tooth pulps, muscle spindles and major salivary glands were also innervated by SCN2B-IR nerve fibers. A double immunofluorescence method revealed that about 40 % of SCN2B-IR neurons exhibited calcitonin gene-related peptide (CGRP)-immunoreactivity. However, distributions of SCN2B- and CGRP-IR nerve fibers were mostly different in facial, oral and cranial structures. By retrograde tracing method, 60.4 and 85.3 % of TG neurons innervating the facial skin and tooth pulp, respectively, showed SCN2B-immunoreactivity. CGRP-immunoreactivity was co-localized by about 40 % of SCN2B-IR cutaneous and tooth pulp TG neurons. In trigeminal sensory nuclei of the brainstem, SCN2B-IR neuronal cell bodies were common in deep laminae of the subnucleus caudalis, and the subnuclei interpolaris and oralis. In the mesencephalic trigeminal tract nucleus, primary sensory neurons also exhibited SCN2B-immunoreactivity. In other regions of trigeminal sensory nuclei, SCN2B-IR cells were very infrequent. SCN2B-IR neuropil was detected in deep laminae of the subnucleus caudalis as well as in the subnuclei interpolaris, oralis and principalis. These findings suggest that SCN2B is expressed by various types of sensory neurons in the TG. There appears to be SCN2B-containing pathway in the TG and trigeminal sensory nuclei.
Collapse
Affiliation(s)
- Yusuke Shimada
- Division of Oral Diagnosis, Graduate School of Dentistry, Tohoku University, Sendai, 980-8575, Japan
| | - Tadasu Sato
- Division of Oral and Craniofacial Anatomy, Graduate School of Dentistry, Tohoku University, 4-1 Seiryo-machi, Sendai, 980-8575, Japan
| | - Takehiro Yajima
- Division of Operative Dentistry, Graduate School of Dentistry, Tohoku University, Sendai, 980-8575, Japan
| | - Masatoshi Fujita
- Division of Dental Anesthesiology and Pain Management, Graduate School of Dentistry, Tohoku University, Sendai, 980-8575, Japan
| | - Naoya Hashimoto
- Division of Oral Diagnosis, Graduate School of Dentistry, Tohoku University, Sendai, 980-8575, Japan
| | - Noriaki Shoji
- Division of Oral Diagnosis, Graduate School of Dentistry, Tohoku University, Sendai, 980-8575, Japan
| | - Takashi Sasano
- Division of Oral Diagnosis, Graduate School of Dentistry, Tohoku University, Sendai, 980-8575, Japan
| | - Hiroyuki Ichikawa
- Division of Oral and Craniofacial Anatomy, Graduate School of Dentistry, Tohoku University, 4-1 Seiryo-machi, Sendai, 980-8575, Japan.
| |
Collapse
|
18
|
Patel F, Brackenbury WJ. Dual roles of voltage-gated sodium channels in development and cancer. THE INTERNATIONAL JOURNAL OF DEVELOPMENTAL BIOLOGY 2016; 59:357-66. [PMID: 26009234 DOI: 10.1387/ijdb.150171wb] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Voltage-gated Na(+) channels (VGSCs) are heteromeric protein complexes containing pore-forming α subunits together with non-pore-forming β subunits. There are nine α subunits, Nav1.1-Nav1.9, and four β subunits, β1-β4. The β subunits are multifunctional, modulating channel activity, cell surface expression, and are members of the immunoglobulin superfamily of cell adhesion molecules. VGSCs are classically responsible for action potential initiation and conduction in electrically excitable cells, including neurons and muscle cells. In addition, through the β1 subunit, VGSCs regulate neurite outgrowth and pathfinding in the developing central nervous system. Reciprocal signalling through Nav1.6 and β1 collectively regulates Na(+) current, electrical excitability and neurite outgrowth in cerebellar granule neurons. Thus, α and β subunits may have diverse interacting roles dependent on cell/tissue type. VGSCs are also expressed in non-excitable cells, including cells derived from a number of types of cancer. In cancer cells, VGSC α and β subunits regulate cellular morphology, migration, invasion and metastasis. VGSC expression associates with poor prognosis in several studies. It is hypothesised that VGSCs are up-regulated in metastatic tumours, favouring an invasive phenotype. Thus, VGSCs may have utility as prognostic markers, and/or as novel therapeutic targets for reducing/preventing metastatic disease burden. VGSCs appear to regulate a number of key cellular processes, both during normal postnatal development of the CNS and during cancer metastasis, by a combination of conducting (i.e. via Na(+) current) and non-conducting mechanisms.
Collapse
|
19
|
Winters JJ, Isom LL. Developmental and Regulatory Functions of Na(+) Channel Non-pore-forming β Subunits. CURRENT TOPICS IN MEMBRANES 2016; 78:315-51. [PMID: 27586289 DOI: 10.1016/bs.ctm.2016.07.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Voltage-gated Na(+) channels (VGSCs) isolated from mammalian neurons are heterotrimeric complexes containing one pore-forming α subunit and two non-pore-forming β subunits. In excitable cells, VGSCs are responsible for the initiation of action potentials. VGSC β subunits are type I topology glycoproteins, containing an extracellular amino-terminal immunoglobulin (Ig) domain with homology to many neural cell adhesion molecules (CAMs), a single transmembrane segment, and an intracellular carboxyl-terminal domain. VGSC β subunits are encoded by a gene family that is distinct from the α subunits. While α subunits are expressed in prokaryotes, β subunit orthologs did not arise until after the emergence of vertebrates. β subunits regulate the cell surface expression, subcellular localization, and gating properties of their associated α subunits. In addition, like many other Ig-CAMs, β subunits are involved in cell migration, neurite outgrowth, and axon pathfinding and may function in these roles in the absence of associated α subunits. In sum, these multifunctional proteins are critical for both channel regulation and central nervous system development.
Collapse
Affiliation(s)
- J J Winters
- University of Michigan Neuroscience Program, Ann Arbor, MI, United States
| | - L L Isom
- University of Michigan Neuroscience Program, Ann Arbor, MI, United States; University of Michigan Medical School, Ann Arbor, MI, United States
| |
Collapse
|
20
|
Das S, Gilchrist J, Bosmans F, Van Petegem F. Binary architecture of the Nav1.2-β2 signaling complex. eLife 2016; 5. [PMID: 26894959 PMCID: PMC4769172 DOI: 10.7554/elife.10960] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 01/07/2016] [Indexed: 12/15/2022] Open
Abstract
To investigate the mechanisms by which β-subunits influence Nav channel function, we solved the crystal structure of the β2 extracellular domain at 1.35Å. We combined these data with known bacterial Nav channel structural insights and novel functional studies to determine the interactions of specific residues in β2 with Nav1.2. We identified a flexible loop formed by (72)Cys and (75)Cys, a unique feature among the four β-subunit isoforms. Moreover, we found that (55)Cys helps to determine the influence of β2 on Nav1.2 toxin susceptibility. Further mutagenesis combined with the use of spider toxins reveals that (55)Cys forms a disulfide bond with (910)Cys in the Nav1.2 domain II pore loop, thereby suggesting a 1:1 stoichiometry. Our results also provide clues as to which disulfide bonds are formed between adjacent Nav1.2 (912/918)Cys residues. The concepts emerging from this work will help to form a model reflecting the β-subunit location in a Nav channel complex.
Collapse
Affiliation(s)
- Samir Das
- Department of Biochemistry and Molecular Biology, Life Sciences Institute, University of British Columbia, Vancouver, Canada
| | - John Gilchrist
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Frank Bosmans
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, United States.,Solomon H Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Filip Van Petegem
- Department of Biochemistry and Molecular Biology, Life Sciences Institute, University of British Columbia, Vancouver, Canada
| |
Collapse
|
21
|
Petracca M, Fleysher L, Oesingmann N, Inglese M. Sodium MRI of multiple sclerosis. NMR IN BIOMEDICINE 2016; 29:153-61. [PMID: 25851455 PMCID: PMC5771413 DOI: 10.1002/nbm.3289] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2014] [Revised: 02/25/2015] [Accepted: 02/26/2015] [Indexed: 05/11/2023]
Abstract
Multiple sclerosis (MS) is the most common cause of non-traumatic disability in young adults. The mechanisms underlying neurodegeneration and disease progression are poorly understood, in part as a result of the lack of non-invasive methods to measure and monitor neurodegeneration in vivo. Sodium MRI is a topic of increasing interest in MS research as it allows the metabolic characterization of brain tissue in vivo, and integration with the structural information provided by (1)H MRI, helping in the exploration of pathogenetic mechanisms and possibly offering insights into disease progression and monitoring of treatment outcomes. We present an up-to-date review of the sodium MRI application in MS organized into four main sections: (i) biological and pathogenetic role of sodium; (ii) brief overview of sodium imaging techniques; (iii) results of sodium MRI application in clinical studies; and (iv) future perspectives.
Collapse
Affiliation(s)
- Maria Petracca
- Department of Neurology, Icahn School of Medicine, Mount Sinai, New York, USA
- Department of Neuroscience, Federico II University, Naples, Italy
| | - Lazar Fleysher
- Department of Radiology, Icahn School of Medicine, Mount Sinai, New York, USA
| | | | - Matilde Inglese
- Department of Neurology, Icahn School of Medicine, Mount Sinai, New York, USA
- Department of Radiology, Icahn School of Medicine, Mount Sinai, New York, USA
- Department of Neuroscience, Icahn School of Medicine, Mount Sinai, New York, USA
| |
Collapse
|
22
|
Abstract
Voltage-gated sodium channels (VGSCs) are responsible for the initiation and propagation of action potentials in excitable cells. VGSCs in mammalian brain are heterotrimeric complexes of α and β subunits. Although β subunits were originally termed auxiliary, we now know that they are multifunctional signaling molecules that play roles in both excitable and nonexcitable cell types and with or without the pore-forming α subunit present. β subunits function in VGSC and potassium channel modulation, cell adhesion, and gene regulation, with particularly important roles in brain development. Mutations in the genes encoding β subunits are linked to a number of diseases, including epilepsy, sudden death syndromes like SUDEP and SIDS, and cardiac arrhythmia. Although VGSC β subunit-specific drugs have not yet been developed, this protein family is an emerging therapeutic target.
Collapse
Affiliation(s)
- Heather A O'Malley
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan 48109;
| | | |
Collapse
|
23
|
XiYang YB, Wang YC, Zhao Y, Ru J, Lu BT, Zhang YN, Wang NC, Hu WY, Liu J, Yang JW, Wang ZJ, Hao CG, Feng ZT, Xiao ZC, Dong W, Quan XZ, Zhang LF, Wang TH. Sodium Channel Voltage-Gated Beta 2 Plays a Vital Role in Brain Aging Associated with Synaptic Plasticity and Expression of COX5A and FGF-2. Mol Neurobiol 2015; 53:955-967. [PMID: 25575679 DOI: 10.1007/s12035-014-9048-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2014] [Accepted: 12/02/2014] [Indexed: 02/05/2023]
Abstract
The role of sodium channel voltage-gated beta 2 (SCN2B) in brain aging is largely unknown. The present study was therefore designed to determine the role of SCN2B in brain aging by using the senescence-accelerated mice prone 8 (SAMP8), a brain senescence-accelerated animal model, together with the SCN2B transgenic mice. The results showed that SAMP8 exhibited impaired learning and memory functions, assessed by the Morris water maze test, as early as 8 months of age. The messenger RNA (mRNA) and protein expressions of SCN2B were also upregulated in the prefrontal cortex at this age. Treatment with traditional Chinese anti-aging medicine Xueshuangtong (Panax notoginseng saponins, PNS) significantly reversed the SCN2B expressions in the prefrontal cortex, resulting in improved learning and memory. Moreover, SCN2B knockdown transgenic mice were generated and bred to determine the roles of SCN2B in brain senescence. A reduction in the SCN2B level by 60.68% resulted in improvement in the hippocampus-dependent spatial recognition memory and long-term potential (LTP) slope of field excitatory postsynaptic potential (fEPSP), followed by an upregulation of COX5A mRNA levels and downregulation of fibroblast growth factor-2 (FGF-2) mRNA expression. Together, the present findings indicated that SCN2B could play an important role in the aging-related cognitive deterioration, which is associated with the regulations of COX5A and FGF-2. These findings could provide the potential strategy of candidate target to develop antisenescence drugs for the treatment of brain aging.
Collapse
Affiliation(s)
- Yan-Bin XiYang
- Institute of Neuroscience, Kunming Medical University, 1168 West Chunrong Road, Yuhua Avenue, Chenggong District, Kunming, 650500, Yunnan, China.,Institute of Neurological Disease, State Key Lab of Biotherapy, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, China
| | - You-Cui Wang
- Institute of Neurological Disease, State Key Lab of Biotherapy, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, China
| | - Ya Zhao
- Institute of Neuroscience, Kunming Medical University, 1168 West Chunrong Road, Yuhua Avenue, Chenggong District, Kunming, 650500, Yunnan, China
| | - Jin Ru
- Institute of Neuroscience, Kunming Medical University, 1168 West Chunrong Road, Yuhua Avenue, Chenggong District, Kunming, 650500, Yunnan, China
| | - Bing-Tuan Lu
- Institute of Neuroscience, Kunming Medical University, 1168 West Chunrong Road, Yuhua Avenue, Chenggong District, Kunming, 650500, Yunnan, China.,Institute of Neurological Disease, State Key Lab of Biotherapy, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yue-Ning Zhang
- Institute of Neuroscience, Kunming Medical University, 1168 West Chunrong Road, Yuhua Avenue, Chenggong District, Kunming, 650500, Yunnan, China
| | - Nai-Chao Wang
- Institute of Neuroscience, Kunming Medical University, 1168 West Chunrong Road, Yuhua Avenue, Chenggong District, Kunming, 650500, Yunnan, China
| | - Wei-Yan Hu
- Institute of Molecular and Clinical Medicine, Kunming Medical University, 1168 West Chunrong Road, Yuhua Avenue, Chenggong District, Kunming, 650500, Yunnan, China.,Monash Immunology and Stem Cell Laboratories (MISCL), Monash University, Clayton, VIC, Australia
| | - Jia Liu
- Institute of Neuroscience, Kunming Medical University, 1168 West Chunrong Road, Yuhua Avenue, Chenggong District, Kunming, 650500, Yunnan, China.,Institute of Neurological Disease, State Key Lab of Biotherapy, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, China
| | - Jin-Wei Yang
- Institute of Neuroscience, Kunming Medical University, 1168 West Chunrong Road, Yuhua Avenue, Chenggong District, Kunming, 650500, Yunnan, China
| | - Zhao-Jun Wang
- Institute of Neuroscience, Kunming Medical University, 1168 West Chunrong Road, Yuhua Avenue, Chenggong District, Kunming, 650500, Yunnan, China
| | - Chun-Guang Hao
- Institute of Neuroscience, Kunming Medical University, 1168 West Chunrong Road, Yuhua Avenue, Chenggong District, Kunming, 650500, Yunnan, China
| | - Zhong-Tang Feng
- Institute of Neuroscience, Kunming Medical University, 1168 West Chunrong Road, Yuhua Avenue, Chenggong District, Kunming, 650500, Yunnan, China.,Institute of Neurological Disease, State Key Lab of Biotherapy, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, China
| | - Zhi-Cheng Xiao
- Institute of Molecular and Clinical Medicine, Kunming Medical University, 1168 West Chunrong Road, Yuhua Avenue, Chenggong District, Kunming, 650500, Yunnan, China.,Monash Immunology and Stem Cell Laboratories (MISCL), Monash University, Clayton, VIC, Australia
| | - Wei Dong
- Key Laboratory of Human Diseases Comparative Medicine, Ministry of Health, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences (CAMS), 100021, Beijing, China.,Comparative Medicine Centre, Peking Union Medical College (PUMC), 100021, Beijing, China
| | - Xiong-Zhi Quan
- Key Laboratory of Human Diseases Comparative Medicine, Ministry of Health, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences (CAMS), 100021, Beijing, China.,Comparative Medicine Centre, Peking Union Medical College (PUMC), 100021, Beijing, China
| | - Lian-Feng Zhang
- Key Laboratory of Human Diseases Comparative Medicine, Ministry of Health, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences (CAMS), 100021, Beijing, China. .,Comparative Medicine Centre, Peking Union Medical College (PUMC), 100021, Beijing, China.
| | - Ting-Hua Wang
- Institute of Neuroscience, Kunming Medical University, 1168 West Chunrong Road, Yuhua Avenue, Chenggong District, Kunming, 650500, Yunnan, China. .,Institute of Neurological Disease, State Key Lab of Biotherapy, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
24
|
Schattling B, Eggert B, Friese MA. Acquired channelopathies as contributors to development and progression of multiple sclerosis. Exp Neurol 2014; 262 Pt A:28-36. [PMID: 24656770 DOI: 10.1016/j.expneurol.2013.12.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Accepted: 12/13/2013] [Indexed: 12/18/2022]
Abstract
Multiple sclerosis (MS), the most frequent inflammatory disease of the central nervous system (CNS), affects about two and a half million individuals worldwide and causes major burdens to the patients, which develop the disease usually at the age of 20 to 40. MS is likely referable to a breakdown of immune cell tolerance to CNS self-antigens resulting in focal immune cell infiltration, activation of microglia and astrocytes, demyelination and axonal and neuronal loss. Here we discuss how altered expression patterns and dysregulated functions of ion channels contribute on a molecular level to nearly all pathophysiological steps of the disease. In particular the detrimental redistribution of ion channels along axons, as well as neuronal excitotoxicity with regard to imbalanced glutamate homeostasis during chronic CNS inflammation will be discussed in detail. Together, we describe which ion channels in the immune and nervous system commend as attractive future drugable targets in MS treatment.
Collapse
Affiliation(s)
- Benjamin Schattling
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Falkenried 94, D-20251 Hamburg, Germany
| | - Britta Eggert
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Falkenried 94, D-20251 Hamburg, Germany
| | - Manuel A Friese
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Falkenried 94, D-20251 Hamburg, Germany.
| |
Collapse
|
25
|
Abstract
Multiple sclerosis (MS) is the most frequent chronic inflammatory disease of the CNS, and imposes major burdens on young lives. Great progress has been made in understanding and moderating the acute inflammatory components of MS, but the pathophysiological mechanisms of the concomitant neurodegeneration--which causes irreversible disability--are still not understood. Chronic inflammatory processes that continuously disturb neuroaxonal homeostasis drive neurodegeneration, so the clinical outcome probably depends on the balance of stressor load (inflammation) and any remaining capacity for neuronal self-protection. Hence, suitable drugs that promote the latter state are sorely needed. With the aim of identifying potential novel therapeutic targets in MS, we review research on the pathological mechanisms of neuroaxonal dysfunction and injury, such as altered ion channel activity, and the endogenous neuroprotective pathways that counteract oxidative stress and mitochondrial dysfunction. We focus on mechanisms inherent to neurons and their axons, which are separable from those acting on inflammatory responses and might, therefore, represent bona fide neuroprotective drug targets with the capability to halt MS progression.
Collapse
|
26
|
Calhoun JD, Isom LL. The role of non-pore-forming β subunits in physiology and pathophysiology of voltage-gated sodium channels. Handb Exp Pharmacol 2014; 221:51-89. [PMID: 24737232 DOI: 10.1007/978-3-642-41588-3_4] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Voltage-gated sodium channel β1 and β2 subunits were discovered as auxiliary proteins that co-purify with pore-forming α subunits in brain. The other family members, β1B, β3, and β4, were identified by homology and shown to modulate sodium current in heterologous systems. Work over the past 2 decades, however, has provided strong evidence that these proteins are not simply ancillary ion channel subunits, but are multifunctional signaling proteins in their own right, playing both conducting (channel modulatory) and nonconducting roles in cell signaling. Here, we discuss evidence that sodium channel β subunits not only regulate sodium channel function and localization but also modulate voltage-gated potassium channels. In their nonconducting roles, VGSC β subunits function as immunoglobulin superfamily cell adhesion molecules that modulate brain development by influencing cell proliferation and migration, axon outgrowth, axonal fasciculation, and neuronal pathfinding. Mutations in genes encoding β subunits are linked to paroxysmal diseases including epilepsy, cardiac arrhythmia, and sudden infant death syndrome. Finally, β subunits may be targets for the future development of novel therapeutics.
Collapse
Affiliation(s)
- Jeffrey D Calhoun
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, 48109-5632, USA
| | | |
Collapse
|
27
|
Maschietto M, Girardi S, Dal Maschio M, Scorzeto M, Vassanelli S. Sodium channel β2 subunit promotes filopodia-like processes and expansion of the dendritic tree in developing rat hippocampal neurons. Front Cell Neurosci 2013; 7:2. [PMID: 23355803 PMCID: PMC3555079 DOI: 10.3389/fncel.2013.00002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2012] [Accepted: 01/07/2013] [Indexed: 12/19/2022] Open
Abstract
The β2 auxiliary subunit of voltage-gated sodium channels (VGSCs) appears at early stages of brain development. It is abundantly expressed in the mammalian central nervous system where it forms complexes with different channel isoforms, including Nav1.2. From the structural point of view, β2 is a transmembrane protein: at its extracellular N-terminus an Ig-like type C2 domain mediates the binding to the pore-forming alpha subunit with disulfide bonds and the interactions with the extracellular matrix. Given this structural versatility, β2 has been suggested to play multiple functions ranging from channel targeting to the plasma membrane and gating modulation to control of cell adhesion. We report that, when expressed in Chinese Hamster Ovary cells CHO-K1, the subunit accumulates at the perimetral region of adhesion and particularly in large lamellipodia-like membrane processes where it induces formation of filopodia-like structures. When overexpressed in developing embryonic rat hippocampal neurons in vitro, β2 specifically promotes formation of filopodia-like processes in dendrites leading to expansion of the arborization tree, while axonal branching remains unaltered. In contrast to this striking and highly specific effect on dendritic morphology, the targeting of functional sodium channels to the plasma membrane, including the preferential localization of Nav1.2 at the axon, and their gating properties are only minimally affected. From these and previously reported observations it is suggested that β2, among its multiple functions, may contribute to promote dendritic outgrowth and to regulate neuronal wiring at specific stages of neuronal development.
Collapse
Affiliation(s)
- Marta Maschietto
- Department of Biomedical Sciences, University of Padova Padova, Italy
| | | | | | | | | |
Collapse
|
28
|
Eijkelkamp N, Linley JE, Baker MD, Minett MS, Cregg R, Werdehausen R, Rugiero F, Wood JN. Neurological perspectives on voltage-gated sodium channels. Brain 2012; 135:2585-612. [PMID: 22961543 PMCID: PMC3437034 DOI: 10.1093/brain/aws225] [Citation(s) in RCA: 255] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The activity of voltage-gated sodium channels has long been linked to disorders of neuronal excitability such as epilepsy and chronic pain. Recent genetic studies have now expanded the role of sodium channels in health and disease, to include autism, migraine, multiple sclerosis, cancer as well as muscle and immune system disorders. Transgenic mouse models have proved useful in understanding the physiological role of individual sodium channels, and there has been significant progress in the development of subtype selective inhibitors of sodium channels. This review will outline the functions and roles of specific sodium channels in electrical signalling and disease, focusing on neurological aspects. We also discuss recent advances in the development of selective sodium channel inhibitors.
Collapse
Affiliation(s)
- Niels Eijkelkamp
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, Gower Street, London WC1E 6BT, UK.
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Chen C, Calhoun JD, Zhang Y, Lopez-Santiago L, Zhou N, Davis TH, Salzer JL, Isom LL. Identification of the cysteine residue responsible for disulfide linkage of Na+ channel α and β2 subunits. J Biol Chem 2012; 287:39061-9. [PMID: 22992729 PMCID: PMC3493947 DOI: 10.1074/jbc.m112.397646] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2012] [Revised: 09/13/2012] [Indexed: 01/28/2023] Open
Abstract
Voltage-gated Na(+) channels in the brain are composed of a single pore-forming α subunit, one non-covalently linked β subunit (β1 or β3), and one disulfide-linked β subunit (β2 or β4). The final step in Na(+) channel biosynthesis in central neurons is concomitant α-β2 disulfide linkage and insertion into the plasma membrane. Consistent with this, Scn2b (encoding β2) null mice have reduced Na(+) channel cell surface expression in neurons, and action potential conduction is compromised. Here we generated a series of mutant β2 cDNA constructs to investigate the cysteine residue(s) responsible for α-β2 subunit covalent linkage. We demonstrate that a single cysteine-to-alanine substitution at extracellular residue Cys-26, located within the immunoglobulin (Ig) domain, abolishes the covalent linkage between α and β2 subunits. Loss of α-β2 covalent complex formation disrupts the targeting of β2 to nodes of Ranvier in a myelinating co-culture system and to the axon initial segment in primary hippocampal neurons, suggesting that linkage with α is required for normal β2 subcellular localization in vivo. WT β2 subunits are resistant to live cell Triton X-100 detergent extraction from the hippocampal axon initial segment, whereas mutant β2 subunits, which cannot form disulfide bonds with α, are removed by detergent. Taken together, our results demonstrate that α-β2 covalent association via a single, extracellular disulfide bond is required for β2 targeting to specialized neuronal subcellular domains and for β2 association with the neuronal cytoskeleton within those domains.
Collapse
Affiliation(s)
- Chunling Chen
- From the Department of Pharmacology and Cellular and Molecular Biology Program, University of Michigan Medical School, Ann Arbor, Michigan 48109 and
| | - Jeffrey D. Calhoun
- From the Department of Pharmacology and Cellular and Molecular Biology Program, University of Michigan Medical School, Ann Arbor, Michigan 48109 and
| | - Yanqing Zhang
- the Departments of Cell Biology and Neurology and the New York University Neuroscience Institute, New York University School of Medicine, New York, New York 10016
| | - Luis Lopez-Santiago
- From the Department of Pharmacology and Cellular and Molecular Biology Program, University of Michigan Medical School, Ann Arbor, Michigan 48109 and
| | - Ningna Zhou
- From the Department of Pharmacology and Cellular and Molecular Biology Program, University of Michigan Medical School, Ann Arbor, Michigan 48109 and
| | - Tigwa H. Davis
- From the Department of Pharmacology and Cellular and Molecular Biology Program, University of Michigan Medical School, Ann Arbor, Michigan 48109 and
| | - James L. Salzer
- the Departments of Cell Biology and Neurology and the New York University Neuroscience Institute, New York University School of Medicine, New York, New York 10016
| | - Lori L. Isom
- From the Department of Pharmacology and Cellular and Molecular Biology Program, University of Michigan Medical School, Ann Arbor, Michigan 48109 and
| |
Collapse
|
30
|
Savio-Galimberti E, Gollob MH, Darbar D. Voltage-gated sodium channels: biophysics, pharmacology, and related channelopathies. Front Pharmacol 2012; 3:124. [PMID: 22798951 PMCID: PMC3394224 DOI: 10.3389/fphar.2012.00124] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2011] [Accepted: 06/11/2012] [Indexed: 12/19/2022] Open
Abstract
Voltage-gated sodium channels (VGSC) are multi-molecular protein complexes expressed in both excitable and non-excitable cells. They are primarily formed by a pore-forming multi-spanning integral membrane glycoprotein (α-subunit) that can be associated with one or more regulatory β-subunits. The latter are single-span integral membrane proteins that modulate the sodium current (INa) and can also function as cell adhesion molecules. In vitro some of the cell-adhesive functions of the β-subunits may play important physiological roles independently of the α-subunits. Other endogenous regulatory proteins named “channel partners” or “channel interacting proteins” (ChiPs) like caveolin-3 and calmodulin/calmodulin kinase II (CaMKII) can also interact and modulate the expression and/or function of VGSC. In addition to their physiological roles in cell excitability and cell adhesion, VGSC are the site of action of toxins (like tetrodotoxin and saxitoxin), and pharmacologic agents (like antiarrhythmic drugs, local anesthetics, antiepileptic drugs, and newly developed analgesics). Mutations in genes that encode α- and/or β-subunits as well as the ChiPs can affect the structure and biophysical properties of VGSC, leading to the development of diseases termed sodium “channelopathies”. This review will outline the structure, function, and biophysical properties of VGSC as well as their pharmacology and associated channelopathies and highlight some of the recent advances in this field.
Collapse
Affiliation(s)
- Eleonora Savio-Galimberti
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Nashville, TN, USA
| | | | | |
Collapse
|
31
|
Strength-duration time constant in peripheral nerve: no abnormality in multiple sclerosis. Mult Scler Int 2012; 2012:390157. [PMID: 22645684 PMCID: PMC3356942 DOI: 10.1155/2012/390157] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2012] [Revised: 03/13/2012] [Accepted: 03/20/2012] [Indexed: 11/23/2022] Open
Abstract
Objectives. To investigate the properties of the strength-duration time constant (SDTC) in multiple sclerosis (MS) patients. Methods. The SDTC and rheobase in 16 MS patients and 19 healthy controls were obtained following stimulation of the right median nerve at the wrist. Results. SDTC and rheobase values were 408.3 ± 60.0 μs and 4.0 ± 1.8 mA in MS patients, versus 408.0 ± 62.4 μs and 3.8 ± 2.1 mA in controls. The differences were not significant in SDTC or rheobase values between the patients and controls (P = 0.988 for SDTC and P = 0.722 for rheobase). Conclusion. Our study showed no abnormality in relapsing remitting MS patients in terms of SDTC, which gives some indirect information about peripheral Na+ channel function. This may indicate that alterations in the Na+ channel pattern in central nervous system (CNS) couldnot be shown in the peripheral nervous system (PNS) in the MS patients by SDTC. The opinion that MS can be a kind of channelopathy might be proven by performing other axonal excitability tests or SDTC in progressive forms of MS.
Collapse
|
32
|
Constantinescu CS, Farooqi N, O'Brien K, Gran B. Experimental autoimmune encephalomyelitis (EAE) as a model for multiple sclerosis (MS). Br J Pharmacol 2012; 164:1079-106. [PMID: 21371012 DOI: 10.1111/j.1476-5381.2011.01302.x] [Citation(s) in RCA: 1029] [Impact Index Per Article: 85.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Experimental autoimmune encephalomyelitis (EAE) is the most commonly used experimental model for the human inflammatory demyelinating disease, multiple sclerosis (MS). EAE is a complex condition in which the interaction between a variety of immunopathological and neuropathological mechanisms leads to an approximation of the key pathological features of MS: inflammation, demyelination, axonal loss and gliosis. The counter-regulatory mechanisms of resolution of inflammation and remyelination also occur in EAE, which, therefore can also serve as a model for these processes. Moreover, EAE is often used as a model of cell-mediated organ-specific autoimmune conditions in general. EAE has a complex neuropharmacology, and many of the drugs that are in current or imminent use in MS have been developed, tested or validated on the basis of EAE studies. There is great heterogeneity in the susceptibility to the induction, the method of induction and the response to various immunological or neuropharmacological interventions, many of which are reviewed here. This makes EAE a very versatile system to use in translational neuro- and immunopharmacology, but the model needs to be tailored to the scientific question being asked. While creating difficulties and underscoring the inherent weaknesses of this model of MS in straightforward translation from EAE to the human disease, this variability also creates an opportunity to explore multiple facets of the immune and neural mechanisms of immune-mediated neuroinflammation and demyelination as well as intrinsic protective mechanisms. This allows the eventual development and preclinical testing of a wide range of potential therapeutic interventions.
Collapse
Affiliation(s)
- Cris S Constantinescu
- Division of Clinical Neurology, School of Clinical Sciences, University of Nottingham, Queen's Medical Centre, Nottingham, UK.
| | | | | | | |
Collapse
|
33
|
Cruz-Orengo L, Chen YJ, Kim JH, Dorsey D, Song SK, Klein RS. CXCR7 antagonism prevents axonal injury during experimental autoimmune encephalomyelitis as revealed by in vivo axial diffusivity. J Neuroinflammation 2011; 8:170. [PMID: 22145790 PMCID: PMC3305694 DOI: 10.1186/1742-2094-8-170] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2011] [Accepted: 12/06/2011] [Indexed: 12/13/2022] Open
Abstract
Background Multiple Sclerosis (MS) is characterized by the pathological trafficking of leukocytes into the central nervous system (CNS). Using the murine MS model, experimental autoimmune encephalomyelitis (EAE), we previously demonstrated that antagonism of the chemokine receptor CXCR7 blocks endothelial cell sequestration of CXCL12, thereby enhancing the abluminal localization of CXCR4-expressing leukocytes. CXCR7 antagonism led to decreased parenchymal entry of leukocytes and amelioration of ongoing disease during EAE. Of note, animals that received high doses of CXCR7 antagonist recovered to baseline function, as assessed by standard clinical scoring. Because functional recovery reflects axonal integrity, we utilized diffusion tensor imaging (DTI) to evaluate axonal injury in CXCR7 antagonist- versus vehicle-treated mice after recovery from EAE. Methods C57BL6/J mice underwent adoptive transfer of MOG-reactive Th1 cells and were treated daily with either CXCR7 antagonist or vehicle for 28 days; and then evaluated by DTI to assess for axonal injury. After imaging, spinal cords underwent histological analysis of myelin and oligodendrocytes via staining with luxol fast blue (LFB), and immunofluorescence for myelin basic protein (MBP) and glutathione S-transferase-π (GST-π). Detection of non-phosphorylated neurofilament H (NH-F) was also performed to detect injured axons. Statistical analysis for EAE scores, DTI parameters and non-phosphorylated NH-F immunofluorescence were done by ANOVA followed by Bonferroni post-hoc test. For all statistical analysis a p < 0.05 was considered significant. Results In vivo DTI maps of spinal cord ventrolateral white matter (VLWM) axial diffusivities of naïve and CXCR7 antagonist-treated mice were indistinguishable, while vehicle-treated animals exhibited decreased axial diffusivities. Quantitative differences in injured axons, as assessed via detection of non-phosphorylated NH-F, were consistent with axial diffusivity measurements. Overall, qualitative myelin content and presence of oligodendrocytes were similar in all treatment groups, as expected by their radial diffusivity values. Quantitative assessment of persistent inflammatory infiltrates revealed significant decreases within the parenchyma of CXCR7 antagonist-treated mice versus controls. Conclusions These data suggest that CXCR7 antagonism not only prevents persistent inflammation but also preserves axonal integrity. Thus, targeting CXCR7 modifies both disease severity and recovery during EAE, suggesting a role for this molecule in both phases of disease.
Collapse
Affiliation(s)
- Lillian Cruz-Orengo
- Department of Internal Medicine, Washington University School of Medicine, 660 S, Euclid Ave, St. Louis, MO 63110, USA
| | | | | | | | | | | |
Collapse
|
34
|
Brackenbury WJ, Isom LL. Na Channel β Subunits: Overachievers of the Ion Channel Family. Front Pharmacol 2011; 2:53. [PMID: 22007171 PMCID: PMC3181431 DOI: 10.3389/fphar.2011.00053] [Citation(s) in RCA: 229] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2011] [Accepted: 09/12/2011] [Indexed: 11/13/2022] Open
Abstract
Voltage-gated Na+ channels (VGSCs) in mammals contain a pore-forming α subunit and one or more β subunits. There are five mammalian β subunits in total: β1, β1B, β2, β3, and β4, encoded by four genes: SCN1B–SCN4B. With the exception of the SCN1B splice variant, β1B, the β subunits are type I topology transmembrane proteins. In contrast, β1B lacks a transmembrane domain and is a secreted protein. A growing body of work shows that VGSC β subunits are multifunctional. While they do not form the ion channel pore, β subunits alter gating, voltage-dependence, and kinetics of VGSCα subunits and thus regulate cellular excitability in vivo. In addition to their roles in channel modulation, β subunits are members of the immunoglobulin superfamily of cell adhesion molecules and regulate cell adhesion and migration. β subunits are also substrates for sequential proteolytic cleavage by secretases. An example of the multifunctional nature of β subunits is β1, encoded by SCN1B, that plays a critical role in neuronal migration and pathfinding during brain development, and whose function is dependent on Na+ current and γ-secretase activity. Functional deletion of SCN1B results in Dravet Syndrome, a severe and intractable pediatric epileptic encephalopathy. β subunits are emerging as key players in a wide variety of physiopathologies, including epilepsy, cardiac arrhythmia, multiple sclerosis, Huntington’s disease, neuropsychiatric disorders, neuropathic and inflammatory pain, and cancer. β subunits mediate multiple signaling pathways on different timescales, regulating electrical excitability, adhesion, migration, pathfinding, and transcription. Importantly, some β subunit functions may operate independently of α subunits. Thus, β subunits perform critical roles during development and disease. As such, they may prove useful in disease diagnosis and therapy.
Collapse
|
35
|
Interleukin-1β Inhibits Voltage-Gated Sodium Currents in a Time- and Dose-Dependent Manner in Cortical Neurons. Neurochem Res 2011; 36:1116-23. [DOI: 10.1007/s11064-011-0456-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/18/2011] [Indexed: 01/21/2023]
|
36
|
Hashiba N, Nagayama S, Araya SI, Inada H, Sonobe Y, Suzumura A, Matsui M. Phenytoin at optimum doses ameliorates experimental autoimmune encephalomyelitis via modulation of immunoregulatory cells. J Neuroimmunol 2011; 233:112-9. [PMID: 21237519 DOI: 10.1016/j.jneuroim.2010.12.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2010] [Revised: 11/26/2010] [Accepted: 12/14/2010] [Indexed: 12/21/2022]
Abstract
We investigated the optimum doses of phenytoin for treatment of myelin oligodendrocyte glycoprotein-induced experimental autoimmune encephalomyelitis (EAE). Oral and intraperitoneal administrations of 0.25 to 1.0mg per mouse (12.5-50mg/kg) 3 times a week improved the clinical course. Intraperitoneal injections of 1.0mg phenytoin were the most effective, as a significant reduction in EAE severity was seen after only 2 administrations with that protocol. Treatment efficacy was associated with amelioration of cellular infiltrates in the CNS, and an increase in CD4(+)Foxp3(+) and CD4(+)CD25(+)CD127(-) regulatory T cells as well as CD8(+) suppressor/cytotoxic T cells in blood.
Collapse
MESH Headings
- Animals
- Anticonvulsants/therapeutic use
- Disease Models, Animal
- Encephalomyelitis, Autoimmune, Experimental/drug therapy
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Immunologic Factors/therapeutic use
- Mice
- Mice, Inbred C57BL
- Phenytoin/therapeutic use
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/pathology
Collapse
Affiliation(s)
- Naomi Hashiba
- Department of Neurology, Kanazawa Medical University, Uchinada, Ishikawa Prefecture 920-0293, Japan
| | | | | | | | | | | | | |
Collapse
|
37
|
Abstract
Axons depend critically on axonal transport both for supplying materials and for communicating with cell bodies. This chapter looks at each activity, asking what aspects are essential for axon survival. Axonal transport declines in neurodegenerative disorders, such as Alzheimer's disease, amyotrophic lateral sclerosis, and multiple sclerosis, and in normal ageing, but whether all cargoes are equally affected and what limits axon survival remains unclear. Cargoes can be differentially blocked in some disorders, either individually or in groups. Each missing protein cargo results in localized loss-of-function that can be partially modeled by disrupting the corresponding gene, sometimes with surprising results. The axonal response to losing specific proteins also depends on the rates of protein turnover and on whether the protein can be locally synthesized. Among cargoes with important axonal roles are components of the PI3 kinase, Mek/Erk, and Jnk signaling pathways, which help to communicate with cell bodies and to regulate axonal transport itself. Bidirectional trafficking of Bdnf, NT-3, and other neurotrophic factors contribute to intra- and intercellular signaling, affecting the axon's cellular environment and survival. Finally, several adhesion molecules and gangliosides are key determinants of axon survival, probably by mediating axon-glia interactions. Thus, failure of long-distance intracellular transport can deprive axons of one, few, or many cargoes. This can lead to axon degeneration either directly, through the absence of essential axonal proteins, or indirectly, through failures in communication with cell bodies and nonneuronal cells.
Collapse
|
38
|
Kim DY, Gersbacher MT, Inquimbert P, Kovacs DM. Reduced sodium channel Na(v)1.1 levels in BACE1-null mice. J Biol Chem 2010; 286:8106-8116. [PMID: 21190943 DOI: 10.1074/jbc.m110.134692] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The Alzheimer BACE1 enzyme cleaves numerous substrates, with largely unknown physiological consequences. We have previously identified the contribution of elevated BACE1 activity to voltage-gated sodium channel Na(v)1.1 density and neuronal function. Here, we analyzed physiological changes in sodium channel metabolism in BACE1-null mice. Mechanistically, we first confirmed that endogenous BACE1 requires its substrate, the β-subunit Na(v)β(2), to regulate levels of the pore-forming α-subunit Na(v)1.1 in cultured primary neurons. Next, we analyzed sodium channel α-subunit levels in brains of BACE1-null mice at 1 and 3 months of age. At both ages, we found that Na(v)1.1 protein levels were significantly decreased in BACE1-null versus wild-type mouse brains, remaining unchanged in BACE1-heterozygous mouse brains. Interestingly, levels of Na(v)1.2 and Na(v)1.6 α-subunits also decreased in 1-month-old BACE1-null mice. In the hippocampus of BACE1-null mice, we found a robust 57% decrease of Na(v)1.1 levels. Next, we performed surface biotinylation studies in acutely dissociated hippocampal slices from BACE1-null mice. Hippocampal surface Na(v)1.1 levels were significantly decreased, but Na(v)1.2 surface levels were increased in BACE1-null mice perhaps as a compensatory mechanism for reduced surface Na(v)1.1. We also found that Na(v)β(2) processing and Na(v)1.1 mRNA levels were significantly decreased in brains of BACE1-null mice. This suggests a mechanism consistent with BACE1 activity regulating mRNA levels of the α-subunit Na(v)1.1 via cleavage of cell-surface Na(v)β(2). Together, our data show that endogenous BACE1 activity regulates total and surface levels of voltage-gated sodium channels in mouse brains. Both decreased Na(v)1.1 and elevated surface Na(v)1.2 may result in a seizure phenotype. Our data caution that therapeutic BACE1 activity inhibition in Alzheimer disease patients may affect Na(v)1 metabolism and alter neuronal membrane excitability in Alzheimer disease patients.
Collapse
Affiliation(s)
- Doo Yeon Kim
- From the Neurobiology of Disease Laboratory, Genetics and Aging Research Unit, Massachusetts General Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129 and
| | - Manuel T Gersbacher
- From the Neurobiology of Disease Laboratory, Genetics and Aging Research Unit, Massachusetts General Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129 and
| | - Perrine Inquimbert
- the Department of Neurology, F. M. Kirby Neurobiology Center, Children's Hospital, Boston, Massachusetts 02115
| | - Dora M Kovacs
- From the Neurobiology of Disease Laboratory, Genetics and Aging Research Unit, Massachusetts General Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129 and.
| |
Collapse
|
39
|
Zindler E, Zipp F. Neuronal injury in chronic CNS inflammation. Best Pract Res Clin Anaesthesiol 2010; 24:551-62. [PMID: 21619866 DOI: 10.1016/j.bpa.2010.11.001] [Citation(s) in RCA: 101] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2010] [Accepted: 11/01/2010] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Multiple sclerosis (MS) is the most common chronic inflammatory disease of the central nervous system which is characterized by inflammatory demyelination and neurodegeneration. Neurological symptoms include sensory disturbances, optic neuritis, limb weakness, ataxia, bladder dysfunction, cognitive deficits and fatigue. PATHOPHYSIOLOGY The inflammation process with MS is promoted by several inflammatory cytokines produced by the immune cells themselves and local resident cells like activated microglia. Consecutive damaging pathways involve the transmigration of activated B lymphocytes and plasma cells, which synthesize antibodies against the myelin sheath, boost the immune attack, and result in ultimate loss of myelin. Likewise, activated macrophages and microglia are present outside the lesions in the normal-appearing CNS tissue contributing to tissue damage. In parallel to inflammatory demyelination, axonal pathology occurs in the early phase which correlates with the number of infiltrating immune cells, and critically contributes to disease severity. The spectrum of neuronal white matter and cortical damage ranges from direct cell death to subtle neurodegenerative changes such as loss of dendritic ramification and the extent of neuronal damage is regarded as a critical factor for persisting neurological deficits. Under normal conditions, CNS microglia safeguards organ integrity by constantly scanning the tissue and responding rapidly to danger signals. The main task of microglial cells is to encapsulate dangerous foci and remove apoptotic cells and debris to protect the surrounding CNS tissue; this assists with tissue regeneration in toxin-induced demyelination. In the absence of lymphocytic inflammation and in the context of non-autoimmune, pathogen-associated triggered inflammation, microglial cells protect the neuronal compartment. These mechanisms seem to be inverted in MS and other chronic neurodegenerative disorders because activated microglia and peripherally derived macrophages are shifted towards a strongly pro-inflammatory phenotype and produce the proinflammatory cytokines TNF-α and interleukin (IL)1-β, as well as potentially neurotoxic substances including nitric oxide, oxygen radicals and proteolytic enzymes. Microglial silencing reduces clinical severity, demonstrating their active involvement in damage processes and in the immune attack against the CNS. In light of this, it is questionable whether microglia and monocyte-derived macrophages, the very last downstream effector cells in the immune reaction, actually have the capacity to influence their fate. It is more likely that the adaptive immune system orchestrates the attack against CNS cells and drives microglia and macrophages to attack oligodendrocytes and neurons. NEUROPROTECTIVE STRATEGIES Currently, Glatiramer acetate (GA) and the interferon-β (IFN-β) variants are established as first-line disease modifying treatments that reduce the relapse rate, ameliorate relapse severity and delay the progression of disability in patients with relapsing-remitting MS. Similarily, sphingosine-1-phosphate (S1P) receptor agonists which influence lymphocyte migration through T cells-trapping in secondary lymphatic organs ameliorates astrogliosis and promotes remyelination by acting on S1P-receptors on astrocytes and oligodendrocytes. Ion channel blockers (e.g. sodium channel blockers), currently used for other indications, are now tested in neurodegenerative diseases to restore intracellular ion homeostasis in neurons. Axonal degeneration was significantly reduced and functional outcome was improved during treatment with Phenytoin, Flecainide and Lamotrigine. Although evidence for a direct protective effect on axons is still missing, additional immune-modulatory actions of sodium channel blockers on microglia and macrophages are likely available. In vitro-studies in axons subjected to anoxia in vitro or exposure to elevated levels of nitric oxide (NO) in vivo demonstrated the involvement of a direct effect on axons. As increased intracellular calcium levels contribute to axonal damage through activation of different enzymes such as proteases, blockade of voltage gated calcium channels is another promising target. For example, nitrendipin and bepridil ameliorate axonal loss and clinical symptoms in different models of chronic neurodegeneration. In addition to these exogenous neuroprotective patheways, endogenous neuroprotective mechanisms including neurotrophins, (re)myelination and, neurogenesis support restauration of neuronal integrity.
Collapse
Affiliation(s)
- Eva Zindler
- Universitätsmedizin der Johannes Gutenberg Universität, Klinik und Poliklinik für Neurologie, Langenbeckstr. 1, 55131 Mainz, Germany
| | | |
Collapse
|
40
|
Kovacs DM, Gersbacher MT, Kim DY. Alzheimer's secretases regulate voltage-gated sodium channels. Neurosci Lett 2010; 486:68-72. [PMID: 20817076 DOI: 10.1016/j.neulet.2010.08.048] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2010] [Revised: 07/16/2010] [Accepted: 08/15/2010] [Indexed: 01/05/2023]
Abstract
BACE1 and presenilin (PS)/γ-secretase are primary proteolytic enzymes responsible for the generation of pathogenic amyloid β-peptides (Aβ) in Alzheimer's disease. We and others have found that β-subunits of the voltage-gated sodium channel (Na(v)βs) also undergo sequential proteolytic cleavages mediated by BACE1 and PS/γ-secretase. In a follow-up study, we reported that elevated BACE1 activity regulates total and surface expression of voltage-gated sodium channels (Na(v)1 channels) and thereby modulates sodium currents in neuronal cells and mouse brains. In this review, we focus on the molecular mechanism of how BACE1 and PS/γ-secretase regulate Na(v)1 channels in neuronal cells. We will also discuss potential physiological and pathological roles of BACE1- and PS/γ-secretase-mediated processing of Na(v)βs in relation to Na(v)1 channel function.
Collapse
Affiliation(s)
- Dora M Kovacs
- Neurobiology of Disease Laboratory, Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | | | | |
Collapse
|
41
|
Herz J, Zipp F, Siffrin V. Neurodegeneration in autoimmune CNS inflammation. Exp Neurol 2010; 225:9-17. [DOI: 10.1016/j.expneurol.2009.11.019] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2009] [Accepted: 11/24/2009] [Indexed: 02/06/2023]
|
42
|
Patino GA, Isom LL. Electrophysiology and beyond: multiple roles of Na+ channel β subunits in development and disease. Neurosci Lett 2010; 486:53-9. [PMID: 20600605 DOI: 10.1016/j.neulet.2010.06.050] [Citation(s) in RCA: 133] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2010] [Revised: 06/02/2010] [Accepted: 06/16/2010] [Indexed: 12/19/2022]
Abstract
Voltage-gated Na+ channel (VGSC) β Subunits are not "auxiliary." These multi-functional molecules not only modulate Na+ current (I(Na)), but also function as cell adhesion molecules (CAMs)-playing roles in aggregation, migration, invasion, neurite outgrowth, and axonal fasciculation. β subunits are integral members of VGSC signaling complexes at nodes of Ranvier, axon initial segments, and cardiac intercalated disks, regulating action potential propagation through critical intermolecular and cell-cell communication events. At least in vitro, many β subunit cell adhesive functions occur both in the presence and absence of pore-forming VGSC α subunits, and in vivo β subunits are expressed in excitable as well as non-excitable cells, thus β subunits may play important functional roles on their own, in the absence of α subunits. VGSC β1 subunits are essential for life and appear to be especially important during brain development. Mutations in β subunit genes result in a variety of human neurological and cardiovascular diseases. Moreover, some cancer cells exhibit alterations in β subunit expression during metastasis. In short, these proteins, originally thought of as merely accessory to α subunits, are critical players in their own right in human health and disease. Here we discuss the role of VGSC β subunits in the nervous system.
Collapse
Affiliation(s)
- Gustavo A Patino
- Department of Pharmacology and Neuroscience Program, University of Michigan, Ann Arbor, MI 48109, United States
| | | |
Collapse
|
43
|
Initiation and progression of axonopathy in experimental autoimmune encephalomyelitis. J Neurosci 2010; 29:14965-79. [PMID: 19940192 DOI: 10.1523/jneurosci.3794-09.2009] [Citation(s) in RCA: 112] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Axonal loss is the principal cause of chronic disability in multiple sclerosis and experimental autoimmune encephalomyelitis (EAE). In C57BL/6 mice with EAE induced by immunization with myelin oligodendrocyte glycoprotein peptide 35-55, the first evidences of axonal damage in spinal cord were in acute subpial and perivascular foci of infiltrating neutrophils and lymphocytes and included intra-axonal accumulations of the endovesicular Toll-like receptor TLR8, and the inflammasome protein NAcht leucine-rich repeat protein 1 (NALP1). Later in the course of this illness, focal inflammatory infiltrates disappeared from the spinal cord, but there was persistent activation of spinal cord innate immunity and progressive, bilaterally symmetric loss of small-diameter corticospinal tract axons. These results support the hypothesis that both contact-dependent and paracrine interactions of systemic inflammatory cells with axons and an innate immune-mediated neurodegenerative process contribute to axonal loss in this multiple sclerosis model.
Collapse
|
44
|
McEwen DP, Chen C, Meadows LS, Lopez-Santiago L, Isom LL. The voltage-gated Na+ channel beta3 subunit does not mediate trans homophilic cell adhesion or associate with the cell adhesion molecule contactin. Neurosci Lett 2009; 462:272-5. [PMID: 19596049 DOI: 10.1016/j.neulet.2009.07.020] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2009] [Revised: 07/07/2009] [Accepted: 07/08/2009] [Indexed: 10/20/2022]
Abstract
Voltage-gated Na(+) channel (VGSC) beta1 and beta2 subunits are multifunctional, serving as both channel modulators and cell adhesion molecules (CAMs). The purpose of this study was to determine whether VGSC beta3 subunits function as CAMs. The beta3 extracellular domain is highly homologous to beta1, suggesting that beta3 may also be a functional CAM. We investigated the trans homophilic cell adhesive properties of beta3, its association with the beta1-interacting CAM contactin, as well as its ability to interact with the cytoskeletal protein ankyrin. Our results demonstrate that, unlike beta1, beta3 does not participate in trans homophilic cell-cell adhesion or associate with contactin. Further, beta3 does not associate with ankyrin(G) in a heterologous system. Previous studies have shown that beta3 interacts with the CAM neurofascin-186 but not with VGSC beta1. Taken together, these findings suggest that, although beta1 and beta3 exhibit similar channel modulatory properties in heterologous systems, these subunits differ with regard to their homophilic and heterophilic CAM binding profiles.
Collapse
Affiliation(s)
- Dyke P McEwen
- Department of Pharmacology, University of Michigan, Ann Arbor, MI 48109-5632, USA
| | | | | | | | | |
Collapse
|