1
|
Fang H, Li M, Yang J, Ma S, Zhang L, Yang H, Tang Q, Cao J, Yang W. Repressing iron overload ameliorates central post-stroke pain via the Hdac2-Kv1.2 axis in a rat model of hemorrhagic stroke. Neural Regen Res 2024; 19:2708-2722. [PMID: 38595289 PMCID: PMC11168507 DOI: 10.4103/nrr.nrr-d-23-01498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 11/21/2023] [Accepted: 02/04/2024] [Indexed: 04/11/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202412000-00027/figure1/v/2024-04-08T165401Z/r/image-tiff Thalamic hemorrhage can lead to the development of central post-stroke pain. Changes in histone acetylation levels, which are regulated by histone deacetylases, affect the excitability of neurons surrounding the hemorrhagic area. However, the regulatory mechanism of histone deacetylases in central post-stroke pain remains unclear. Here, we show that iron overload leads to an increase in histone deacetylase 2 expression in damaged ventral posterolateral nucleus neurons. Inhibiting this increase restored histone H3 acetylation in the Kcna2 promoter region of the voltage-dependent potassium (Kv) channel subunit gene in a rat model of central post-stroke pain, thereby increasing Kcna2 expression and relieving central pain. However, in the absence of nerve injury, increasing histone deacetylase 2 expression decreased Kcna2 expression, decreased Kv current, increased the excitability of neurons in the ventral posterolateral nucleus area, and led to neuropathic pain symptoms. Moreover, treatment with the iron chelator deferiprone effectively reduced iron overload in the ventral posterolateral nucleus after intracerebral hemorrhage, reversed histone deacetylase 2 upregulation and Kv1.2 downregulation, and alleviated mechanical hypersensitivity in central post-stroke pain rats. These results suggest that histone deacetylase 2 upregulation and Kv1.2 downregulation, mediated by iron overload, are important factors in central post-stroke pain pathogenesis and could serve as new targets for central post-stroke pain treatment.
Collapse
Affiliation(s)
- He Fang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Mengjie Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Jingchen Yang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Shunping Ma
- Department of Nutrition, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Li Zhang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Hongqi Yang
- Department of Neurology, Henan Provincial People’s Hospital, Zhengzhou, Henan Province, China
| | - Qiongyan Tang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Jing Cao
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China
- Neuroscience Research Institute, Zhengzhou University Academy of Medical Sciences, Zhengzhou, Henan Province, China
| | - Weimin Yang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| |
Collapse
|
2
|
Lu Y, Sciaccotta F, Kiely L, Bellanger B, Erisir A, Meliza CD. Rapid, Activity-Dependent Intrinsic Plasticity in the Developing Zebra Finch Auditory Cortex. J Neurosci 2023; 43:6872-6883. [PMID: 37648449 PMCID: PMC10573762 DOI: 10.1523/jneurosci.0354-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 07/14/2023] [Accepted: 08/23/2023] [Indexed: 09/01/2023] Open
Abstract
The acoustic environment an animal experiences early in life shapes the structure and function of its auditory system. This process of experience-dependent development is thought to be primarily orchestrated by potentiation and depression of synapses, but plasticity of intrinsic voltage dynamics may also contribute. Here, we show that in juvenile male and female zebra finches, neurons in a cortical-level auditory area, the caudal mesopallium (CM), can rapidly change their firing dynamics. This plasticity was only observed in birds that were reared in a complex acoustic and social environment, which also caused increased expression of the low-threshold potassium channel Kv1.1 in the plasma membrane and endoplasmic reticulum (ER). Intrinsic plasticity depended on activity, was reversed by blocking low-threshold potassium currents, and was prevented by blocking intracellular calcium signaling. Taken together, these results suggest that Kv1.1 is rapidly mobilized to the plasma membrane by activity-dependent elevation of intracellular calcium. This produces a shift in the excitability and temporal integration of CM neurons that may be permissive for auditory learning in complex acoustic environments during a crucial period for the development of vocal perception and production.SIGNIFICANCE STATEMENT Neurons can change not only the strength of their connections to other neurons, but also how they integrate synaptic currents to produce patterns of action potentials. In contrast to synaptic plasticity, the mechanisms and functional roles of intrinisic plasticity remain poorly understood. We found that neurons in the zebra finch auditory cortex can rapidly shift their spiking dynamics within a few minutes in response to intracellular stimulation. This plasticity involves increased conductance of a low-threshold potassium current associated with the Kv1.1 channel, but it only occurs in birds reared in a rich acoustic environment. Thus, auditory experience regulates a mechanism of neural plasticity that allows neurons to rapidly adapt their firing dynamics to stimulation.
Collapse
Affiliation(s)
| | | | | | | | - Alev Erisir
- Psychology Department
- Neuroscience Graduate Program, University of Virginia, Charlottesville, Virginia 22904
| | - C Daniel Meliza
- Psychology Department
- Neuroscience Graduate Program, University of Virginia, Charlottesville, Virginia 22904
| |
Collapse
|
3
|
Dziadkowiak E, Nowakowska-Kotas M, Budrewicz S, Koszewicz M. Pathology of Initial Axon Segments in Chronic Inflammatory Demyelinating Polyradiculoneuropathy and Related Disorders. Int J Mol Sci 2022; 23:13621. [PMID: 36362407 PMCID: PMC9658771 DOI: 10.3390/ijms232113621] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 10/27/2022] [Accepted: 11/01/2022] [Indexed: 07/30/2023] Open
Abstract
The diagnosis of chronic inflammatory demyelinating polyradiculoneuropathy (CIDP) is based on a combination of clinical, electrodiagnostic and laboratory features. The different entities of the disease include chronic immune sensory polyradiculopathy (CISP) and autoimmune nodopathies. It is debatable whether CIDP occurring in the course of other conditions, i.e., monoclonal IgG or IgA gammopathy, should be treated as a separate disease entity from idiopathic CIDP. This study aims to evaluate the molecular differences of the nodes of Ranvier and the initial axon segment (AIS) and juxtaparanode region (JXP) as the potential cause of phenotypic variation of CIDP while also seeking new pathomechanisms since JXP is sequestered behind the paranode and autoantibodies may not access the site easily. The authors initially present the structure of the different parts of the neuron and its functional significance, then discuss the problem of whether damage to the juxtaparanodal region, Schwann cells and axons could cause CIDP or if these damages should be separated as separate disease entities. In particular, AIS's importance for modulating neural excitability and carrying out transport along the axon is highlighted. The disclosure of specific pathomechanisms, including novel target antigens, in the heterogeneous CIDP syndrome is important for diagnosing and treating these patients.
Collapse
|
4
|
Tanenhaus A, Stowe T, Young A, McLaughlin J, Aeran R, Lin IW, Li J, Hosur R, Chen M, Leedy J, Chou T, Pillay S, Vila MC, Kearney JA, Moorhead M, Belle A, Tagliatela S. Cell-Selective Adeno-Associated Virus-Mediated SCN1A Gene Regulation Therapy Rescues Mortality and Seizure Phenotypes in a Dravet Syndrome Mouse Model and Is Well Tolerated in Nonhuman Primates. Hum Gene Ther 2022; 33:579-597. [PMID: 35435735 PMCID: PMC9242722 DOI: 10.1089/hum.2022.037] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Dravet syndrome (DS) is a developmental and epileptic encephalopathy caused by monoallelic loss-of-function variants in the SCN1A gene. SCN1A encodes for the alpha subunit of the voltage-gated type I sodium channel (NaV1.1), the primary voltage-gated sodium channel responsible for generation of action potentials in GABAergic inhibitory interneurons. In these studies, we tested the efficacy of an adeno-associated virus serotype 9 (AAV9) SCN1A gene regulation therapy, AAV9-REGABA-eTFSCN1A, designed to target transgene expression to GABAergic inhibitory neurons and reduce off-target expression within excitatory cells, in the Scn1a+/- mouse model of DS. Biodistribution and preliminary safety were evaluated in nonhuman primates (NHPs). AAV9-REGABA-eTFSCN1A was engineered to upregulate SCN1A expression levels within GABAergic inhibitory interneurons to correct the underlying haploinsufficiency and circuit dysfunction. A single bilateral intracerebroventricular (ICV) injection of AAV9-REGABA-eTFSCN1A in Scn1a+/- postnatal day 1 mice led to increased SCN1A mRNA transcripts, specifically within GABAergic inhibitory interneurons, and NaV1.1 protein levels in the brain. This was associated with a significant decrease in the occurrence of spontaneous and hyperthermia-induced seizures, and prolonged survival for over a year. In NHPs, delivery of AAV9-REGABA-eTFSCN1A by unilateral ICV injection led to widespread vector biodistribution and transgene expression throughout the brain, including key structures involved in epilepsy and cognitive behaviors, such as hippocampus and cortex. AAV9-REGABA-eTFSCN1A was well tolerated, with no adverse events during administration, no detectable changes in clinical observations, no adverse findings in histopathology, and no dorsal root ganglion-related toxicity. Our results support the clinical development of AAV9-REGABA-eTFSCN1A (ETX101) as an effective and targeted disease-modifying approach to SCN1A+ DS.
Collapse
Affiliation(s)
- Annie Tanenhaus
- Encoded Therapeutics, Inc., South San Francisco, California, USA
| | - Timothy Stowe
- Encoded Therapeutics, Inc., South San Francisco, California, USA
| | - Andrew Young
- Encoded Therapeutics, Inc., South San Francisco, California, USA
| | - John McLaughlin
- Encoded Therapeutics, Inc., South San Francisco, California, USA
| | - Rangoli Aeran
- Encoded Therapeutics, Inc., South San Francisco, California, USA
| | - I. Winnie Lin
- Encoded Therapeutics, Inc., South San Francisco, California, USA
| | - Jianmin Li
- Encoded Therapeutics, Inc., South San Francisco, California, USA
| | | | - Ming Chen
- Encoded Therapeutics, Inc., South San Francisco, California, USA
| | - Jennifer Leedy
- Encoded Therapeutics, Inc., South San Francisco, California, USA
| | - Tiffany Chou
- Encoded Therapeutics, Inc., South San Francisco, California, USA
| | - Sirika Pillay
- Encoded Therapeutics, Inc., South San Francisco, California, USA
| | | | - Jennifer A. Kearney
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Martin Moorhead
- Encoded Therapeutics, Inc., South San Francisco, California, USA
| | - Archana Belle
- Encoded Therapeutics, Inc., South San Francisco, California, USA
| | - Stephanie Tagliatela
- Encoded Therapeutics, Inc., South San Francisco, California, USA.,Correspondence: Stephanie Tagliatela, Encoded Therapeutics, Inc., 341 Oyster Point Boulevard, South San Francisco, CA 94080, USA.
| |
Collapse
|
5
|
Abd El-Aziz TM, Soares AG, Mironova E, Boiko N, Kaur A, Archer CR, Stockand JD, Berman JM. Mechanisms and consequences of casein kinase II and ankyrin-3 regulation of the epithelial Na + channel. Sci Rep 2021; 11:14600. [PMID: 34272444 PMCID: PMC8285517 DOI: 10.1038/s41598-021-94118-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Accepted: 07/01/2021] [Indexed: 01/21/2023] Open
Abstract
Activity of the Epithelial Na+ Channel (ENaC) in the distal nephron fine-tunes renal sodium excretion. Appropriate sodium excretion is a key factor in the regulation of blood pressure. Consequently, abnormalities in ENaC function can cause hypertension. Casein Kinase II (CKII) phosphorylates ENaC. The CKII phosphorylation site in ENaC resides within a canonical "anchor" ankyrin binding motif. CKII-dependent phosphorylation of ENaC is necessary and sufficient to increase channel activity and is thought to influence channel trafficking in a manner that increases activity. We test here the hypothesis that phosphorylation of ENaC by CKII within an anchor motif is necessary for ankyrin-3 (Ank-3) regulation of the channel, which is required for normal channel locale and function, and the proper regulation of renal sodium excretion. This was addressed using a fluorescence imaging strategy combining total internal reflection fluorescence (TIRF) microscopy with fluorescence recovery after photobleaching (FRAP) to quantify ENaC expression in the plasma membrane in living cells; and electrophysiology to quantify ENaC activity in split-open collecting ducts from principal cell-specific Ank-3 knockout mice. Sodium excretion studies also were performed in parallel in this knockout mouse. In addition, we substituted a key serine residue in the consensus CKII site in β-ENaC with alanine to abrogate phosphorylation and disrupt the anchor motif. Findings show that disrupting CKII signaling decreases ENaC activity by decreasing expression in the plasma membrane. In the principal cell-specific Ank-3 KO mouse, ENaC activity and sodium excretion were significantly decreased and increased, respectively. These results are consistent with CKII phosphorylation of ENaC functioning as a "switch" that favors Ank-3 binding to increase channel activity.
Collapse
Affiliation(s)
- Tarek Mohamed Abd El-Aziz
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center At San Antonio, San Antonio, TX, 78229-3900, USA
- Zoology Department, Faculty of Science, Minia University, El-Minia, 61519, Egypt
| | - Antonio G Soares
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center At San Antonio, San Antonio, TX, 78229-3900, USA
| | - Elena Mironova
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center At San Antonio, San Antonio, TX, 78229-3900, USA
| | - Nina Boiko
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center At San Antonio, San Antonio, TX, 78229-3900, USA
| | - Amanpreet Kaur
- Department of Biochemistry, University of Washington, Seattle, Washington, 98195, USA
| | - Crystal R Archer
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center At San Antonio, San Antonio, TX, 78229-3900, USA
| | - James D Stockand
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center At San Antonio, San Antonio, TX, 78229-3900, USA.
| | - Jonathan M Berman
- Department of Basic Science, New York Institute of Technology College of Osteopathic Medicine at Arkansas State University, Jonesboro, AR, 72401, USA
| |
Collapse
|
6
|
Askland KD, Strong D, Wright MN, Moore JH. The Translational Machine: A novel machine-learning approach to illuminate complex genetic architectures. Genet Epidemiol 2021; 45:485-536. [PMID: 33942369 DOI: 10.1002/gepi.22383] [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: 10/23/2020] [Revised: 03/05/2021] [Accepted: 03/23/2021] [Indexed: 11/08/2022]
Abstract
The Translational Machine (TM) is a machine learning (ML)-based analytic pipeline that translates genotypic/variant call data into biologically contextualized features that richly characterize complex variant architectures and permit greater interpretability and biological replication. It also reduces potentially confounding effects of population substructure on outcome prediction. The TM consists of three main components. First, replicable but flexible feature engineering procedures translate genome-scale data into biologically informative features that appropriately contextualize simple variant calls/genotypes within biological and functional contexts. Second, model-free, nonparametric ML-based feature filtering procedures empirically reduce dimensionality and noise of both original genotype calls and engineered features. Third, a powerful ML algorithm for feature selection is used to differentiate risk variant contributions across variant frequency and functional prediction spectra. The TM simultaneously evaluates potential contributions of variants operative under polygenic and heterogeneous models of genetic architecture. Our TM enables integration of biological information (e.g., genomic annotations) within conceptual frameworks akin to geneset-/pathways-based and collapsing methods, but overcomes some of these methods' limitations. The full TM pipeline is executed in R. Our approach and initial findings from its application to a whole-exome schizophrenia case-control data set are presented. These TM procedures extend the findings of the primary investigation and yield novel results.
Collapse
Affiliation(s)
- Kathleen D Askland
- Waypoint Centre for Mental Health Care Penetanguishene, University of Toronto, Toronto, Ontario, Canada
| | - David Strong
- Department of Family Medicine and Public Health, University of California San Diego, San Diego, California, USA
| | - Marvin N Wright
- Department Biometry and Data Management, Leibniz Institute for Prevention Research and Epidemiology - BIPS GmbH, Germany
| | - Jason H Moore
- Department of Biostatistics, Epidemiology, & Informatics, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
7
|
McCoy MT, Jayanthi S, Cadet JL. Potassium Channels and Their Potential Roles in Substance Use Disorders. Int J Mol Sci 2021; 22:1249. [PMID: 33513859 PMCID: PMC7865894 DOI: 10.3390/ijms22031249] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/19/2021] [Accepted: 01/22/2021] [Indexed: 01/12/2023] Open
Abstract
Substance use disorders (SUDs) are ubiquitous throughout the world. However, much remains to be done to develop pharmacotherapies that are very efficacious because the focus has been mostly on using dopaminergic agents or opioid agonists. Herein we discuss the potential of using potassium channel activators in SUD treatment because evidence has accumulated to support a role of these channels in the effects of rewarding drugs. Potassium channels regulate neuronal action potential via effects on threshold, burst firing, and firing frequency. They are located in brain regions identified as important for the behavioral responses to rewarding drugs. In addition, their expression profiles are influenced by administration of rewarding substances. Genetic studies have also implicated variants in genes that encode potassium channels. Importantly, administration of potassium agonists have been shown to reduce alcohol intake and to augment the behavioral effects of opioid drugs. Potassium channel expression is also increased in animals with reduced intake of methamphetamine. Together, these results support the idea of further investing in studies that focus on elucidating the role of potassium channels as targets for therapeutic interventions against SUDs.
Collapse
Affiliation(s)
| | | | - Jean Lud Cadet
- Molecular Neuropsychiatry Research Branch, NIDA Intramural Research Program, Baltimore, MD 21224, USA; (M.T.M.); (S.J.)
| |
Collapse
|
8
|
Assembly and Function of the Juxtaparanodal Kv1 Complex in Health and Disease. Life (Basel) 2020; 11:life11010008. [PMID: 33374190 PMCID: PMC7824554 DOI: 10.3390/life11010008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 12/21/2020] [Accepted: 12/23/2020] [Indexed: 02/07/2023] Open
Abstract
The precise axonal distribution of specific potassium channels is known to secure the shape and frequency of action potentials in myelinated fibers. The low-threshold voltage-gated Kv1 channels located at the axon initial segment have a significant influence on spike initiation and waveform. Their role remains partially understood at the juxtaparanodes where they are trapped under the compact myelin bordering the nodes of Ranvier in physiological conditions. However, the exposure of Kv1 channels in de- or dys-myelinating neuropathy results in alteration of saltatory conduction. Moreover, cell adhesion molecules associated with the Kv1 complex, including Caspr2, Contactin2, and LGI1, are target antigens in autoimmune diseases associated with hyperexcitability such as encephalitis, neuromyotonia, or neuropathic pain. The clustering of Kv1.1/Kv1.2 channels at the axon initial segment and juxtaparanodes is based on interactions with cell adhesion molecules and cytoskeletal linkers. This review will focus on the trafficking and assembly of the axonal Kv1 complex in the peripheral and central nervous system (PNS and CNS), during development, and in health and disease.
Collapse
|
9
|
Lee M, Liu YC, Chen C, Lu CH, Lu ST, Huang TN, Hsu MT, Hsueh YP, Cheng PL. Ecm29-mediated proteasomal distribution modulates excitatory GABA responses in the developing brain. J Cell Biol 2020; 219:133566. [PMID: 31910261 PMCID: PMC7041676 DOI: 10.1083/jcb.201903033] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 10/14/2019] [Accepted: 11/13/2019] [Indexed: 12/31/2022] Open
Abstract
Neuronal GABAergic responses switch from excitatory to inhibitory at an early postnatal period in rodents. The timing of this switch is controlled by intracellular Cl− concentrations, but factors determining local levels of cation-chloride cotransporters remain elusive. Here, we report that local abundance of the chloride importer NKCC1 and timely emergence of GABAergic inhibition are modulated by proteasome distribution, which is mediated through interactions of proteasomes with the adaptor Ecm29 and the axon initial segment (AIS) scaffold protein ankyrin G. Mechanistically, both the Ecm29 N-terminal domain and an intact AIS structure are required for transport and tethering of proteasomes in the AIS region. In mice, Ecm29 knockout (KO) in neurons increases the density of NKCC1 protein in the AIS region, a change that positively correlates with a delay in the GABAergic response switch. Phenotypically, Ecm29 KO mice showed increased firing frequency of action potentials at early postnatal ages and were hypersusceptible to chemically induced convulsive seizures. Finally, Ecm29 KO neurons exhibited accelerated AIS developmental positioning, reflecting a perturbed AIS morphological plastic response to hyperexcitability arising from proteasome inhibition, a phenotype rescued by ectopic Ecm29 expression or NKCC1 inhibition. Together, our findings support the idea that neuronal maturation requires regulation of proteasomal distribution controlled by Ecm29.
Collapse
Affiliation(s)
- Min Lee
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Yen-Chen Liu
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Chen Chen
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Chi-Huan Lu
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Shao-Tzu Lu
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Tzyy-Nan Huang
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Meng-Tsung Hsu
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Yi-Ping Hsueh
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Pei-Lin Cheng
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
10
|
Wu RN, Hung WC, Chen CT, Tsai LP, Lai WS, Min MY, Wong SB. Firing activity of locus coeruleus noradrenergic neurons decreases in necdin-deficient mice, an animal model of Prader-Willi syndrome. J Neurodev Disord 2020; 12:21. [PMID: 32727346 PMCID: PMC7389383 DOI: 10.1186/s11689-020-09323-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 07/17/2020] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Prader-Willi syndrome (PWS) is a neurodevelopmental disorder characterized by multiple respiratory, cognitive, endocrine, and behavioral symptoms, such as central apnea, intellectual disabilities, exaggerated stress responses, and temper tantrums. The locus coeruleus noradrenergic system (LC-NE) modulates a diverse range of behaviors, including arousal, learning, pain modulation, and stress-induced negative affective states, which are possibly correlated with the pathogenesis of PWS phenotypes. Therefore, we evaluated the LC-NE neuronal activity of necdin-deficient mice, an animal model of PWS. METHODS Heterozygous necdin-deficient mice (B6.Cg-Ndntm1ky) were bred from wild-type (WT) females to generate WT (+m/+p) and heterozygotes (+m/-p) animals, which were examined of LC-NE neuronal activity, developmental reflexes, and plethysmography. RESULTS On slice electrophysiology, LC-NE neurons of Ndntm1ky mice with necdin deficiency showed significantly decreased spontaneous activities and impaired excitability, which was mediated by enhanced A-type voltage-dependent potassium currents. Ndntm1ky mice also exhibited the neonatal phenotypes of PWS, such as hypotonia and blunt respiratory responses to hypercapnia. CONCLUSIONS LC-NE neuronal firing activity decreased in necdin-deficient mice, suggesting that LC, the primary source of norepinephrine in the central nervous system, is possibly involved in PWS pathogenesis.
Collapse
Affiliation(s)
- Rui-Ni Wu
- Department of Pediatrics, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, No. 289, Jiangguo Rd, Xindian Dist, New Taipei City, 23142, Taiwan
| | - Wei-Chen Hung
- Department of Life Science, College of Life Science, National Taiwan University, No. 1, Sec 4, Roosevelt Rd, Taipei, 10617, Taiwan
| | - Ching-Tsuey Chen
- Department of Life Science, College of Life Science, National Taiwan University, No. 1, Sec 4, Roosevelt Rd, Taipei, 10617, Taiwan
| | - Li-Ping Tsai
- Department of Pediatrics, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, No. 289, Jiangguo Rd, Xindian Dist, New Taipei City, 23142, Taiwan
- School of Medicine, Tzu Chi University, No. 701, Sec 3, Jhongyang Rd, Hualien, 97071, Taiwan
| | - Wen-Sung Lai
- Department of Psychology, National Taiwan University, No. 1, Sec 4, Roosevelt Rd, Taipei, 10617, Taiwan
| | - Ming-Yuan Min
- Department of Life Science, College of Life Science, National Taiwan University, No. 1, Sec 4, Roosevelt Rd, Taipei, 10617, Taiwan
| | - Shi-Bing Wong
- Department of Pediatrics, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, No. 289, Jiangguo Rd, Xindian Dist, New Taipei City, 23142, Taiwan.
- School of Medicine, Tzu Chi University, No. 701, Sec 3, Jhongyang Rd, Hualien, 97071, Taiwan.
| |
Collapse
|
11
|
Gavrilovici C, Jiang Y, Kiroski I, Teskey GC, Rho JM, Nguyen MD. Postnatal Role of the Cytoskeleton in Adult Epileptogenesis. Cereb Cortex Commun 2020; 1:tgaa024. [PMID: 32864616 PMCID: PMC7446231 DOI: 10.1093/texcom/tgaa024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 06/08/2020] [Accepted: 06/11/2020] [Indexed: 02/07/2023] Open
Abstract
Mutations in cytoskeletal proteins can cause early infantile and childhood epilepsies by misplacing newly born neurons and altering neuronal connectivity. In the adult epileptic brain, cytoskeletal disruption is often viewed as being secondary to aberrant neuronal activity and/or death, and hence simply represents an epiphenomenon. Here, we review the emerging evidence collected in animal models and human studies implicating the cytoskeleton as a potential causative factor in adult epileptogenesis. Based on the emerging evidence, we propose that cytoskeletal disruption may be an important pathogenic mechanism in the mature epileptic brain.
Collapse
Affiliation(s)
- Cezar Gavrilovici
- Departments of Neurosciences & Pediatrics, University of California San Diego, Rady Children’s Hospital San Diego, San Diego, CA 92123, USA
| | - Yulan Jiang
- Departments of Clinical Neurosciences, Cell Biology & Anatomy, and Biochemistry & Molecular Biology, Hotchkiss Brain Institute, Alberta Children Hospital Research Institute, University of Calgary, Calgary T2N 4N1, Canada
| | - Ivana Kiroski
- Departments of Clinical Neurosciences, Cell Biology & Anatomy, and Biochemistry & Molecular Biology, Hotchkiss Brain Institute, Alberta Children Hospital Research Institute, University of Calgary, Calgary T2N 4N1, Canada
| | - G Campbell Teskey
- Department of Cell Biology & Anatomy, Hotchkiss Brain Institute, Alberta Children Hospital Research Institute, University of Calgary, Calgary T2N 4N1, Canada
| | - Jong M Rho
- Departments of Neurosciences & Pediatrics, University of California San Diego, Rady Children’s Hospital San Diego, San Diego, CA 92123, USA
| | - Minh Dang Nguyen
- Departments of Clinical Neurosciences, Cell Biology & Anatomy, and Biochemistry & Molecular Biology, Hotchkiss Brain Institute, Alberta Children Hospital Research Institute, University of Calgary, Calgary T2N 4N1, Canada
| |
Collapse
|
12
|
Jayanthi S, Torres OV, Ladenheim B, Cadet JL. A Single Prior Injection of Methamphetamine Enhances Methamphetamine Self-Administration (SA) and Blocks SA-Induced Changes in DNA Methylation and mRNA Expression of Potassium Channels in the Rat Nucleus Accumbens. Mol Neurobiol 2019; 57:1459-1472. [PMID: 31758400 PMCID: PMC7060962 DOI: 10.1007/s12035-019-01830-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 11/01/2019] [Indexed: 12/27/2022]
Abstract
The transition from occasional to escalated psychostimulant use is accelerated by prior drug exposure. These behavioral observations may be related to long-lasting transcriptional and/or epigenetic changes induced by the drug pre-exposure. Herein, we investigated if a single methamphetamine (METH) injection would enhance METH self-administration (SA) and impact any METH SA-induced epigenetic or transcriptional alterations. We thus injected a single METH dose (10 mg/kg) or saline to rats before training them to self-administer METH or saline. There were three experimental groups in SA experiments: (1) a single saline injection followed by saline SA (SS); (2) a single saline injection followed by METH SA (SM); and (3) a single METH injection followed by METH SA (MM). METH-pretreated rats escalated METH SA earlier and took more METH than saline-pretreated animals. Both groups showed similar incubation of cue-induced METH craving. Because compulsive METH takers and METH-abstinent rats show differences in potassium (K+) channel mRNA levels in their nucleus accumbens (NAc), we wondered if K+ channel expression might also help to distinguish between SM and MM groups. We found increases in mRNA and protein expression of shaker-related voltage-gated K+ channels (Kv1: Kcna1, Kcna3, and Kcna6) and calcium-activated K+ channels (Kcnn1) in the SM compared to MM rats. SM rats also showed decreased DNA methylation at the CpG-rich sites near the promoter region of Kcna1, Kcna3 and Kcnn1 genes in comparison to MM rats. Together, these results provide additional evidence for potentially using K+ channels as therapeutic targets against METH use disorder.
Collapse
Affiliation(s)
- Subramaniam Jayanthi
- Molecular Neuropsychiatry Research Branch, Intramural Research Program, NIDA/NIH/DHHS, 251 Bayview Boulevard, Baltimore, MD, 21224, USA
| | - Oscar V Torres
- Department of Behavioral Sciences, San Diego Mesa College, San Diego, CA, USA
| | - Bruce Ladenheim
- Molecular Neuropsychiatry Research Branch, Intramural Research Program, NIDA/NIH/DHHS, 251 Bayview Boulevard, Baltimore, MD, 21224, USA
| | - Jean Lud Cadet
- Molecular Neuropsychiatry Research Branch, Intramural Research Program, NIDA/NIH/DHHS, 251 Bayview Boulevard, Baltimore, MD, 21224, USA.
| |
Collapse
|
13
|
Metabolic regulation of Kv channels and cardiac repolarization by Kvβ2 subunits. J Mol Cell Cardiol 2019; 137:93-106. [PMID: 31639389 DOI: 10.1016/j.yjmcc.2019.09.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 09/24/2019] [Accepted: 09/28/2019] [Indexed: 11/21/2022]
Abstract
Voltage-gated potassium (Kv) channels control myocardial repolarization. Pore-forming Kvα proteins associate with intracellular Kvβ subunits, which bind pyridine nucleotides with high affinity and differentially regulate channel trafficking, plasmalemmal localization and gating properties. Nevertheless, it is unclear how Kvβ subunits regulate myocardial K+ currents and repolarization. Here, we tested the hypothesis that Kvβ2 subunits regulate the expression of myocardial Kv channels and confer redox sensitivity to Kv current and cardiac repolarization. Co-immunoprecipitation and in situ proximity ligation showed that in cardiac myocytes, Kvβ2 interacts with Kv1.4, Kv1.5, Kv4.2, and Kv4.3. Cardiac myocytes from mice lacking Kcnab2 (Kvβ2-/-) had smaller cross sectional areas, reduced sarcolemmal abundance of Kvα binding partners, reduced Ito, IK,slow1, and IK,slow2 densities, and prolonged action potential duration compared with myocytes from wild type mice. These differences in Kvβ2-/- mice were associated with greater P wave duration and QT interval in electrocardiograms, and lower ejection fraction, fractional shortening, and left ventricular mass in echocardiographic and morphological assessments. Direct intracellular dialysis with a high NAD(P)H:NAD(P)+ accelerated Kv inactivation in wild type, but not Kvβ2-/- myocytes. Furthermore, elevated extracellular levels of lactate increased [NADH]i and prolonged action potential duration in wild type cardiac myocytes and perfused wild type, but not Kvβ2-/-, hearts. Taken together, these results suggest that Kvβ2 regulates myocardial electrical activity by supporting the functional expression of proteins that generate Ito and IK,slow, and imparting redox and metabolic sensitivity to Kv channels, thereby coupling cardiac repolarization to myocyte metabolism.
Collapse
|
14
|
Simultaneous electrophysiological and morphological assessment of functional damage to neural networks in vitro after 30-300 g impacts. Sci Rep 2019; 9:14994. [PMID: 31628381 PMCID: PMC6802386 DOI: 10.1038/s41598-019-51541-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 09/26/2019] [Indexed: 11/08/2022] Open
Abstract
An enigma of mild traumatic brain injury are observations of substantial behavior and performance deficits in the absence of bleeding or other observable structural damage. Altered behavior and performance reflect changes in action potential (AP) patterns within neuronal networks, which could result from subtle subcellular responses that affect synaptic efficacy and AP production. The aim of this study was to investigate and quantify network activity changes after simulated concussions in vitro and therewith develop a platform for simultaneous and direct observations of morphological and electrophysiological changes in neural networks. We used spontaneously active networks grown on microelectrode arrays (MEAs) to allow long-term multisite monitoring with simultaneous optical observations before and after impacts delivered by a ballistic pendulum (30 to 300 g accelerations). The monitoring of AP waveshape templates for long periods before and after impact provided an internal control for cell death or loss of cell-electrode coupling in the observed set of neurons. Network activity patterns were linked in real-time to high power phase contrast microscopy. There was no overt loss of glial or neuronal adhesion, even at high-g impacts. All recording experiments showed repeatable spike production responses: a loss of activity with recovery to near reference in 1 hr, followed by a slow activity decay to a stable, level plateau approximately 30–40% below reference. The initial recovery occurred in two steps: a rapid return of activity to an average 24% below reference, forming a level plateau lasting from 5 to 20 min, followed by a climb to within 10% of reference where a second plateau was established for 1 to 2 hrs. Cross correlation profiles revealed changes in firing hierarchy as well as in Phase 1 in spontaneous network oscillations that were reduced by as much as 20% 6–8 min post impact with only a partial recovery at 30 min. We also observed that normally stable nuclei developed irregular rotational motion after impact in 27 out of 30 networks. The evolution of network activity deficits and recovery can be linked with microscopically observable changes in the very cells that are generating the activity. The repeatable electrophysiological impact response profiles and oscillation changes can provide a quantitative basis for systematic evaluations of pharmacological intervention strategies. Future expansion to include fluorescent microscopy should allow detailed investigations of damage mechanisms on the subcellular level.
Collapse
|
15
|
Adachi R, Yamada R, Kuba H. Tonotopic Differentiation of Coupling between Ca 2+ and Kv1.1 Expression in Brainstem Auditory Circuit. iScience 2019; 13:199-213. [PMID: 30856389 PMCID: PMC6411580 DOI: 10.1016/j.isci.2019.02.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 12/17/2018] [Accepted: 02/22/2019] [Indexed: 12/25/2022] Open
Abstract
Tonotopic differentiations of ion channels ensure sound processing across frequencies. Afferent input plays a critical role in differentiations. We demonstrate here in organotypic culture of chicken cochlear nucleus that expression of Kv1.1 was coupled with Ca2+ to a different degree depending on tonotopic regions, thereby differentiating the level of expression within the nucleus. In the culture, Kv1.1 was down-regulated and not differentiated tonotopically. Chronic depolarization increased Kv1.1 expression in a level-dependent manner. Moreover, the dependence was steeper at higher-frequency regions, which restored the differentiation. The depolarization increased Kv1.1 via activation of Cav1 channels, whereas basal Ca2+ level elevated similarly irrespective of tonotopic regions. Thus, the efficiency of Ca2+-dependent Kv1.1 expression would be fine-tuned in a tonotopic-region-specific manner, emphasizing the importance of neuronal tonotopic identity as well as pattern of afferent input in the tonotopic differentiation of the channel in the auditory circuit. Kv1.1 expression is down-regulated in slice culture of chicken cochlear nucleus Depolarization up-regulates Kv1.1 in a tonotopic-region-specific manner Level of Kv1.1 expression is dependent on basal calcium concentration Efficiency of calcium-dependent Kv1.1 expression is differentiated tonotopically
Collapse
Affiliation(s)
- Ryota Adachi
- Department of Cell Physiology, Graduate School of Medicine, Nagoya University, Nagoya 466-8550, Japan
| | - Rei Yamada
- Department of Cell Physiology, Graduate School of Medicine, Nagoya University, Nagoya 466-8550, Japan
| | - Hiroshi Kuba
- Department of Cell Physiology, Graduate School of Medicine, Nagoya University, Nagoya 466-8550, Japan.
| |
Collapse
|
16
|
Atanasoska M, Vazharova R, Ivanov I, Balabanski L, Andonova S, Ivanov S, Pacheva I, Malinov M, Toncheva D. SCN8A p.Arg1872Gln mutation in early infantile epileptic encephalopathy type 13: Review and case report. BIOTECHNOL BIOTEC EQ 2018. [DOI: 10.1080/13102818.2018.1532815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Affiliation(s)
- Maya Atanasoska
- Genetic Laboratory, Gynecology and Assisted Reproduction Hospital “Dr Malinov D.M.”, Sofia, Bulgaria
| | - Radoslava Vazharova
- Genetic Laboratory, Gynecology and Assisted Reproduction Hospital “Dr Malinov D.M.”, Sofia, Bulgaria
- Department of Biology, Medical Genetics and Microbiology, Faculty of Medicine, Sofia University “St. Kliment Ohridski”, Sofia, Bulgaria
| | - Ivan Ivanov
- Department of Paediatrics and Medical Genetics, St. George University Hospital, Medical University - Plovdiv, Plovdiv, Bulgaria
| | - Lubomir Balabanski
- Genetic Laboratory, Gynecology and Assisted Reproduction Hospital “Dr Malinov D.M.”, Sofia, Bulgaria
| | - Silvia Andonova
- National Genetic Laboratory, University Hospital of Obstetrics and Gynecology “Maichin dom”, Medical University of Sofia, Sofia, Bulgaria
| | - Samuil Ivanov
- Genetic Laboratory, Gynecology and Assisted Reproduction Hospital “Dr Malinov D.M.”, Sofia, Bulgaria
| | - Iliana Pacheva
- Department of Paediatrics and Medical Genetics, St. George University Hospital, Medical University - Plovdiv, Plovdiv, Bulgaria
| | - Maxim Malinov
- Genetic Laboratory, Gynecology and Assisted Reproduction Hospital “Dr Malinov D.M.”, Sofia, Bulgaria
| | - Draga Toncheva
- Genetic Laboratory, Gynecology and Assisted Reproduction Hospital “Dr Malinov D.M.”, Sofia, Bulgaria
- Department of Medical Genetics, Faculty of Medicine, Medical University of Sofia, Sofia, Bulgaria
| |
Collapse
|
17
|
Hines RM, Maric HM, Hines DJ, Modgil A, Panzanelli P, Nakamura Y, Nathanson AJ, Cross A, Deeb T, Brandon NJ, Davies P, Fritschy JM, Schindelin H, Moss SJ. Developmental seizures and mortality result from reducing GABA A receptor α2-subunit interaction with collybistin. Nat Commun 2018; 9:3130. [PMID: 30087324 PMCID: PMC6081406 DOI: 10.1038/s41467-018-05481-1] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 07/05/2018] [Indexed: 01/08/2023] Open
Abstract
Fast inhibitory synaptic transmission is mediated by γ-aminobutyric acid type A receptors (GABAARs) that are enriched at functionally diverse synapses via mechanisms that remain unclear. Using isothermal titration calorimetry and complementary methods we demonstrate an exclusive low micromolar binding of collybistin to the α2-subunit of GABAARs. To explore the biological relevance of collybistin-α2-subunit selectivity, we generate mice with a mutation in the α2-subunit-collybistin binding region (Gabra2-1). The mutation results in loss of a distinct subset of inhibitory synapses and decreased amplitude of inhibitory synaptic currents. Gabra2-1 mice have a striking phenotype characterized by increased susceptibility to seizures and early mortality. Surviving Gabra2-1 mice show anxiety and elevations in electroencephalogram δ power, which are ameliorated by treatment with the α2/α3-selective positive modulator, AZD7325. Taken together, our results demonstrate an α2-subunit selective binding of collybistin, which plays a key role in patterned brain activity, particularly during development.
Collapse
Affiliation(s)
- Rochelle M Hines
- Department of Neuroscience, Tufts University School of Medicine, Boston, 02111, MA, USA.
- Department of Psychology, University of Nevada Las Vegas, Las Vegas, 89154, Ne, USA.
| | - Hans Michael Maric
- Rudolf Virchow Center for Experimental Biomedicine, University of Würzburg, Würzburg, D-97080, Germany
- Department of Biotechnology and Biophysics, Biocenter, University of Würzburg, Würzburg, D-97080, Germany
| | - Dustin J Hines
- Department of Neuroscience, Tufts University School of Medicine, Boston, 02111, MA, USA
- Department of Psychology, University of Nevada Las Vegas, Las Vegas, 89154, Ne, USA
| | - Amit Modgil
- Department of Neuroscience, Tufts University School of Medicine, Boston, 02111, MA, USA
| | - Patrizia Panzanelli
- Department of Neuroscience Rita Levi Montalcini, University of Turin, Turin, 10126, Italy
| | - Yasuko Nakamura
- Department of Neuroscience, Tufts University School of Medicine, Boston, 02111, MA, USA
| | - Anna J Nathanson
- Department of Neuroscience, Tufts University School of Medicine, Boston, 02111, MA, USA
| | - Alan Cross
- AstraZeneca Neuroscience iMED, Biotech Unit, Boston, 02451, MA, USA
| | - Tarek Deeb
- Department of Neuroscience, Tufts University School of Medicine, Boston, 02111, MA, USA
- AstraZeneca Tufts Laboratory for Basic and Translational Neuroscience, Boston, 02111, MA, USA
| | - Nicholas J Brandon
- AstraZeneca Neuroscience iMED, Biotech Unit, Boston, 02451, MA, USA
- AstraZeneca Tufts Laboratory for Basic and Translational Neuroscience, Boston, 02111, MA, USA
| | - Paul Davies
- Department of Neuroscience, Tufts University School of Medicine, Boston, 02111, MA, USA
| | - Jean-Marc Fritschy
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, 8057, Switzerland
- Center for Neuroscience Zurich, University of Zurich and ETH Zurich, Zurich, 8057, Switzerland
| | - Hermann Schindelin
- Rudolf Virchow Center for Experimental Biomedicine, University of Würzburg, Würzburg, D-97080, Germany
| | - Stephen J Moss
- Department of Neuroscience, Tufts University School of Medicine, Boston, 02111, MA, USA.
- AstraZeneca Tufts Laboratory for Basic and Translational Neuroscience, Boston, 02111, MA, USA.
- Department of Neuroscience, Physiology and Pharmacology, University College, London, WC1E 6BT, UK.
| |
Collapse
|
18
|
Two Distinct Secretory Pathways for Differential Kv2.1 Localization in Neurons. J Neurosci 2018; 38:4261-4263. [PMID: 29720558 DOI: 10.1523/jneurosci.0236-18.2018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 03/24/2018] [Accepted: 03/29/2018] [Indexed: 11/21/2022] Open
|
19
|
mTOR-dependent alterations of Kv1.1 subunit expression in the neuronal subset-specific Pten knockout mouse model of cortical dysplasia with epilepsy. Sci Rep 2018; 8:3568. [PMID: 29476105 PMCID: PMC5824782 DOI: 10.1038/s41598-018-21656-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 02/08/2018] [Indexed: 01/03/2023] Open
Abstract
Cortical dysplasia (CD) is a common cause for intractable epilepsy. Hyperactivation of the mechanistic target of rapamycin (mTOR) pathway has been implicated in CD; however, the mechanisms by which mTOR hyperactivation contribute to the epilepsy phenotype remain elusive. Here, we investigated whether constitutive mTOR hyperactivation in the hippocampus is associated with altered voltage-gated ion channel expression in the neuronal subset-specific Pten knockout (NS-Pten KO) mouse model of CD with epilepsy. We found that the protein levels of Kv1.1, but not Kv1.2, Kv1.4, or Kvβ2, potassium channel subunits were increased, along with altered Kv1.1 distribution, within the hippocampus of NS-Pten KO mice. The aberrant Kv1.1 protein levels were present in young adult (≥postnatal week 6) but not juvenile (≤postnatal week 4) NS-Pten KO mice. No changes in hippocampal Kv1.1 mRNA levels were found between NS-Pten KO and WT mice. Interestingly, mTOR inhibition with rapamycin treatment at early and late stages of the pathology normalized Kv1.1 protein levels in NS-Pten KO mice to WT levels. Together, these studies demonstrate altered Kv1.1 protein expression in association with mTOR hyperactivation in NS-Pten KO mice and suggest a role for mTOR signaling in the modulation of voltage-gated ion channel expression in this model.
Collapse
|
20
|
Auditory Input Shapes Tonotopic Differentiation of Kv1.1 Expression in Avian Cochlear Nucleus during Late Development. J Neurosci 2018; 38:2967-2980. [PMID: 29439165 DOI: 10.1523/jneurosci.2472-17.2018] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 01/30/2018] [Accepted: 02/05/2018] [Indexed: 11/21/2022] Open
Abstract
Tonotopic differentiation is fundamental for signal processing in the auditory system. However, when and how this differentiation arises remain elusive. We addressed this issue using electrophysiology and immunohistochemistry in nucleus magnocellularis of chickens of both sexes, which is known to differ in the expression of Kv1.1 channels depending on characteristic frequency (CF). Just after hearing onset (embryonic day 12-14), Kv1 current gradually increased to a slightly larger extent in neurons with higher CF, causing a tonotopic difference of Kv1 current before hatch. However, after hatch, a much larger increase of Kv1 current occurred, particularly in higher-CF neurons, due to an augmentation of Kv1.1 expression at the plasma membrane. This later change in expression led to the large tonotopic difference of Kv1 current characteristic of mature animals. Attenuation of auditory input by inducing conductive or sensorineural hearing loss around hatch suppressed the differentiation in a level-dependent manner. Moreover, elevation of auditory input during embryonic periods could not reproduce the differentiation, suggesting that the capacity of neurons to drive Kv1.1 expression via auditory input develops in a cell-specific manner, thus underlying the frequency-specific expression of the channel within the nucleus. The results indicated that the tonotopic differentiation of Kv1.1 in nucleus magnocellularis is partially determined before hatch, but largely driven by afferent input after hatch. Our results highlight the importance of neuronal capacity for sound to drive ion channel expression as well as the level of auditory experience in the frequency tuning of brainstem auditory circuits.SIGNIFICANCE STATEMENT Tuning-frequency-specific expression of ion channels is a prerequisite for auditory system function, but its underlying mechanisms remain unclear. Here, we revealed in avian cochlear nucleus that the expression of Kv1.1 became more dependent on auditory input at a late period of maturation in neurons tuned to higher-frequency sound, leading to frequency-specific Kv1.1 expression. Attenuation of auditory input during this period suppressed the differentiation in a level-dependent manner, whereas elevation of input in earlier periods could not reproduce the differentiation. Thus, the capacity of neurons to drive Kv1.1 expression via auditory input develops in a cell-specific manner and directs differentiation, highlighting the importance of neuronal character as well as the level of input in the frequency tuning of auditory circuits.
Collapse
|
21
|
Hivert B, Marien L, Agbam KN, Faivre-Sarrailh C. ADAM22 and ADAM23 modulate the targeting of the Kv1 channel-associated protein LGI1 to the axon initial segment. J Cell Sci 2018; 132:jcs.219774. [DOI: 10.1242/jcs.219774] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 12/19/2018] [Indexed: 01/30/2023] Open
Abstract
The distribution of voltage-gated potassium channels Kv1 at the axon initial segment (AIS) influences neuronal intrinsic excitability. Kv1.1/1.2 subunits are associated with cell adhesion molecules (CAMs), including Caspr2 and LGI1 that are implicated in autoimmune and genetic neurological diseases with seizures. In particular, mutations in the LGI1 gene cause autosomal dominant lateral temporal lobe epilepsy (ADLTE). Here, using rat hippocampal neurons in culture, we showed that LGI1 is recruited at the AIS and colocalized with ADAM22 and Kv1 channels. Strikingly, the missense mutations S473L and R474Q of LGI1 identified in ADLTE prevent its association with ADAM22 and enrichment at the AIS. Moreover, we observed that ADAM22 or ADAM23 modulates the trafficking of LGI1, and promotes its ER export and expression at the overall neuronal cell surface. Live-cell imaging indicated that LGI1 is co-transported in axonal vesicles with ADAM22 or ADAM23. Finally, we showed that ADAM22 and ADAM23 also associate with Caspr2 and TAG-1 to be selectively targeted within different axonal sub-regions. So, the combinatorial expression of Kv1-associated CAMs may be critical to tune intrinsic excitability in physiological or epileptogenic context.
Collapse
Affiliation(s)
- Bruno Hivert
- Aix Marseille Université, INSERM UMR1249, F-13273 Marseille, France
- Present address: Aix Marseille Université, CNRS UMR7289, Institut de Neurosciences de la Timone, F-13385 Marseille, France
| | - Laurène Marien
- Aix Marseille Université, INSERM UMR1249, F-13273 Marseille, France
| | | | | |
Collapse
|
22
|
Duménieu M, Oulé M, Kreutz MR, Lopez-Rojas J. The Segregated Expression of Voltage-Gated Potassium and Sodium Channels in Neuronal Membranes: Functional Implications and Regulatory Mechanisms. Front Cell Neurosci 2017; 11:115. [PMID: 28484374 PMCID: PMC5403416 DOI: 10.3389/fncel.2017.00115] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 04/05/2017] [Indexed: 01/25/2023] Open
Abstract
Neurons are highly polarized cells with apparent functional and morphological differences between dendrites and axon. A critical determinant for the molecular and functional identity of axonal and dendritic segments is the restricted expression of voltage-gated ion channels (VGCs). Several studies show an uneven distribution of ion channels and their differential regulation within dendrites and axons, which is a prerequisite for an appropriate integration of synaptic inputs and the generation of adequate action potential (AP) firing patterns. This review article will focus on the signaling pathways leading to segmented expression of voltage-gated potassium and sodium ion channels at the neuronal plasma membrane and the regulatory mechanisms ensuring segregated functions. We will also discuss the relevance of proper ion channel targeting for neuronal physiology and how alterations in polarized distribution contribute to neuronal pathology.
Collapse
Affiliation(s)
- Maël Duménieu
- Research Group Neuroplasticity, Leibniz Institute for NeurobiologyMagdeburg, Germany
| | - Marie Oulé
- Research Group Neuroplasticity, Leibniz Institute for NeurobiologyMagdeburg, Germany
| | - Michael R Kreutz
- Research Group Neuroplasticity, Leibniz Institute for NeurobiologyMagdeburg, Germany.,Leibniz Group "Dendritic Organelles and Synaptic Function", University Medical Center Hamburg-Eppendorf, Center for Molecular Neurobiology (ZMNH)Hamburg, Germany
| | - Jeffrey Lopez-Rojas
- Research Group Neuroplasticity, Leibniz Institute for NeurobiologyMagdeburg, Germany
| |
Collapse
|
23
|
Goetz SM, Deng ZD. The development and modelling of devices and paradigms for transcranial magnetic stimulation. Int Rev Psychiatry 2017; 29:115-145. [PMID: 28443696 PMCID: PMC5484089 DOI: 10.1080/09540261.2017.1305949] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 03/03/2017] [Accepted: 03/09/2017] [Indexed: 12/20/2022]
Abstract
Magnetic stimulation is a non-invasive neurostimulation technique that can evoke action potentials and modulate neural circuits through induced electric fields. Biophysical models of magnetic stimulation have become a major driver for technological developments and the understanding of the mechanisms of magnetic neurostimulation and neuromodulation. Major technological developments involve stimulation coils with different spatial characteristics and pulse sources to control the pulse waveform. While early technological developments were the result of manual design and invention processes, there is a trend in both stimulation coil and pulse source design to mathematically optimize parameters with the help of computational models. To date, macroscopically highly realistic spatial models of the brain, as well as peripheral targets, and user-friendly software packages enable researchers and practitioners to simulate the treatment-specific and induced electric field distribution in the brains of individual subjects and patients. Neuron models further introduce the microscopic level of neural activation to understand the influence of activation dynamics in response to different pulse shapes. A number of models that were designed for online calibration to extract otherwise covert information and biomarkers from the neural system recently form a third branch of modelling.
Collapse
Affiliation(s)
- Stefan M Goetz
- a Department of Psychiatry & Behavioral Sciences, Division for Brain Stimulation & Neurophysiology , Duke University , Durham , NC , USA
- b Department of Electrical & Computer Engineering , Duke University , Durham , NC , USA
- c Department of Neurosurgery , Duke University , Durham , NC , USA
| | - Zhi-De Deng
- a Department of Psychiatry & Behavioral Sciences, Division for Brain Stimulation & Neurophysiology , Duke University , Durham , NC , USA
- d Intramural Research Program, Experimental Therapeutics & Pathophysiology Branch, Noninvasive Neuromodulation Unit , National Institutes of Health, National Institute of Mental Health , Bethesda , MD , USA
- e Duke Institute for Brain Sciences , Duke University , Durham , NC , USA
| |
Collapse
|
24
|
Cornejo VH, Luarte A, Couve A. Global and local mechanisms sustain axonal proteostasis of transmembrane proteins. Traffic 2017; 18:255-266. [PMID: 28220989 DOI: 10.1111/tra.12472] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Revised: 02/02/2017] [Accepted: 02/16/2017] [Indexed: 12/22/2022]
Abstract
The control of neuronal protein homeostasis or proteostasis is tightly regulated both spatially and temporally, assuring accurate and integrated responses to external or intrinsic stimuli. Local or autonomous responses in dendritic and axonal compartments are crucial to sustain function during development, physiology and in response to damage or disease. Axons are responsible for generating and propagating electrical impulses in neurons, and the establishment and maintenance of their molecular composition are subject to extreme constraints exerted by length and size. Proteins that require the secretory pathway, such as receptors, transporters, ion channels or cell adhesion molecules, are fundamental for axonal function, but whether axons regulate their abundance autonomously and how they achieve this is not clear. Evidence supports the role of three complementary mechanisms to maintain proteostasis of these axonal proteins, namely vesicular transport, local translation and trafficking and transfer from supporting cells. Here, we review these mechanisms, their molecular machineries and contribution to neuronal function. We also examine the signaling pathways involved in local translation and their role during development and nerve injury. We discuss the relative contributions of a transport-controlled proteome directed by the soma (global regulation) versus a local-controlled proteome based on local translation or cell transfer (local regulation).
Collapse
Affiliation(s)
- Víctor Hugo Cornejo
- Program of Physiology and Biophysics, ICBM, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Biomedical Neuroscience Institute, Universidad de Chile, Santiago, Chile
| | - Alejandro Luarte
- Program of Physiology and Biophysics, ICBM, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Biomedical Neuroscience Institute, Universidad de Chile, Santiago, Chile
| | - Andrés Couve
- Program of Physiology and Biophysics, ICBM, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Biomedical Neuroscience Institute, Universidad de Chile, Santiago, Chile
| |
Collapse
|
25
|
Heteromeric complexes of aldo-keto reductase auxiliary K Vβ subunits (AKR6A) regulate sarcolemmal localization of K V1.5 in coronary arterial myocytes. Chem Biol Interact 2017; 276:210-217. [PMID: 28342889 DOI: 10.1016/j.cbi.2017.03.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Revised: 03/02/2017] [Accepted: 03/21/2017] [Indexed: 01/20/2023]
Abstract
Redox-sensitive potassium channels consisting of the voltage-gated K+ (KV) channel pore subunit KV1.5 regulate resting membrane potential and thereby contractility of vascular smooth muscle cells. Members of the KV1 family associate with cytosolic auxiliary β subunits, which are members of the aldo-keto reductase (AKR) superfamily (AKR6A subfamily). The Kvβ subunits have been proposed to regulate Kv1 gating via pyridine nucleotide cofactor binding. However, the molecular identity of KVβ subunits that associate with native KV1.5 channels in the vasculature is unknown. Here, we examined mRNA and protein expression of KVβ subunits and tested whether KVβ isoforms interact with KV1.5 channels in murine coronary arteries. We detected KVβ1 (AKR6A3), KVβ2 (AKR6A5) and KVβ3 (AKR6A9) transcripts and KVβ1 and KVβ2 protein in left anterior descending coronary arteries by real time quantitative PCR and Western blot, respectively. In situ proximity ligation assays indicated abundant protein-protein interactions between KV1.5/KVβ1, KV1.5/KVβ2 and KVβ1/β2 in coronary arterial myocytes. Confocal microscopy and membrane fractionation analyses suggest that arterial myocytes from KVβ2-null mice have reduced abundance of sarcolemmal KV1.5. Together, data suggest that in coronary arterial myocytes, KV1.5 channels predominantly associate with KVβ1 and KVβ2 proteins and that KVβ2 performs a chaperone function for KV1.5 channels in arterial myocytes, thereby facilitating Kv1α trafficking and membrane localization.
Collapse
|
26
|
Kyung JW, Cho IH, Lee S, Song WK, Ryan TA, Hoppa MB, Kim SH. Adaptor Protein 2 (AP-2) complex is essential for functional axogenesis in hippocampal neurons. Sci Rep 2017; 7:41620. [PMID: 28139716 PMCID: PMC5282494 DOI: 10.1038/srep41620] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 12/21/2016] [Indexed: 11/18/2022] Open
Abstract
The complexity and diversity of a neural network requires regulated elongation and branching of axons, as well as the formation of synapses between neurons. In the present study we explore the role of AP-2, a key endocytic adaptor protein complex, in the development of rat hippocampal neurons. We found that the loss of AP-2 during the early stage of development resulted in impaired axon extension and failed maturation of the axon initial segment (AIS). Normally the AIS performs two tasks in concert, stabilizing neural polarity and generating action potentials. In AP-2 silenced axons polarity is established, however there is a failure to establish action potential firing. Consequently, this impairs activity-driven Ca2+ influx and exocytosis at nerve terminals. In contrast, removal of AP-2 from older neurons does not impair axonal growth or signaling and synaptic function. Our data reveal that AP-2 has important roles in functional axogenesis by proper extension of axon as well as the formation of AIS during the early step of neurodevelopment.
Collapse
Affiliation(s)
- Jae Won Kyung
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, 02447, South Korea
| | - In Ha Cho
- Department of Biology, Molecular Cellular Biology Program, Dartmouth College, Hanover, NH, 03755, USA
| | - Sukmook Lee
- Laboratory of Molecular Cancer Therapeutics, Scripps Korea Antibody Institute, Chuncheon, 24341, South Korea
| | - Woo Keun Song
- School of Life Science, Bioimaging Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, South Korea
| | - Timothy A Ryan
- Department of Biochemistry, Weill Cornell Medical College, New York, NY, 10065, USA
| | - Michael B Hoppa
- Department of Biology, Molecular Cellular Biology Program, Dartmouth College, Hanover, NH, 03755, USA
| | - Sung Hyun Kim
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, 02447, South Korea.,Department of Physiology, Neurodegeneration Control Research Center, School of Medicine, Kyung Hee University, Seoul, 02447, South Korea
| |
Collapse
|
27
|
Valkova C, Liebmann L, Krämer A, Hübner CA, Kaether C. The sorting receptor Rer1 controls Purkinje cell function via voltage gated sodium channels. Sci Rep 2017; 7:41248. [PMID: 28117367 PMCID: PMC5259745 DOI: 10.1038/srep41248] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 12/19/2016] [Indexed: 01/08/2023] Open
Abstract
Rer1 is a sorting receptor in the early secretory pathway that controls the assembly and the cell surface transport of selected multimeric membrane protein complexes. Mice with a Purkinje cell (PC) specific deletion of Rer1 showed normal polarization and differentiation of PCs and normal development of the cerebellum. However, PC-specific loss of Rer1 led to age-dependent motor deficits in beam walk, ladder climbing and gait. Analysis of brain sections revealed a specific degeneration of PCs in the anterior cerebellar lobe in old animals. Electrophysiological recordings demonstrated severe deficits in spontaneous action potential generation. Measurements of resurgent currents indicated decreased surface densities of voltage-gated sodium channels (Nav), but not changes in individual channels. Analysis of mice with a whole brain Rer1-deletion demonstrated a strong down-regulation of Nav1.6 and 1.1 in the absence of Rer1, whereas protein levels of the related Cav2.1 and of Kv3.3 and 7.2 channels were not affected. The data suggest that Rer1 controls the assembly and transport of Nav1.1 and 1.6, the principal sodium channels responsible for recurrent firing, in PCs.
Collapse
Affiliation(s)
- Christina Valkova
- Leibniz Institut für Alternsforschung-Fritz Lipmann Institut, 07743 Jena, Germany
| | - Lutz Liebmann
- Institut für Humangenetik, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Germany
| | - Andreas Krämer
- Leibniz Institut für Alternsforschung-Fritz Lipmann Institut, 07743 Jena, Germany
| | - Christian A Hübner
- Institut für Humangenetik, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Germany
| | - Christoph Kaether
- Leibniz Institut für Alternsforschung-Fritz Lipmann Institut, 07743 Jena, Germany
| |
Collapse
|
28
|
Abstract
Voltage-gated sodium channels (VGSC) are critical determinants of cellular electrical activity through the control of initiation and propagation of action potential. To ensure this role, these proteins are not consistently delivered to the plasma membrane but undergo drastic quality controls throughout various adaptive processes such as biosynthesis, anterograde and retrograde trafficking, and membrane targeting. In pathological conditions, this quality control could lead to the retention of functional VGSC and is therefore the target of different pharmacological approaches. The present chapter gives an overview of the current understanding of the facets of VGSC life cycle in the context of both cardiac and neuronal cell types.
Collapse
Affiliation(s)
- A Mercier
- Laboratoire de Signalisation et Transports Ioniques Membranaires, Pôle Biologie Santé, Université de Poitiers, CNRS, 1 rue Georges Bonnet, TSA 51106, 86073, Poitiers Cedex 9, France
| | - P Bois
- Laboratoire de Signalisation et Transports Ioniques Membranaires, Pôle Biologie Santé, Université de Poitiers, CNRS, 1 rue Georges Bonnet, TSA 51106, 86073, Poitiers Cedex 9, France
| | - A Chatelier
- Laboratoire de Signalisation et Transports Ioniques Membranaires, Pôle Biologie Santé, Université de Poitiers, CNRS, 1 rue Georges Bonnet, TSA 51106, 86073, Poitiers Cedex 9, France.
| |
Collapse
|
29
|
Abstract
KCNQ2/3 (Kv7.2/7.3) channels and voltage-gated sodium channels (VGSCs) are enriched in the axon initial segment (AIS) where they bind to ankyrin-G and coregulate membrane potential in central nervous system neurons. The molecular mechanisms supporting coordinated regulation of KCNQ and VGSCs and the cellular mechanisms governing KCNQ trafficking to the AIS are incompletely understood. Here, we show that fibroblast growth factor 14 (FGF14), previously described as a VGSC regulator, also affects KCNQ function and localization. FGF14 knockdown leads to a reduction of KCNQ2 in the AIS and a reduction in whole-cell KCNQ currents. FGF14 positively regulates KCNQ2/3 channels in a simplified expression system. FGF14 interacts with KCNQ2 at a site distinct from the FGF14-VGSC interaction surface, thus enabling the bridging of NaV1.6 and KCNQ2. These data implicate FGF14 as an organizer of channel localization in the AIS and provide insight into the coordination of KCNQ and VGSC conductances in the regulation of membrane potential.
Collapse
|
30
|
Soo Hoo L, Banna CD, Radeke CM, Sharma N, Albertolle ME, Low SH, Weimbs T, Vandenberg CA. The SNARE Protein Syntaxin 3 Confers Specificity for Polarized Axonal Trafficking in Neurons. PLoS One 2016; 11:e0163671. [PMID: 27662481 PMCID: PMC5035089 DOI: 10.1371/journal.pone.0163671] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 09/12/2016] [Indexed: 11/19/2022] Open
Abstract
Cell polarity and precise subcellular protein localization are pivotal to neuronal function. The SNARE machinery underlies intracellular membrane fusion events, but its role in neuronal polarity and selective protein targeting remain unclear. Here we report that syntaxin 3 is involved in orchestrating polarized trafficking in cultured rat hippocampal neurons. We show that syntaxin 3 localizes to the axonal plasma membrane, particularly to axonal tips, whereas syntaxin 4 localizes to the somatodendritic plasma membrane. Disruption of a conserved N-terminal targeting motif, which causes mislocalization of syntaxin 3, results in coincident mistargeting of the axonal cargos neuron-glia cell adhesion molecule (NgCAM) and neurexin, but not transferrin receptor, a somatodendritic cargo. Similarly, RNAi-mediated knockdown of endogenous syntaxin 3 leads to partial mistargeting of NgCAM, demonstrating that syntaxin 3 plays an important role in its targeting. Additionally, overexpression of syntaxin 3 results in increased axonal growth. Our findings suggest an important role for syntaxin 3 in maintaining neuronal polarity and in the critical task of selective trafficking of membrane protein to axons.
Collapse
Affiliation(s)
- Linda Soo Hoo
- Department of Molecular, Cellular, and Developmental Biology, University of California Santa Barbara, Santa Barbara, California, United States of America
- Neuroscience Research Institute, University of California Santa Barbara, Santa Barbara, California, United States of America
| | - Chris D. Banna
- Department of Molecular, Cellular, and Developmental Biology, University of California Santa Barbara, Santa Barbara, California, United States of America
- Neuroscience Research Institute, University of California Santa Barbara, Santa Barbara, California, United States of America
| | - Carolyn M. Radeke
- Department of Molecular, Cellular, and Developmental Biology, University of California Santa Barbara, Santa Barbara, California, United States of America
- Neuroscience Research Institute, University of California Santa Barbara, Santa Barbara, California, United States of America
| | - Nikunj Sharma
- Department of Molecular, Cellular, and Developmental Biology, University of California Santa Barbara, Santa Barbara, California, United States of America
- Neuroscience Research Institute, University of California Santa Barbara, Santa Barbara, California, United States of America
| | - Mary E. Albertolle
- Department of Molecular, Cellular, and Developmental Biology, University of California Santa Barbara, Santa Barbara, California, United States of America
- Neuroscience Research Institute, University of California Santa Barbara, Santa Barbara, California, United States of America
| | - Seng Hui Low
- Department of Molecular, Cellular, and Developmental Biology, University of California Santa Barbara, Santa Barbara, California, United States of America
- Neuroscience Research Institute, University of California Santa Barbara, Santa Barbara, California, United States of America
| | - Thomas Weimbs
- Department of Molecular, Cellular, and Developmental Biology, University of California Santa Barbara, Santa Barbara, California, United States of America
- Neuroscience Research Institute, University of California Santa Barbara, Santa Barbara, California, United States of America
| | - Carol A. Vandenberg
- Department of Molecular, Cellular, and Developmental Biology, University of California Santa Barbara, Santa Barbara, California, United States of America
- Neuroscience Research Institute, University of California Santa Barbara, Santa Barbara, California, United States of America
- * E-mail:
| |
Collapse
|
31
|
Benítez-King G, Valdés-Tovar M, Trueta C, Galván-Arrieta T, Argueta J, Alarcón S, Lora-Castellanos A, Solís-Chagoyán H. The microtubular cytoskeleton of olfactory neurons derived from patients with schizophrenia or with bipolar disorder: Implications for biomarker characterization, neuronal physiology and pharmacological screening. Mol Cell Neurosci 2016; 73:84-95. [PMID: 26837043 DOI: 10.1016/j.mcn.2016.01.013] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Revised: 01/26/2016] [Accepted: 01/29/2016] [Indexed: 01/29/2023] Open
Abstract
Schizophrenia (SZ) and Bipolar Disorder (BD) are highly inheritable chronic mental disorders with a worldwide prevalence of around 1%. Despite that many efforts had been made to characterize biomarkers in order to allow for biological testing for their diagnoses, these disorders are currently detected and classified only by clinical appraisal based on the Diagnostic and Statistical Manual of Mental Disorders. Olfactory neuroepithelium-derived neuronal precursors have been recently proposed as a model for biomarker characterization. Because of their peripheral localization, they are amenable to collection and suitable for being cultured and propagated in vitro. Olfactory neuroepithelial cells can be obtained by a non-invasive brush-exfoliation technique from neuropsychiatric patients and healthy subjects. Neuronal precursors isolated from these samples undergo in vitro the cytoskeletal reorganization inherent to the neurodevelopment process which has been described as one important feature in the etiology of both diseases. In this paper, we will review the current knowledge on microtubular organization in olfactory neurons of patients with SZ and with BD that may constitute specific cytoskeletal endophenotypes and their relation with alterations in L-type voltage-activated Ca(2+) currents. Finally, the potential usefulness of neuronal precursors for pharmacological screening will be discussed.
Collapse
Affiliation(s)
- G Benítez-King
- Laboratorio de Neurofarmacología, Subdirección de Investigaciones Clínicas, Mexico.
| | - M Valdés-Tovar
- Laboratorio de Neurofarmacología, Subdirección de Investigaciones Clínicas, Mexico
| | - C Trueta
- Departamento de Neurofisiología, Dirección de Investigaciones en Neurociencias, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz (INPRFM), Calzada México-Xochimilco No. 101, Col. San Lorenzo-Huipulco, C.P. 14370, Tlalpan, Distrito Federal, Mexico
| | - T Galván-Arrieta
- Laboratorio de Neurofarmacología, Subdirección de Investigaciones Clínicas, Mexico
| | - J Argueta
- Laboratorio de Neurofarmacología, Subdirección de Investigaciones Clínicas, Mexico
| | - S Alarcón
- Laboratorio de Neurofarmacología, Subdirección de Investigaciones Clínicas, Mexico
| | - A Lora-Castellanos
- Laboratorio de Neurofarmacología, Subdirección de Investigaciones Clínicas, Mexico
| | - H Solís-Chagoyán
- Laboratorio de Neurofarmacología, Subdirección de Investigaciones Clínicas, Mexico
| |
Collapse
|
32
|
Tétreault MP, Bourdin B, Briot J, Segura E, Lesage S, Fiset C, Parent L. Identification of Glycosylation Sites Essential for Surface Expression of the CaVα2δ1 Subunit and Modulation of the Cardiac CaV1.2 Channel Activity. J Biol Chem 2016; 291:4826-43. [PMID: 26742847 DOI: 10.1074/jbc.m115.692178] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2015] [Indexed: 12/15/2022] Open
Abstract
Alteration in the L-type current density is one aspect of the electrical remodeling observed in patients suffering from cardiac arrhythmias. Changes in channel function could result from variations in the protein biogenesis, stability, post-translational modification, and/or trafficking in any of the regulatory subunits forming cardiac L-type Ca(2+) channel complexes. CaVα2δ1 is potentially the most heavily N-glycosylated subunit in the cardiac L-type CaV1.2 channel complex. Here, we show that enzymatic removal of N-glycans produced a 50-kDa shift in the mobility of cardiac and recombinant CaVα2δ1 proteins. This change was also observed upon simultaneous mutation of the 16 Asn sites. Nonetheless, the mutation of only 6/16 sites was sufficient to significantly 1) reduce the steady-state cell surface fluorescence of CaVα2δ1 as characterized by two-color flow cytometry assays and confocal imaging; 2) decrease protein stability estimated from cycloheximide chase assays; and 3) prevent the CaVα2δ1-mediated increase in the peak current density and voltage-dependent gating of CaV1.2. Reversing the N348Q and N812Q mutations in the non-operational sextuplet Asn mutant protein partially restored CaVα2δ1 function. Single mutation N663Q and double mutations N348Q/N468Q, N348Q/N812Q, and N468Q/N812Q decreased protein stability/synthesis and nearly abolished steady-state cell surface density of CaVα2δ1 as well as the CaVα2δ1-induced up-regulation of L-type currents. These results demonstrate that Asn-663 and to a lesser extent Asn-348, Asn-468, and Asn-812 contribute to protein stability/synthesis of CaVα2δ1, and furthermore that N-glycosylation of CaVα2δ1 is essential to produce functional L-type Ca(2+) channels.
Collapse
Affiliation(s)
| | - Benoîte Bourdin
- From the Départment de Physiologie Moléculaire et Intégrative, Faculté de Médecine, and
| | - Julie Briot
- From the Départment de Physiologie Moléculaire et Intégrative, Faculté de Médecine, and
| | - Emilie Segura
- From the Départment de Physiologie Moléculaire et Intégrative, Faculté de Médecine, and
| | - Sylvie Lesage
- Départment de Microbiologie, Infectiologie, and Immunologie, Faculté de Médecine, Centre de Recherche de l'Hôpital Maisonneuve-Rosemont, Université de Montréal, Montréal, Québec H3C 3J7, Canada
| | - Céline Fiset
- Faculté de Pharmacie, Institut de Cardiologie de Montréal and
| | - Lucie Parent
- From the Départment de Physiologie Moléculaire et Intégrative, Faculté de Médecine, and
| |
Collapse
|
33
|
Szklarczyk R, Megchelenbrink W, Cizek P, Ledent M, Velemans G, Szklarczyk D, Huynen MA. WeGET: predicting new genes for molecular systems by weighted co-expression. Nucleic Acids Res 2015; 44:D567-73. [PMID: 26582928 PMCID: PMC4702868 DOI: 10.1093/nar/gkv1228] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Accepted: 10/30/2015] [Indexed: 01/04/2023] Open
Abstract
We have developed the Weighted Gene Expression Tool and database (WeGET, http://weget.cmbi.umcn.nl) for the prediction of new genes of a molecular system by correlated gene expression. WeGET utilizes a compendium of 465 human and 560 murine gene expression datasets that have been collected from multiple tissues under a wide range of experimental conditions. It exploits this abundance of expression data by assigning a high weight to datasets in which the known genes of a molecular system are harmoniously up- and down-regulated. WeGET ranks new candidate genes by calculating their weighted co-expression with that system. A weighted rank is calculated for human genes and their mouse orthologs. Then, an integrated gene rank and p-value is computed using a rank-order statistic. We applied our method to predict novel genes that have a high degree of co-expression with Gene Ontology terms and pathways from KEGG and Reactome. For each query set we provide a list of predicted novel genes, computed weights for transcription datasets used and cell and tissue types that contributed to the final predictions. The performance for each query set is assessed by 10-fold cross-validation. Finally, users can use the WeGET to predict novel genes that co-express with a custom query set.
Collapse
Affiliation(s)
- Radek Szklarczyk
- Maastricht University Medical Centre, P. Debyelaan 25, 6226XM Maastricht, The Netherlands Centre for Molecular and Biomolecular Informatics (CMBI), Radboud University Medical Centre, Geert Grooteplein Zuid 26-28, 6525 GA Nijmegen, Nijmegen, The Netherlands
| | - Wout Megchelenbrink
- Centre for Molecular and Biomolecular Informatics (CMBI), Radboud University Medical Centre, Geert Grooteplein Zuid 26-28, 6525 GA Nijmegen, Nijmegen, The Netherlands Institute for Computing and Information Sciences (ICIS), Radboud University, Toernooiveld 212, 6525 EC Nijmegen, The Netherlands Centre for Systems Biology and Bioenergetics (CSBB), Radboud University Medical Centre, Geert Grooteplein Zuid 10, 6500 HB Nijmegen, Nijmegen, The Netherlands
| | - Pavel Cizek
- Centre for Molecular and Biomolecular Informatics (CMBI), Radboud University Medical Centre, Geert Grooteplein Zuid 26-28, 6525 GA Nijmegen, Nijmegen, The Netherlands
| | - Marie Ledent
- Maastricht University Medical Centre, P. Debyelaan 25, 6226XM Maastricht, The Netherlands
| | - Gonny Velemans
- Centre for Molecular and Biomolecular Informatics (CMBI), Radboud University Medical Centre, Geert Grooteplein Zuid 26-28, 6525 GA Nijmegen, Nijmegen, The Netherlands
| | - Damian Szklarczyk
- Institute of Molecular Life Sciences and Swiss Institute of Bioinformatics, University of Zurich, 8057 Zurich, Switzerland
| | - Martijn A Huynen
- Centre for Molecular and Biomolecular Informatics (CMBI), Radboud University Medical Centre, Geert Grooteplein Zuid 26-28, 6525 GA Nijmegen, Nijmegen, The Netherlands Centre for Systems Biology and Bioenergetics (CSBB), Radboud University Medical Centre, Geert Grooteplein Zuid 10, 6500 HB Nijmegen, Nijmegen, The Netherlands
| |
Collapse
|
34
|
Spear JM, Koborssy DA, Schwartz AB, Johnson AJ, Audhya A, Fadool DA, Stagg SM. Kv1.3 contains an alternative C-terminal ER exit motif and is recruited into COPII vesicles by Sec24a. BMC BIOCHEMISTRY 2015; 16:16. [PMID: 26156069 PMCID: PMC4497498 DOI: 10.1186/s12858-015-0045-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Accepted: 06/26/2015] [Indexed: 12/11/2022]
Abstract
Background Potassium channels play a fundamental role in resetting the resting membrane potential of excitable cells. Determining the intracellular trafficking and localization mechanisms of potassium channels provides a platform to fully characterize their maturation and functionality. Previous investigations have discovered residues or motifs that exist in their primary structure, which directly promote anterograde trafficking of nascent potassium channels. Recently, a non-conical di-acidic motif (E483/484) has been discovered in the C-terminus of the mammalian homologue of the Shaker voltage-gated potassium channel subfamily member 3 (Kv1.3), and was shown to disrupt the anterograde trafficking of Kv1.3. Results We have further investigated the intracellular trafficking requirements of Kv1.3 both in vivo and in vitro. First, three alternative C-terminal acidic residues, E443, E445, E447 were probed for their involvement within the early secretory pathway of Kv1.3. Single point (E443A, E445A, and E447A) and double point (E443A-E445A, E445A-E447A) mutations exhibited no significant changes in their endoplasmic reticulum (ER) retention. The triple point mutant E443A-E445A-E447A displayed a modest ER retention while deletion of the C-terminus showed dramatic ER retention. Second, we demonstrate in vivo the requirement for the Sec24a isoform to confer anterograde trafficking using a siRNA knockdown assay. Third, we show in vitro the association of recombinantly expressed Kv1.3 and Sec24a proteins. Conclusion These results expand upon previous studies aimed at deciphering the Kv1.3 secretory trafficking mechanisms and further show in vitro evidence of the association between Kv1.3 and the COPII cargo adaptor subunit isoform Sec24a. Electronic supplementary material The online version of this article (doi:10.1186/s12858-015-0045-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- John M Spear
- Institute of Molecular Biophysics, Florida State University, 91 Chieftan Way, Tallahassee, FL, 32306, USA.
| | - Dolly Al Koborssy
- Program in Neuroscience, Florida State University, 1107 West Call Street, Tallahassee, FL, 32306, USA.
| | - Austin B Schwartz
- Institute of Molecular Biophysics, Florida State University, 91 Chieftan Way, Tallahassee, FL, 32306, USA.
| | - Adam J Johnson
- Biomolecular Chemistry, University of Wisconsin-Madison, 440 Henry Mall, Madison, WI, 53706, USA.
| | - Anjon Audhya
- Biomolecular Chemistry, University of Wisconsin-Madison, 440 Henry Mall, Madison, WI, 53706, USA.
| | - Debra A Fadool
- Institute of Molecular Biophysics, Florida State University, 91 Chieftan Way, Tallahassee, FL, 32306, USA. .,Program in Neuroscience, Florida State University, 1107 West Call Street, Tallahassee, FL, 32306, USA. .,Department of Biological Science, Florida State University, 319 Stadium Drive, Tallahassee, FL, 32306, USA.
| | - Scott M Stagg
- Institute of Molecular Biophysics, Florida State University, 91 Chieftan Way, Tallahassee, FL, 32306, USA. .,Department of Chemistry and Biochemistry, Florida State University, 91 Chieftan Way, Tallahassee, FL, 32306, USA.
| |
Collapse
|
35
|
Ahmed Z, Wieraszko A. Pulsed magnetic stimulation modifies amplitude of action potentials in vitro via ionic channels-dependent mechanism. Bioelectromagnetics 2015; 36:386-97. [DOI: 10.1002/bem.21917] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2013] [Accepted: 03/11/2015] [Indexed: 12/18/2022]
Affiliation(s)
- Zaghloul Ahmed
- Department of Physical Therapy; The Program for Developmental Neuroscience; The College of Staten Island, Staten Island, and Graduate Center/The City University of New York; New York NY
| | - Andrzej Wieraszko
- The Department of Biology; The Program for Developmental Neuroscience; The College of Staten Island, Staten Island, and Graduate Center/The City University of New York; New York NY
| |
Collapse
|
36
|
Trimmer JS. Subcellular localization of K+ channels in mammalian brain neurons: remarkable precision in the midst of extraordinary complexity. Neuron 2015; 85:238-56. [PMID: 25611506 DOI: 10.1016/j.neuron.2014.12.042] [Citation(s) in RCA: 155] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Potassium channels (KChs) are the most diverse ion channels, in part due to extensive combinatorial assembly of a large number of principal and auxiliary subunits into an assortment of KCh complexes. Their structural and functional diversity allows KChs to play diverse roles in neuronal function. Localization of KChs within specialized neuronal compartments defines their physiological role and also fundamentally impacts their activity, due to localized exposure to diverse cellular determinants of channel function. Recent studies in mammalian brain reveal an exquisite refinement of KCh subcellular localization. This includes axonal KChs at the initial segment, and near/within nodes of Ranvier and presynaptic terminals, dendritic KChs found at sites reflecting specific synaptic input, and KChs defining novel neuronal compartments. Painting the remarkable diversity of KChs onto the complex architecture of mammalian neurons creates an elegant picture of electrical signal processing underlying the sophisticated function of individual neuronal compartments, and ultimately neurotransmission and behavior.
Collapse
Affiliation(s)
- James S Trimmer
- Department of Neurobiology, Physiology, and Behavior, University of California, Davis, Davis, CA 95616, USA; Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA 95616, USA.
| |
Collapse
|
37
|
King AN, Manning CF, Trimmer JS. A unique ion channel clustering domain on the axon initial segment of mammalian neurons. J Comp Neurol 2015; 522:2594-608. [PMID: 24477962 DOI: 10.1002/cne.23551] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2013] [Revised: 01/21/2014] [Accepted: 01/22/2014] [Indexed: 01/11/2023]
Abstract
The axon initial segment (AIS) plays a key role in initiation of action potentials and neuronal output. The plasma membrane of the AIS contains high densities of voltage-gated ion channels required for these electrical events, and much recent work has focused on defining the mechanisms for generating and maintaining this unique neuronal plasma membrane domain. The Kv2.1 voltage-gated potassium channel is abundantly present in large clusters on the soma and proximal dendrites of mammalian brain neurons. Kv2.1 is also a component of the ion channel repertoire at the AIS. Here we show that Kv2.1 clusters on the AIS of brain neurons across diverse mammalian species including humans define a noncanonical ion channel clustering domain deficient in Ankyrin-G. The sites of Kv2.1 clustering on the AIS are sites where cisternal organelles, specialized intracellular calcium release membranes, come into close apposition with the plasma membrane, and are also sites of clustering of γ-aminobutyric acid (GABA)ergic synapses. Using an antibody specific for a single Kv2.1 phosphorylation site, we find that the phosphorylation state differs between Kv2.1 clusters on the proximal and distal portions of the AIS. Together, these studies show that the sites of Kv2.1 clustering on the AIS represent specialized domains containing components of diverse neuronal signaling pathways that may contribute to local regulation of Kv2.1 function and AIS membrane excitability.
Collapse
Affiliation(s)
- Anna N King
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, California, 95616
| | | | | |
Collapse
|
38
|
Curran J, Mohler PJ. Alternative Paradigms for Ion Channelopathies: Disorders of Ion Channel Membrane Trafficking and Posttranslational Modification. Annu Rev Physiol 2015; 77:505-24. [DOI: 10.1146/annurev-physiol-021014-071838] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Jerry Curran
- The Dorothy M. Davis Heart & Lung Research Institute,
- Department of Physiology and Cell Biology, and
| | - Peter J. Mohler
- The Dorothy M. Davis Heart & Lung Research Institute,
- Department of Physiology and Cell Biology, and
- Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio 43210;
| |
Collapse
|
39
|
Pablo JL, Pitt GS. Fibroblast Growth Factor Homologous Factors: New Roles in Neuronal Health and Disease. Neuroscientist 2014; 22:19-25. [PMID: 25492945 DOI: 10.1177/1073858414562217] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Fibroblast growth factor homologous factors (FHFs) are a noncanonical subset of intracellular fibroblast growth factors that have been implicated in a variety of neurobiological processes and in disease. They are most prominently regulators of voltage-gated Na(+) channels (NaVs). In this review, we discuss new insights into how FHFs modulate NaVs. This is followed by a summary of a growing body of evidence that FHFs operate in much broader fashion. Finally, we highlight unknown aspects of FHF function as areas of future interest.
Collapse
Affiliation(s)
- Juan L Pablo
- Ion Channel Research Unit, Duke University Medical Center, Durham, NC, USA Department of Neurobiology, Duke University Medical Center, Durham, NC, USA
| | - Geoffrey S Pitt
- Ion Channel Research Unit, Duke University Medical Center, Durham, NC, USA Department of Neurobiology, Duke University Medical Center, Durham, NC, USA Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
40
|
Kim GE, Kronengold J, Barcia G, Quraishi IH, Martin HC, Blair E, Taylor JC, Dulac O, Colleaux L, Nabbout R, Kaczmarek LK. Human slack potassium channel mutations increase positive cooperativity between individual channels. Cell Rep 2014; 9:1661-1672. [PMID: 25482562 DOI: 10.1016/j.celrep.2014.11.015] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2014] [Revised: 10/06/2014] [Accepted: 11/10/2014] [Indexed: 11/24/2022] Open
Abstract
Disease-causing mutations in ion channels generally alter intrinsic gating properties such as activation, inactivation, and voltage dependence. We examined nine different mutations of the KCNT1 (Slack) Na(+)-activated K(+) channel that give rise to three distinct forms of epilepsy. All produced many-fold increases in current amplitude compared to the wild-type channel. This could not be accounted for by increases in the intrinsic open probability of individual channels. Rather, greatly increased opening was a consequence of cooperative interactions between multiple channels in a patch. The degree of cooperative gating was much greater for all of the mutant channels than for the wild-type channel, and could explain increases in current even in a mutant with reduced unitary conductance. We also found that the same mutation gave rise to different forms of epilepsy in different individuals. Our findings indicate that a major consequence of these mutations is to alter channel-channel interactions.
Collapse
Affiliation(s)
- Grace E Kim
- Department of Pharmacology, Yale University, New Haven, CT 06520, USA; Department of Cellular and Molecular Physiology, Yale University, New Haven, CT 06520, USA
| | - Jack Kronengold
- Department of Pharmacology, Yale University, New Haven, CT 06520, USA
| | - Giulia Barcia
- Department of Pediatric Neurology, Centre de Reference Epilepsies Rares, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, 75015 Paris, France
| | - Imran H Quraishi
- Comprehensive Epilepsy Center, Department of Neurology, Yale University, New Haven, CT 06520, USA
| | - Hilary C Martin
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Edward Blair
- Oxford University Hospitals Trust, Oxford OX3 9DU, UK
| | - Jenny C Taylor
- Oxford Biomedical Research Centre, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Olivier Dulac
- Department of Pediatric Neurology, Centre de Reference Epilepsies Rares, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, 75015 Paris, France
| | - Laurence Colleaux
- INSERM U781, Université Paris Descartes, Sorbonne Paris Cité, Institut Imagine, Hôpital Necker-Enfants Malades, 75015 Paris, France
| | - Rima Nabbout
- Department of Pediatric Neurology, Centre de Reference Epilepsies Rares, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, 75015 Paris, France
| | - Leonard K Kaczmarek
- Department of Pharmacology, Yale University, New Haven, CT 06520, USA; Department of Cellular and Molecular Physiology, Yale University, New Haven, CT 06520, USA.
| |
Collapse
|
41
|
Ohba C, Kato M, Takahashi S, Lerman-Sagie T, Lev D, Terashima H, Kubota M, Kawawaki H, Matsufuji M, Kojima Y, Tateno A, Goldberg-Stern H, Straussberg R, Marom D, Leshinsky-Silver E, Nakashima M, Nishiyama K, Tsurusaki Y, Miyake N, Tanaka F, Matsumoto N, Saitsu H. Early onset epileptic encephalopathy caused by de novoSCN8Amutations. Epilepsia 2014; 55:994-1000. [DOI: 10.1111/epi.12668] [Citation(s) in RCA: 129] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/22/2014] [Indexed: 12/13/2022]
Affiliation(s)
- Chihiro Ohba
- Department of Human Genetics; Graduate School of Medicine; Yokohama City University; Yokohama Japan
- Department of Clinical Neurology and Stroke Medicine; Yokohama City University; Yokohama Japan
| | - Mitsuhiro Kato
- Department of Pediatrics; Yamagata University Faculty of Medicine; Yamagata Japan
| | - Satoru Takahashi
- Department of Pediatrics; Asahikawa Medical University; Asahikawa Japan
| | | | - Dorit Lev
- Metabolic Neurogenetic Clinic; Wolfson Medical Center; Holon Israel
| | - Hiroshi Terashima
- Division of Neurology; National Center for Child Health and Development; Tokyo Japan
| | - Masaya Kubota
- Division of Neurology; National Center for Child Health and Development; Tokyo Japan
| | - Hisashi Kawawaki
- Department of Pediatric Neurology; Osaka City General Hospital; Osaka Japan
| | - Mayumi Matsufuji
- Department of Pediatrics; Japan Community Health Care Organization Kyusyu Hospital; Kitakyushu Japan
| | - Yasuko Kojima
- Department of Pediatrics; Toho University Sakura Medical Center; Chiba Japan
| | - Akihiko Tateno
- Department of Pediatrics; Toho University Sakura Medical Center; Chiba Japan
| | | | - Rachel Straussberg
- Department of Neurogenetics; Schneider's Children Medical Center; Petah Tiqwa Israel
| | - Dafna Marom
- Department of Neurogenetics; Schneider's Children Medical Center; Petah Tiqwa Israel
| | | | - Mitsuko Nakashima
- Department of Human Genetics; Graduate School of Medicine; Yokohama City University; Yokohama Japan
| | - Kiyomi Nishiyama
- Department of Human Genetics; Graduate School of Medicine; Yokohama City University; Yokohama Japan
| | - Yoshinori Tsurusaki
- Department of Human Genetics; Graduate School of Medicine; Yokohama City University; Yokohama Japan
| | - Noriko Miyake
- Department of Human Genetics; Graduate School of Medicine; Yokohama City University; Yokohama Japan
| | - Fumiaki Tanaka
- Department of Clinical Neurology and Stroke Medicine; Yokohama City University; Yokohama Japan
| | - Naomichi Matsumoto
- Department of Human Genetics; Graduate School of Medicine; Yokohama City University; Yokohama Japan
| | - Hirotomo Saitsu
- Department of Human Genetics; Graduate School of Medicine; Yokohama City University; Yokohama Japan
| |
Collapse
|
42
|
Galea CA, Nguyen HM, George Chandy K, Smith BJ, Norton RS. Domain structure and function of matrix metalloprotease 23 (MMP23): role in potassium channel trafficking. Cell Mol Life Sci 2014; 71:1191-210. [PMID: 23912897 PMCID: PMC11113776 DOI: 10.1007/s00018-013-1431-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2013] [Revised: 07/17/2013] [Accepted: 07/18/2013] [Indexed: 10/26/2022]
Abstract
MMP23 is a member of the matrix metalloprotease family of zinc- and calcium-dependent endopeptidases, which are involved in a wide variety of cellular functions. Its catalytic domain displays a high degree of structural homology with those of other metalloproteases, but its atypical domain architecture suggests that it may possess unique functional properties. The N-terminal MMP23 pro-domain contains a type-II transmembrane domain that anchors the protein to the plasma membrane and lacks the cysteine-switch motif that is required to maintain other MMPs in a latent state during passage to the cell surface. Instead of the C-terminal hemopexin domain common to other MMPs, MMP23 contains a small toxin-like domain (TxD) and an immunoglobulin-like cell adhesion molecule (IgCAM) domain. The MMP23 pro-domain can trap Kv1.3 but not closely-related Kv1.2 channels in the endoplasmic reticulum, preventing their passage to the cell surface, while the TxD can bind to the channel pore and block the passage of potassium ions. The MMP23 C-terminal IgCAM domain displays some similarity to Ig-like C2-type domains found in IgCAMs of the immunoglobulin superfamily, which are known to mediate protein-protein and protein-lipid interactions. MMP23 and Kv1.3 are co-expressed in a variety of tissues and together are implicated in diseases including cancer and inflammatory disorders. Further studies are required to elucidate the mechanism of action of this unique member of the MMP family.
Collapse
Affiliation(s)
- Charles A Galea
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, 3052, Australia,
| | | | | | | | | |
Collapse
|
43
|
Lin WH, Baines RA. Regulation of membrane excitability: a convergence on voltage-gated sodium conductance. Mol Neurobiol 2014; 51:57-67. [PMID: 24677068 PMCID: PMC4309913 DOI: 10.1007/s12035-014-8674-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Accepted: 03/11/2014] [Indexed: 11/30/2022]
Abstract
The voltage-gated sodium channel (Nav) plays a key role in regulation of neuronal excitability. Aberrant regulation of Nav expression and/or function can result in an imbalance in neuronal activity which can progress to epilepsy. Regulation of Nav activity is achieved by coordination of a multitude of mechanisms including RNA alternative splicing and translational repression. Understanding of these regulatory mechanisms is complicated by extensive genetic redundancy: the mammalian genome encodes ten Navs. By contrast, the genome of the fruitfly, Drosophila melanogaster, contains just one Nav homologue, encoded by paralytic (DmNa v ). Analysis of splicing in DmNa v shows variants exhibit distinct gating properties including varying magnitudes of persistent sodium current (INaP). Splicing by Pasilla, an identified RNA splicing factor, alters INaP magnitude as part of an activity-dependent mechanism. Enhanced INaP promotes membrane hyperexcitability that is associated with seizure-like behaviour in Drosophila. Nova-2, a mammalian Pasilla homologue, has also been linked to splicing of Navs and, moreover, mouse gene knockouts display seizure-like behaviour.Expression level of Navs is also regulated through a mechanism of translational repression in both flies and mammals. The translational repressor Pumilio (Pum) can bind to Na v transcripts and repress the normal process of translation, thus regulating sodium current (INa) density in neurons. Pum2-deficient mice exhibit spontaneous EEG abnormalities. Taken together, aberrant regulation of Nav function and/or expression is often epileptogenic. As such, a better understanding of regulation of membrane excitability through RNA alternative splicing and translational repression of Navs should provide new leads to treat epilepsy.
Collapse
Affiliation(s)
- Wei-Hsiang Lin
- Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester, UK
| | | |
Collapse
|
44
|
Chuang CC, Jow GM, Lin HM, Weng YH, Hu JH, Peng YJ, Chiu YC, Chiu MM, Jeng CJ. The punctate localization of rat Eag1 K+ channels is conferred by the proximal post-CNBHD region. BMC Neurosci 2014; 15:23. [PMID: 24495567 PMCID: PMC3926332 DOI: 10.1186/1471-2202-15-23] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2013] [Accepted: 01/31/2014] [Indexed: 01/12/2023] Open
Abstract
Background In mammals, Eag K+ channels (KV10) are exclusively expressed in the brain and comprise two isoforms: Eag1 (KV10.1) and Eag2 (KV10.2). Despite their wide presence in various regions of the brain, the functional role of Eag K+ channels remains obscure. Here we address this question by characterizing the subcellular localization of rat Eag1 (rEag1) and rat Eag2 (rEag2) in hippocampal neurons, as well as determining the structural basis underlying their different localization patterns. Results Immunofluorescence analysis of young and mature hippocampal neurons in culture revealed that endogenous rEag1 and rEag2 K+ channels were present in both the dendrosomatic and the axonal compartments. Only rEag1 channels displayed a punctate immunostaining pattern and showed significant co-localization with PSD-95. Subcellular fractionation analysis further demonstrated a distinct enrichment of rEag1 in the synaptosomal fraction. Over-expression of recombinant GFP-tagged Eag constructs in hippocampal neurons also showed a significant punctate localization of rEag1 channels. To identify the protein region dictating the Eag channel subcellular localization pattern, we generated a variety of different chimeric constructs between rEag1 and rEag2. Quantitative studies of neurons over-expressing these GFP-tagged chimeras indicated that punctate localization was conferred by a segment (A723-R807) within the proximal post-cyclic nucleotide-binding homology domain (post-CNBHD) region in the rEag1 carboxyl terminus. Conclusions Our findings suggest that Eag1 and Eag2 K+ channels may modulate membrane excitability in both the dendrosomatic and the axonal compartments and that Eag1 may additionally regulate neurotransmitter release and postsynaptic signaling. Furthermore, we present the first evidence showing that the proximal post-CNBHD region seems to govern the Eag K+ channel subcellular localization pattern.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Chung-Jiuan Jeng
- Institute of Anatomy and Cell Biology, School of Medicine, National Yang-Ming University, No, 155, Section 2, Li-Non Street, Taipei 12212, Taiwan.
| |
Collapse
|
45
|
Dustrude ET, Wilson SM, Ju W, Xiao Y, Khanna R. CRMP2 protein SUMOylation modulates NaV1.7 channel trafficking. J Biol Chem 2013; 288:24316-31. [PMID: 23836888 DOI: 10.1074/jbc.m113.474924] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Voltage-gated sodium channel (NaV) trafficking is incompletely understood. Post-translational modifications of NaVs and/or auxiliary subunits and protein-protein interactions have been posited as NaV-trafficking mechanisms. Here, we tested if modification of the axonal collapsin response mediator protein 2 (CRMP2) by a small ubiquitin-like modifier (SUMO) could affect NaV trafficking; CRMP2 alters the extent of NaV slow inactivation conferred by the anti-epileptic (R)-lacosamide, implying NaV-CRMP2 functional coupling. Expression of a CRMP2 SUMOylation-incompetent mutant (CRMP2-K374A) in neuronal model catecholamine A differentiated (CAD) cells did not alter lacosamide-induced NaV slow inactivation compared with CAD cells expressing wild type CRMP2. Like wild type CRMP2, CRMP2-K374A expressed robustly in CAD cells. Neurite outgrowth, a canonical CRMP2 function, was moderately reduced by the mutation but was still significantly higher than enhanced GFP-transfected cortical neurons. Notably, huwentoxin-IV-sensitive NaV1.7 currents, which predominate in CAD cells, were significantly reduced in CAD cells expressing CRMP2-K374A. Increasing deSUMOylation with sentrin/SUMO-specific protease SENP1 or SENP2 in wild type CRMP2-expressing CAD cells decreased NaV1.7 currents. Consistent with a reduction in current density, biotinylation revealed a significant reduction in surface NaV1.7 levels in CAD cells expressing CRMP2-K374A; surface NaV1.7 expression was also decreased by SENP1 + SENP2 overexpression. Currents in HEK293 cells stably expressing NaV1.7 were reduced by CRMP2-K374A in a manner dependent on the E2-conjugating enzyme Ubc9. No decrement in current density was observed in HEK293 cells co-expressing CRMP2-K374A and NaV1.1 or NaV1.3. Diminution of sodium currents, largely NaV1.7, was recapitulated in sensory neurons expressing CRMP2-K374A. Our study elucidates a novel regulatory mechanism that utilizes CRMP2 SUMOylation to choreograph NaV1.7 trafficking.
Collapse
Affiliation(s)
- Erik T Dustrude
- Paul and Carole Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA
| | | | | | | | | |
Collapse
|