1
|
Yagishita H, Go Y, Okamoto K, Arimura N, Ikegaya Y, Sasaki T. A method to analyze gene expression profiles from hippocampal neurons electrophysiologically recorded in vivo. Front Neurosci 2024; 18:1360432. [PMID: 38694898 PMCID: PMC11061373 DOI: 10.3389/fnins.2024.1360432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Accepted: 03/26/2024] [Indexed: 05/04/2024] Open
Abstract
Hippocampal pyramidal neurons exhibit diverse spike patterns and gene expression profiles. However, their relationships with single neurons are not fully understood. In this study, we designed an electrophysiology-based experimental procedure to identify gene expression profiles using RNA sequencing of single hippocampal pyramidal neurons whose spike patterns were recorded in living mice. This technique involves a sequence of experiments consisting of in vivo juxtacellular recording and labeling, brain slicing, cell collection, and transcriptome analysis. We demonstrated that the expression levels of a subset of genes in individual hippocampal pyramidal neurons were significantly correlated with their spike burstiness, submillisecond-level spike rise times or spike rates, directly measured by in vivo electrophysiological recordings. Because this methodological approach can be applied across a wide range of brain regions, it is expected to contribute to studies on various neuronal heterogeneities to understand how physiological spike patterns are associated with gene expression profiles.
Collapse
Affiliation(s)
- Haruya Yagishita
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, Japan
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Yasuhiro Go
- Graduate School of Information Science, University of Hyogo, Hyogo, Japan
- Department of System Neuroscience, Division of Behavioral Development, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Aichi, Japan
- Cognitive Genomics Research Group, Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki, Aichi, Japan
| | - Kazuki Okamoto
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
- Department of Neuroanatomy, Graduate School of Medicine, Juntendo University, Tokyo, Japan
- Department of Cell Biology and Neuroscience, Graduate School of Medicine, Juntendo University, Bunkyo, Tokyo, Japan
| | - Nariko Arimura
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, Japan
| | - Yuji Ikegaya
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
- Center for Information and Neural Networks, National Institute of Information and Communications Technology, Osaka, Japan
- Institute for AI and Beyond, The University of Tokyo, Tokyo, Japan
| | - Takuya Sasaki
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, Japan
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
2
|
Fang H, Hu W, Kang Q, Kuang X, Wang L, Zhang X, Liao H, Yang L, Yang H, Jiang Z, Wu L. Clinical characteristics and genetic analysis of pediatric patients with sodium channel gene mutation-related childhood epilepsy: a review of 94 patients. Front Neurol 2023; 14:1310419. [PMID: 38174099 PMCID: PMC10764033 DOI: 10.3389/fneur.2023.1310419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 11/27/2023] [Indexed: 01/05/2024] Open
Abstract
Objective This study aimed to examine the clinical and gene-mutation characteristics of pediatric patients with sodium channel gene mutation-related childhood epilepsy and to provide a basis for precision treatment and genetic counseling. Methods The clinical data from 94 patients with sodium channel gene mutation-related childhood epilepsy who were treated at Hunan Children's Hospital from August 2012 to December 2022 were retrospectively evaluated, and the clinical characteristics, gene variants, treatment, and follow-up status were analyzed and summarized. Results Our 94 pediatric patients with sodium channel gene variant-related childhood epilepsy comprised 37 girls and 57 boys. The age of disease onset ranged from 1 day to 3 years. We observed seven different sodium channel gene variants, and 55, 14, 9, 6, 6, 2, and 2 patients had SCNlA, SCN2A, SCN8A, SCN9A, SCN1B, SCN11A, and SCN3A variants, respectively. We noted that 52 were reported variants and 42 were novel variants. Among all gene types, SCN1A, SCN2A, and SCN8A variants were associated with an earlier disease onset age. With the exception of the SCN1B, the other six genes were associated with clustering seizures. Except for variants SCN3A and SCN11A, some patients with other variants had status epilepticus (SE). The main diagnosis of children with SCN1A variants was Dravet syndrome (DS) (72.7%), whereas patients with SCN2A and SCN8A variants were mainly diagnosed with various types of epileptic encephalopathy, accounting for 85.7% (12 of 14) and 88.9% (8 of 9) respectively. A total of five cases of sudden unexpected death in epilepsy (SUDEP) occurred in patients with SCN1A, SCN2A, and SCN8A variants. The proportion of benign epilepsy in patients with SCN9A, SCN11A, and SCN1B variants was relatively high, and the epilepsy control rate was higher than the rate of other variant types. Conclusion Sodium channel gene variants involve different epileptic syndromes, and the treatment responses also vary. We herein reported 42 novel variants, and we are also the first ever to report two patients with SCN11A variants, thereby increasing the gene spectrum and phenotypic profile of sodium channel dysfunction. We provide a basis for precision treatment and prognostic assessment.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Liwen Wu
- Neurology Department, Hunan Children's Hospital, Changsha, China
| |
Collapse
|
3
|
Finkelstein DS, Du Bois J. Trifunctional Saxitoxin Conjugates for Covalent Labeling of Voltage-Gated Sodium Channels. Chembiochem 2023; 24:e202300493. [PMID: 37746898 PMCID: PMC10863845 DOI: 10.1002/cbic.202300493] [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: 07/04/2023] [Revised: 08/24/2023] [Indexed: 09/26/2023]
Abstract
Voltage-gated sodium ion channels (NaV s) are integral membrane protein complexes responsible for electrical signal conduction in excitable cells. Methods that enable selective labeling of NaV s hold potential value for understanding how channel regulation and post-translational modification are influenced during development and in response to diseases and disorders of the nervous system. We have developed chemical reagents patterned after (+)-saxitoxin (STX) - a potent and reversible inhibitor of multiple NaV isoforms - and affixed with a reactive electrophile and either a biotin cofactor, fluorophore, or 'click' functional group for labeling wild-type channels. Our studies reveal enigmatic structural effects of the probes on the potency and efficiency of covalent protein modification. Among the compounds analyzed, a STX-maleimide-coumarin derivative is most effective at irreversibly blocking Na+ conductance when applied to recombinant NaV s and endogenous channels expressed in hippocampal neurons. Mechanistic analysis supports the conclusion that high-affinity toxin binding is a prerequisite for covalent protein modification. Results from these studies are guiding the development of next-generation tool compounds for selective modification of NaV s expressed in the plasma membranes of cells.
Collapse
Affiliation(s)
- Darren S Finkelstein
- Department of Chemistry, Stanford University, 337 Campus Dr., Stanford, CA 94305, USA
- Present address: Pliant Therapeutics, 260 Littlefield Avenue, South San Francisco, CA 94080, USA
| | - J Du Bois
- Department of Chemistry, Stanford University, 337 Campus Dr., Stanford, CA 94305, USA
| |
Collapse
|
4
|
Lu TL, Wu SN. Investigating the Impact of Selective Modulators on the Renin-Angiotensin-Aldosterone System: Unraveling Their Off-Target Perturbations of Transmembrane Ionic Currents. Int J Mol Sci 2023; 24:14007. [PMID: 37762309 PMCID: PMC10530685 DOI: 10.3390/ijms241814007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 09/05/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023] Open
Abstract
The renin-angiotensin-aldosterone system (RAAS) plays a crucial role in maintaining various physiological processes in the body, including blood pressure regulation, electrolyte balance, and overall cardiovascular health. However, any compounds or drugs known to perturb the RAAS might have an additional impact on transmembrane ionic currents. In this retrospective review article, we aimed to present a selection of chemical compounds or medications that have long been recognized as interfering with the RAAS. It is noteworthy that these substances may also exhibit regulatory effects in different types of ionic currents. Apocynin, known to attenuate the angiotensin II-induced activation of epithelial Na+ channels, was shown to stimulate peak and late components of voltage-gated Na+ current (INa). Esaxerenone, an antagonist of the mineralocorticoid receptor, can exert an inhibitory effect on peak and late INa directly. Dexamethasone, a synthetic glucocorticoid, can directly enhance the open probability of large-conductance Ca2+-activated K+ channels. Sparsentan, a dual-acting antagonist of the angiotensin II receptor and endothelin type A receptors, was found to suppress the amplitude of peak and late INa effectively. However, telmisartan, a blocker of the angiotensin II receptor, was effective in stimulating the peak and late INa along with a slowing of the inactivation time course of the current. However, telmisartan's presence can also suppress the erg-mediated K+ current. Moreover, tolvaptan, recognized as an aquaretic agent that can block the vasopressin receptor, was noted to suppress the amplitude of the delayed-rectifier K+ current and the M-type K+ current directly. The above results indicate that these substances not only have an interference effect on the RAAS but also exert regulatory effects on different types of ionic currents. Therefore, to determine their mechanisms of action, it is necessary to gain a deeper understanding.
Collapse
Affiliation(s)
- Te-Ling Lu
- School of Pharmacy, China Medical University, Taichung 406040, Taiwan;
| | - Sheng-Nan Wu
- Department of Research and Education, An Nan Hospital, China Medical University, Tainan 709040, Taiwan
- School of Medicine, College of Medicine, National Sun Yat-sen University, Kaohsiung 804201, Taiwan
| |
Collapse
|
5
|
Gould HJ, Paul D. Targeted Osmotic Lysis: A Novel Approach to Targeted Cancer Therapies. Biomedicines 2022; 10:biomedicines10040838. [PMID: 35453588 PMCID: PMC9027517 DOI: 10.3390/biomedicines10040838] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 03/28/2022] [Accepted: 03/30/2022] [Indexed: 02/04/2023] Open
Abstract
The conventional treatment of cancer has been based on the delivery of non-selective toxins and/or ionizing energy that affect both the cancer and normal tissues in the hope of destroying the offending disease before killing the patient. Unfortunately, resistance often develops to these treatments and patients experience severe, dose-limiting adverse effects that reduce treatment efficacy and compromise quality of life. Recent advances in our knowledge of the biology of tumor cells and their microenvironment, the recognition of surface proteins that are unique to specific cancers and essential to cell growth and survival and signaling pathways associate with invasion and metastasis have led to the development of targeted therapies that are able to identify specific cellular markers and more selectively deliver lethal treatment to the invading cancer thus improving efficacy and limiting adverse effects. In the context of targeted approaches to cancer therapy, we present targeted osmotic lysis as a novel and fundamentally different approach for treating advanced-stage carcinoma that exploits the conserved relationship between voltage-gated sodium channels and Na+, K+-ATPase and has the potential to increase survival without compromising quality of life in a broad spectrum of highly malignant forms of cancer.
Collapse
Affiliation(s)
- Harry J. Gould
- Department of Neurology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
- Correspondence: (H.J.G.III); (D.P.); Tel.: +1-504-568-5080 (H.J.G.III); +1-504-568-4745 (D.P.)
| | - Dennis Paul
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
- Correspondence: (H.J.G.III); (D.P.); Tel.: +1-504-568-5080 (H.J.G.III); +1-504-568-4745 (D.P.)
| |
Collapse
|
6
|
Dvorak NM, Tapia CM, Singh AK, Baumgartner TJ, Wang P, Chen H, Wadsworth PA, Zhou J, Laezza F. Pharmacologically Targeting the Fibroblast Growth Factor 14 Interaction Site on the Voltage-Gated Na + Channel 1.6 Enables Isoform-Selective Modulation. Int J Mol Sci 2021; 22:ijms222413541. [PMID: 34948337 PMCID: PMC8708424 DOI: 10.3390/ijms222413541] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 12/15/2021] [Accepted: 12/15/2021] [Indexed: 01/05/2023] Open
Abstract
Voltage-gated Na+ (Nav) channels are the primary molecular determinant of the action potential. Among the nine isoforms of the Nav channel α subunit that have been described (Nav1.1-Nav1.9), Nav1.1, Nav1.2, and Nav1.6 are the primary isoforms expressed in the central nervous system (CNS). Crucially, these three CNS Nav channel isoforms display differential expression across neuronal cell types and diverge with respect to their subcellular distributions. Considering these differences in terms of their localization, the CNS Nav channel isoforms could represent promising targets for the development of targeted neuromodulators. However, current therapeutics that target Nav channels lack selectivity, which results in deleterious side effects due to modulation of off-target Nav channel isoforms. Among the structural components of the Nav channel α subunit that could be pharmacologically targeted to achieve isoform selectivity, the C-terminal domains (CTD) of Nav channels represent promising candidates on account of displaying appreciable amino acid sequence divergence that enables functionally unique protein–protein interactions (PPIs) with Nav channel auxiliary proteins. In medium spiny neurons (MSNs) of the nucleus accumbens (NAc), a critical brain region of the mesocorticolimbic circuit, the PPI between the CTD of the Nav1.6 channel and its auxiliary protein fibroblast growth factor 14 (FGF14) is central to the generation of electrical outputs, underscoring its potential value as a site for targeted neuromodulation. Focusing on this PPI, we previously developed a peptidomimetic derived from residues of FGF14 that have an interaction site on the CTD of the Nav1.6 channel. In this work, we show that whereas the compound displays dose-dependent effects on the activity of Nav1.6 channels in heterologous cells, the compound does not affect Nav1.1 or Nav1.2 channels at comparable concentrations. In addition, we show that the compound correspondingly modulates the action potential discharge and the transient Na+ of MSNs of the NAc. Overall, these results demonstrate that pharmacologically targeting the FGF14 interaction site on the CTD of the Nav1.6 channel is a strategy to achieve isoform-selective modulation, and, more broadly, that sites on the CTDs of Nav channels interacted with by auxiliary proteins could represent candidates for the development of targeted therapeutics.
Collapse
|
7
|
Quinn RK, Drury HR, Cresswell ET, Tadros MA, Nayagam BA, Callister RJ, Brichta AM, Lim R. Expression and Physiology of Voltage-Gated Sodium Channels in Developing Human Inner Ear. Front Neurosci 2021; 15:733291. [PMID: 34759790 PMCID: PMC8575412 DOI: 10.3389/fnins.2021.733291] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 09/29/2021] [Indexed: 11/13/2022] Open
Abstract
Sodium channel expression in inner ear afferents is essential for the transmission of vestibular and auditory information to the central nervous system. During development, however, there is also a transient expression of Na+ channels in vestibular and auditory hair cells. Using qPCR analysis, we describe the expression of four Na+ channel genes, SCN5A (Nav1.5), SCN8A (Nav1.6), SCN9A (Nav1.7), and SCN10A (Nav1.8) in the human fetal cristae ampullares, utricle, and base, middle, and apex of the cochlea. Our data show distinct patterns of Na+ channel gene expression with age and between these inner ear organs. In the utricle, there was a general trend toward fold-change increases in expression of SCN8A, SCN9A, and SCN10A with age, while the crista exhibited fold-change increases in SCN5A and SCN8A and fold-change decreases in SCN9A and SCN10A. Fold-change differences of each gene in the cochlea were more complex and likely related to distinct patterns of expression based on tonotopy. Generally, the relative expression of SCN genes in the cochlea was greater than that in utricle and cristae ampullares. We also recorded Na+ currents from developing human vestibular hair cells aged 10-11 weeks gestation (WG), 12-13 WG, and 14+ WG and found there is a decrease in the number of vestibular hair cells that exhibit Na+ currents with increasing gestational age. Na+ current properties and responses to the application of tetrodotoxin (TTX; 1 μM) in human fetal vestibular hair cells are consistent with those recorded in other species during embryonic and postnatal development. Both TTX-sensitive and TTX-resistant currents are present in human fetal vestibular hair cells. These results provide a timeline of sodium channel gene expression in inner ear neuroepithelium and the physiological characterization of Na+ currents in human fetal vestibular neuroepithelium. Understanding the normal developmental timeline of ion channel gene expression and when cells express functional ion channels is essential information for regenerative technologies.
Collapse
Affiliation(s)
- Rikki K Quinn
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, Australia.,Hunter Medical Research Institute, The University of Newcastle, New Lambton Heights, NSW, Australia
| | - Hannah R Drury
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, Australia.,Hunter Medical Research Institute, The University of Newcastle, New Lambton Heights, NSW, Australia
| | - Ethan T Cresswell
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, Australia.,Hunter Medical Research Institute, The University of Newcastle, New Lambton Heights, NSW, Australia
| | - Melissa A Tadros
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, Australia.,Hunter Medical Research Institute, The University of Newcastle, New Lambton Heights, NSW, Australia
| | - Bryony A Nayagam
- Department of Audiology and Speech Pathology, The University of Melbourne, Parkville, VIC, Australia
| | - Robert J Callister
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, Australia.,Hunter Medical Research Institute, The University of Newcastle, New Lambton Heights, NSW, Australia
| | - Alan M Brichta
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, Australia.,Hunter Medical Research Institute, The University of Newcastle, New Lambton Heights, NSW, Australia
| | - Rebecca Lim
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, Australia.,Hunter Medical Research Institute, The University of Newcastle, New Lambton Heights, NSW, Australia
| |
Collapse
|
8
|
Kamata S, Kimura M, Ohyama S, Yamashita S, Shibukawa Y. Large-Conductance Calcium-Activated Potassium Channels and Voltage-Dependent Sodium Channels in Human Cementoblasts. Front Physiol 2021; 12:634846. [PMID: 33959036 PMCID: PMC8093401 DOI: 10.3389/fphys.2021.634846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Accepted: 03/17/2021] [Indexed: 12/02/2022] Open
Abstract
Cementum, which is excreted by cementoblasts, provides an attachment site for collagen fibers that connect to the alveolar bone and fix the teeth into the alveolar sockets. Transmembrane ionic signaling, associated with ionic transporters, regulate various physiological processes in a wide variety of cells. However, the properties of the signals generated by plasma membrane ionic channels in cementoblasts have not yet been described in detail. We investigated the biophysical and pharmacological properties of ion channels expressed in human cementoblast (HCEM) cell lines by measuring ionic currents using conventional whole-cell patch-clamp recording. The application of depolarizing voltage steps in 10 mV increments from a holding potential (Vh) of −70 mV evoked outwardly rectifying currents at positive potentials. When intracellular K+ was substituted with an equimolar concentration of Cs+, the outward currents almost disappeared. Using tail current analysis, the contributions of both K+ and background Na+ permeabilities were estimated for the outward currents. Extracellular application of tetraethylammonium chloride (TEA) and iberiotoxin (IbTX) reduced the densities of the outward currents significantly and reversibly, whereas apamin and TRAM-34 had no effect. When the Vh was changed to −100 mV, we observed voltage-dependent inward currents in 30% of the recorded cells. These results suggest that HCEM express TEA- and IbTX-sensitive large-conductance Ca2+-activated K+ channels and voltage-dependent Na+ channels.
Collapse
Affiliation(s)
- Satomi Kamata
- Department of Removable Partial Prosthodontics, Tokyo Dental College, Tokyo, Japan.,Department of Physiology, Tokyo Dental College, Tokyo, Japan
| | - Maki Kimura
- Department of Physiology, Tokyo Dental College, Tokyo, Japan
| | - Sadao Ohyama
- Department of Physiology, Tokyo Dental College, Tokyo, Japan
| | - Shuichiro Yamashita
- Department of Removable Partial Prosthodontics, Tokyo Dental College, Tokyo, Japan
| | | |
Collapse
|
9
|
The Specific Effects of OD-1, a Peptide Activator, on Voltage-Gated Sodium Current and Seizure Susceptibility. Int J Mol Sci 2020; 21:ijms21218254. [PMID: 33158049 PMCID: PMC7663472 DOI: 10.3390/ijms21218254] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Revised: 10/24/2020] [Accepted: 10/29/2020] [Indexed: 12/12/2022] Open
Abstract
OD-1, a scorpion toxin, has been previously recognized as an activator of voltage-gated Na+ currents. To what extent this agent can alter hippocampal neuronal Na+ currents and network excitability and how it can be applied to neuronal hyperexcitability research remains unclear. With the aid of patch-clamp technology, it was revealed that, in mHippoE-14 hippocampal neurons, OD-1 produced a concentration-, time-, and state-dependent rise in the peak amplitude of INa. It shifted the INa inactivation curve to a less negative potential and increased the frequency of spontaneous action currents. Further characterization of neuronal excitability revealed higher excitability in the hippocampal slices treated with OD-1 as compared with the control slices. A stereotaxic intrahippocampal injection of OD-1 generated a significantly higher frequency of spontaneous seizures and epileptiform discharges compared with intraperitoneal injection of lithium-pilocarpine- or kainic acid-induced epilepsy, with comparable pathological changes. Carbamazepine significantly attenuated OD-1 induced seizures and epileptiform discharges. The OD-1-mediated modifications of INa altered the electrical activity of neurons in vivo and OD-1 could potentially serve as a novel seizure and excitotoxicity model.
Collapse
|
10
|
Lai MC, Wu SN, Huang CW. Telmisartan, an Antagonist of Angiotensin II Receptors, Accentuates Voltage-Gated Na + Currents and Hippocampal Neuronal Excitability. Front Neurosci 2020; 14:902. [PMID: 33013297 PMCID: PMC7499822 DOI: 10.3389/fnins.2020.00902] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Accepted: 08/03/2020] [Indexed: 12/19/2022] Open
Abstract
Telmisartan (TEL), a non-peptide blocker of the angiotensin II type 1 receptor, is a widely used antihypertensive agent. Nevertheless, its neuronal ionic effects and how they potentially affect neuronal network excitability remain largely unclear. With the aid of patch-clamp technology, the effects of TEL on membrane ion currents present in hippocampal neurons (mHippoE-14 cells) were investigated. For additional characterization of the effects of TEL on hippocampal neuronal excitability, we undertook in vivo studies on Sprague Dawley (SD) rats using pilocarpine-induced seizure modeling, a hippocampal histopathological analysis, and inhibitory avoidance testing. In these hippocampal neurons, TEL increased the peak amplitude of INa, with a concomitant decline in the current inactivation rate. The TEL concentration dependently enhanced the peak amplitude of depolarization-elicited INa and lessened the inactivation rate of INa. By comparison, TEL was more efficacious in stimulating the peak INa and in prolonging the inactivation time course of this current than tefluthrin or (-)-epicatechin-3-gallate. In the continued presence of pioglitazone, the TEL-perturbed stimulation of INa remained effective. In addition, cell exposure to TEL shifted the steady-state inactivation INa curve to fewer negative potentials with no perturbations of the slope factor. Unlike chlorotoxin, either ranolazine, eugenol, or KMUP-1 reversed TEL-mediated increases in the strength of non-inactivating INa. In the cell-attached voltage-clamp recordings, TEL shortened the latency in the generation of action currents. Meanwhile, TEL increased the peak INa, with a concurrent decrease in current inactivation in HEKT293T cells expressing SCN5A. Furthermore, although TEL did not aggravate pilocarpine-induced chronic seizures and tended to preserve cognitive performance, it significantly accentuated hippocampal mossy fiber sprouting. Collectively, TEL stimulation of peak INa in combination with an apparent retardation in current inactivation could be an important mechanism through which hippocampal neuronal excitability is increased, and hippocampal network excitability is accentuated following status epilepticus, suggesting further attention to this finding.
Collapse
Affiliation(s)
- Ming-Chi Lai
- Department of Pediatrics, Chi-Mei Medical Center, Tainan, Taiwan
| | - Sheng-Nan Wu
- Department of Physiology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chin-Wei Huang
- Department of Neurology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
11
|
Technological advances and computational approaches for alternative splicing analysis in single cells. Comput Struct Biotechnol J 2020; 18:332-343. [PMID: 32099593 PMCID: PMC7033300 DOI: 10.1016/j.csbj.2020.01.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Accepted: 01/26/2020] [Indexed: 12/15/2022] Open
Abstract
Alternative splicing of RNAs generates isoform diversity, resulting in different proteins that are necessary for maintaining cellular function and identity. The discovery of alternative splicing has been revolutionized by next-generation transcriptomic sequencing mainly using bulk RNA-sequencing, which has unravelled RNA splicing and mis-splicing of normal cells under steady-state and stress conditions. Single-cell RNA-sequencing studies have focused on gene-level expression analysis and revealed gene expression signatures distinguishable between different cellular types. Single-cell alternative splicing is an emerging area of research with the promise to reveal transcriptomic dynamics invisible to bulk- and gene-level analysis. In this review, we will discuss the technological advances for single-cell alternative splicing analysis, computational strategies for isoform detection and quantitation in single cells, and current applications of single-cell alternative splicing analysis and its potential future contributions to personalized medicine.
Collapse
|
12
|
Ding J, Zhang JW, Guo YX, Zhang YX, Chen ZH, Zhai QX. Novel mutations in SCN9A occurring with fever-associated seizures or epilepsy. Seizure 2019; 71:214-218. [DOI: 10.1016/j.seizure.2019.06.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Revised: 06/03/2019] [Accepted: 06/06/2019] [Indexed: 10/26/2022] Open
|
13
|
SCN9A Epileptic Encephalopathy Mutations Display a Gain-of-function Phenotype and Distinct Sensitivity to Oxcarbazepine. Neurosci Bull 2019; 36:11-24. [PMID: 31372899 DOI: 10.1007/s12264-019-00413-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 04/12/2019] [Indexed: 02/05/2023] Open
Abstract
Genetic mutants of voltage-gated sodium channels (VGSCs) are considered to be responsible for the increasing number of epilepsy syndromes. Previous research has indicated that mutations of one of the VGSC genes, SCN9A (Nav1.7), result in febrile seizures and Dravet syndrome in humans. Despite these recent efforts, the electrophysiological basis of SCN9A mutations remains unclear. Here, we performed a genetic screen of patients with febrile seizures and identified a novel missense mutation of SCN9A (W1150R). Electrophysiological characterization of different SCN9A mutants in HEK293T cells, the previously-reported N641Y and K655R variants, as well as the newly-found W1150R variant, revealed that the current density of the W1150R and N641Y variants was significantly larger than that of the wild-type (WT) channel. The time constants of recovery from fast inactivation of the N641Y and K655R variants were markedly lower than in the WT channel. The W1150R variant caused a negative shift of the G-V curve in the voltage dependence of steady-state activation. All mutants displayed persistent currents larger than the WT channel. In addition, we found that oxcarbazepine (OXC), one of the antiepileptic drugs targeting VGSCs, caused a significant shift to more negative potential for the activation and inactivation in WT and mutant channels. OXC-induced inhibition of currents was weaker in the W1150R variant than in the WT. Furthermore, with administering OXC the time constant of the N641Y variant was longer than those of the other two SCN9A mutants. In all, our results indicated that the point mutation W1150R resulted in a novel gain-of-function variant. These findings indicated that SCN9A mutants contribute to an increase in seizure, and show distinct sensitivity to OXC.
Collapse
|
14
|
Zhang F, Zhang C, Xu X, Zhang Y, Gong X, Yang Z, Zhang H, Tang D, Liang S, Liu Z. Naja atra venom peptide reduces pain by selectively blocking the voltage-gated sodium channel Nav1.8. J Biol Chem 2019; 294:7324-7334. [PMID: 30804211 DOI: 10.1074/jbc.ra118.007370] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 02/20/2019] [Indexed: 01/14/2023] Open
Abstract
The voltage-gated sodium channel Nav1.8 is preferentially expressed in peripheral nociceptive neurons and contributes to inflammatory and neuropathic pain. Therefore, Nav1.8 has emerged as one of the most promising analgesic targets for pain relief. Using large-scale screening of various animal-derived toxins and venoms for Nav1.8 inhibitors, here we identified μ-EPTX-Na1a, a 62-residue three-finger peptide from the venom of the Chinese cobra (Naja atra), as a potent inhibitor of Nav1.8, exhibiting high selectivity over other voltage-gated sodium channel subtypes. Using whole-cell voltage-clamp recordings, we observed that purified μ-EPTX-Na1a blocked the Nav1.8 current. This blockade was associated with a depolarizing shift of activation and repolarizing shift of inactivation, a mechanism distinct from that of any other gating modifier toxin identified to date. In rodent models of inflammatory and neuropathic pain, μ-EPTX-Na1a alleviated nociceptive behaviors more potently than did morphine, indicating that μ-EPTX-Na1a has a potent analgesic effect. μ-EPTX-Na1a displayed no evident cytotoxicity and cardiotoxicity and produced no obvious adverse responses in mice even at a dose 30-fold higher than that producing a significant analgesic effect. Our study establishes μ-EPTX-Na1a as a promising lead for the development of Nav1.8-targeting analgesics to manage pain.
Collapse
Affiliation(s)
- Fan Zhang
- From The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081 Hunan, China
| | - Changxin Zhang
- From The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081 Hunan, China
| | - Xunxun Xu
- From The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081 Hunan, China
| | - Yunxiao Zhang
- From The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081 Hunan, China
| | - Xue Gong
- From The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081 Hunan, China
| | - Zuqin Yang
- From The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081 Hunan, China
| | - Heng Zhang
- From The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081 Hunan, China
| | - Dongfang Tang
- From The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081 Hunan, China
| | - Songping Liang
- From The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081 Hunan, China
| | - Zhonghua Liu
- From The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081 Hunan, China
| |
Collapse
|
15
|
Alves RM, Uva P, Veiga MF, Oppo M, Zschaber FCR, Porcu G, Porto HP, Persico I, Onano S, Cuccuru G, Atzeni R, Vieira LCN, Pires MVA, Cucca F, Toralles MBP, Angius A, Crisponi L. Novel ANKRD11 gene mutation in an individual with a mild phenotype of KBG syndrome associated to a GEFS+ phenotypic spectrum: a case report. BMC MEDICAL GENETICS 2019; 20:16. [PMID: 30642272 PMCID: PMC6332862 DOI: 10.1186/s12881-019-0745-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 01/03/2019] [Indexed: 11/17/2022]
Abstract
Background KBG syndrome is a very rare autosomal dominant disorder, characterized by macrodontia, distinctive craniofacial findings, skeletal findings, post-natal short stature, and developmental delays, sometimes associated with seizures and EEG abnormalities. So far, there have been over 100 cases of KBG syndrome reported. Case presentation Here, we describe two sisters of a non-consanguineous family, both presenting generalized epilepsy with febrile seizures (GEFS+), and one with a more complex phenotype associated with mild intellectual disability, skeletal and dental anomalies. Whole exome sequencing (WES) analysis in all the family members revealed a heterozygous SCN9A mutation, p.(Lys655Arg), shared among the father and the two probands, and a novel de novo loss of function mutation in the ANKRD11 gene, p.(Tyr1715*), in the proband with the more complex phenotype. The reassessment of the phenotypic features confirmed that the patient fulfilled the proposed diagnostic criteria for KBG syndrome, although complicated by early-onset isolated febrile seizures. EEG abnormalities with or without seizures have been reported previously in some KBG cases. The shared variant, occurring in SCN9A, has been previously found in several individuals with GEFS+ and Dravet syndrome. Conclusions This report describe a novel de novo variant in ANKRD11 causing a mild phenotype of KGB syndrome and further supports the association of monogenic pattern of SCN9A mutations with GEFS+. Our data expand the allelic spectrum of ANKRD11 mutations, providing the first Brazilian case of KBG syndrome. Furthermore, this study offers an example of how WES has been instrumental allowing us to better dissect the clinical phenotype under study, which is a multilocus variation aggregating in one proband, rather than a phenotypic expansion associated with a single genomic locus, underscoring the role of multiple rare variants at different loci in the etiology of clinical phenotypes making problematic the diagnostic path. The successful identification of the causal variant in a gene may not be sufficient, making it necessary to identify other variants that fully explain the clinical picture. The prevalence of blended phenotypes from multiple monogenic disorders is currently unknown and will require a systematic re-analysis of large WES datasets for proper diagnosis in daily practice.
Collapse
Affiliation(s)
- Rita Maria Alves
- Postgraduate Program in Interactive Processes of Organs and Systems - Federal University of Bahia, Salvador, Brazil.,Research group Epi-Genétic, Salvador, Bahia, Brazil
| | - Paolo Uva
- Centre for Advanced Studies, Research and Development in Sardinia (CRS4), Science and Technology Park Polaris, Pula, Italy
| | - Marielza F Veiga
- Postgraduate Program in Interactive Processes of Organs and Systems - Federal University of Bahia, Salvador, Brazil.,EEG Service and Clinical Outpatient of Epilepsy, University Hospital Complex Professor Edgard Santos (C-HUPES), Federal University of Bahia, Salvador, Bahia, Brazil
| | - Manuela Oppo
- Department of Biomedical Science, University of Sassari, Sassari, Italy
| | | | | | | | - Ivana Persico
- Institute of Genetic and Biomedical Research, National Research Council (CNR), Cittadella Universitaria di Cagliari, 09042, Monserrato, Cagliari, Italy
| | - Stefano Onano
- Department of Biomedical Science, University of Sassari, Sassari, Italy.,Institute of Genetic and Biomedical Research, National Research Council (CNR), Cittadella Universitaria di Cagliari, 09042, Monserrato, Cagliari, Italy
| | - Gianmauro Cuccuru
- Centre for Advanced Studies, Research and Development in Sardinia (CRS4), Science and Technology Park Polaris, Pula, Italy
| | - Rossano Atzeni
- Centre for Advanced Studies, Research and Development in Sardinia (CRS4), Science and Technology Park Polaris, Pula, Italy
| | - Lauro C N Vieira
- Clinic Ponto Alto diagnostic by Image, São Marcos, Salvador, Bahia, Brazil
| | - Marcos V A Pires
- Research group Epi-Genétic, Salvador, Bahia, Brazil.,Faculty of Medicine of the ABC, São Paulo, Brazil
| | - Francesco Cucca
- Department of Biomedical Science, University of Sassari, Sassari, Italy.,Institute of Genetic and Biomedical Research, National Research Council (CNR), Cittadella Universitaria di Cagliari, 09042, Monserrato, Cagliari, Italy
| | - Maria Betânia P Toralles
- Postgraduate Program in Interactive Processes of Organs and Systems - Federal University of Bahia, Salvador, Brazil
| | - Andrea Angius
- Department of Biomedical Science, University of Sassari, Sassari, Italy. .,Institute of Genetic and Biomedical Research, National Research Council (CNR), Cittadella Universitaria di Cagliari, 09042, Monserrato, Cagliari, Italy.
| | - Laura Crisponi
- Department of Biomedical Science, University of Sassari, Sassari, Italy.,Institute of Genetic and Biomedical Research, National Research Council (CNR), Cittadella Universitaria di Cagliari, 09042, Monserrato, Cagliari, Italy
| |
Collapse
|
16
|
Feng Y, Zhang S, Zhang Z, Guo J, Tan Z, Zhu Y, Tao J, Ji YH. Understanding Genotypes and Phenotypes of the Mutations in Voltage- Gated Sodium Channel α Subunits in Epilepsy. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2018; 18:266-272. [PMID: 30370865 DOI: 10.2174/1871527317666181026164825] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 05/16/2018] [Accepted: 05/16/2018] [Indexed: 12/19/2022]
Abstract
BACKGROUND & OBJECTIVE Voltage-gated sodium channels (VGSCs) are responsible for the generation and propagation of action potentials in most excitable cells. In general, a VGSC consists of one pore-forming α subunit and two auxiliary β subunits. Genetic alterations in VGSCs genes, including both α and β subunits, are considered to be associated with epileptogenesis as well as seizures. This review aims to summarize the mutations in VGSC α subunits in epilepsy, particularly the pathophysiological and pharmacological properties of relevant VGSC mutants. CONCLUSION The review of epilepsy-associated VGSC α subunits mutants may not only contribute to the understanding of disease mechanism and genetic modifiers, but also provide potential theoretical targets for the precision and individualized medicine for epilepsy.
Collapse
Affiliation(s)
- Yijun Feng
- Laboratory of Neuropharmacology and Neurotoxicology, Shanghai University, Shanghai 200444, China
| | - Shuzhang Zhang
- Laboratory of Neuropharmacology and Neurotoxicology, Shanghai University, Shanghai 200444, China
| | - Zhiping Zhang
- Laboratory of Neuropharmacology and Neurotoxicology, Shanghai University, Shanghai 200444, China
| | - Jingkang Guo
- Laboratory of Neuropharmacology and Neurotoxicology, Shanghai University, Shanghai 200444, China
| | - Zhiyong Tan
- Department of Pharmacology and Toxicology and Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, United States
| | - Yudan Zhu
- Central Laboratory, Department of Neurology and Neurosurgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jie Tao
- Central Laboratory, Department of Neurology and Neurosurgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yong-Hua Ji
- Laboratory of Neuropharmacology and Neurotoxicology, Shanghai University, Shanghai 200444, China
| |
Collapse
|
17
|
Misonou H. Precise localizations of voltage-gated sodium and potassium channels in neurons. Dev Neurobiol 2017; 78:271-282. [PMID: 29218789 DOI: 10.1002/dneu.22565] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 12/01/2017] [Accepted: 12/06/2017] [Indexed: 11/08/2022]
Abstract
Neurons are extremely large and complex cells, and they regulate membrane potentials in multiple subcellular compartments using a variety of ion channels. Voltage-gated sodium (Nav) and potassium (Kv) channels are crucial in regulating neuronal membrane excitability owing to their diversity in subtypes, biophysical properties, and localizations. In particular, specific localizations of Nav and Kv channels in specific membrane compartments are essential to achieve a precise control of local membrane excitability. Recent advancement in super-resolution microscopy further substantiated nanoscale localizations of different ion channels in neuronal membranes. New questions arise from these new lines of evidence regarding how Nav and Kv channels are trafficked to a specific location and maintained against lateral diffusion. In this review, the aim is to summarize current information about ion channel localizations at nanoscopic levels and discuss what we can infer regarding the mechanisms. © 2017 Wiley Periodicals, Inc. Develop Neurobiol 78: 271-282, 2018.
Collapse
Affiliation(s)
- Hiroaki Misonou
- Graduate School of Brain Science, Doshisha University, Kyoto, Japan
| |
Collapse
|
18
|
Fouillet A, Watson JF, Piekarz AD, Huang X, Li B, Priest B, Nisenbaum E, Sher E, Ursu D. Characterisation of Nav1.7 functional expression in rat dorsal root ganglia neurons by using an electrical field stimulation assay. Mol Pain 2017; 13:1744806917745179. [PMID: 29166836 PMCID: PMC5731621 DOI: 10.1177/1744806917745179] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background The Nav1.7 subtype of voltage-gated sodium channels is specifically expressed in sensory and sympathetic ganglia neurons where it plays an important role in the generation and transmission of information related to pain sensation. Human loss or gain-of-function mutations in the gene encoding Nav1.7 channels (SCN9A) are associated with either absence of pain, as reported for congenital insensitivity to pain, or with exacerbation of pain, as reported for primary erythromelalgia and paroxysmal extreme pain disorder. Based on this important human genetic evidence, numerous drug discovery efforts are ongoing in search for Nav1.7 blockers as a novel therapeutic strategy to treat pain conditions. Results We are reporting here a novel approach to study Nav1.7 function in cultured rat sensory neurons. We used live cell imaging combined with electrical field stimulation to evoke and record action potential-driven calcium transients in the neurons. We have shown that the tarantula venom peptide Protoxin-II, a known Nav1.7 subtype selective blocker, inhibited electrical field stimulation-evoked calcium responses in dorsal root ganglia neurons with an IC50 of 72 nM, while it had no activity in embryonic hippocampal neurons. The results obtained in the live cell imaging assay were supported by patch-clamp studies as well as by quantitative PCR and Western blotting experiments that confirmed the presence of Nav1.7 mRNA and protein in dorsal root ganglia but not in embryonic hippocampal neurons. Conclusions The findings presented here point to a selective effect of Protoxin-II in sensory neurons and helped to validate a new method for investigating and comparing Nav1.7 pharmacology in sensory versus central nervous system neurons. This will help in the characterisation of the selectivity of novel Nav1.7 modulators using native ion channels and will provide the basis for the development of higher throughput models for enabling pain-relevant phenotypic screening.
Collapse
Affiliation(s)
- Antoine Fouillet
- 1 Lilly Research Centre, 1539 Eli Lilly and Company , Windlesham, UK
| | - Jake F Watson
- 1 Lilly Research Centre, 1539 Eli Lilly and Company , Windlesham, UK
| | - Andrew D Piekarz
- 2 Lilly Research Laboratories, 1539 Eli Lilly and Company , IN, USA
| | - Xiaofang Huang
- 2 Lilly Research Laboratories, 1539 Eli Lilly and Company , IN, USA
| | - Baolin Li
- 2 Lilly Research Laboratories, 1539 Eli Lilly and Company , IN, USA
| | - Birgit Priest
- 2 Lilly Research Laboratories, 1539 Eli Lilly and Company , IN, USA
| | - Eric Nisenbaum
- 2 Lilly Research Laboratories, 1539 Eli Lilly and Company , IN, USA
| | - Emanuele Sher
- 1 Lilly Research Centre, 1539 Eli Lilly and Company , Windlesham, UK
| | - Daniel Ursu
- 1 Lilly Research Centre, 1539 Eli Lilly and Company , Windlesham, UK
| |
Collapse
|
19
|
Q10R mutation in SCN9A gene is associated with generalized epilepsy with febrile seizures plus. Seizure 2017; 50:186-188. [PMID: 28704742 DOI: 10.1016/j.seizure.2017.06.023] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Revised: 06/07/2017] [Accepted: 06/28/2017] [Indexed: 11/21/2022] Open
|
20
|
Calati R, Gressier F, Balestri M, Serretti A. Genetic modulation of borderline personality disorder: systematic review and meta-analysis. J Psychiatr Res 2013; 47:1275-87. [PMID: 23810197 DOI: 10.1016/j.jpsychires.2013.06.002] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2013] [Revised: 04/08/2013] [Accepted: 06/03/2013] [Indexed: 11/18/2022]
Abstract
Borderline personality disorder (BPD) is a highly prevalent psychiatric disorder with high morbidity and mortality. Early theories ascribed an environmental etiology of BPD, but growing evidence supports a genetic vulnerability as well. The primary aim of this study was to systematically review genetic association studies focused on BPD. PubMed, ISI Web of Knowledge and PsycINFO databases were searched for studies published until December 2012. Meta-analyses were also performed when three or more studies reported genetic data on the same polymorphism. Data were analyzed with Cochrane Collaboration Review Manager Software (RevMan, version 5.0). Quality and publication bias were assessed. The systematic review of association studies examining genetic polymorphisms and BPD produced conflicting results. Meta-analyses were performed for three serotonergic polymorphisms: two common polymorphisms of the serotonin transporter gene (SLC6A4), the promoter insertion/deletion (5-HTTLPR) and the intron 2 VNTR (STin2 VNTR), and the rs1800532 (A218C) polymorphism of the tryptophan hydroxylase 1 gene (TPH1), all showing no association. No direct role of genetic polymorphisms was found in BPD. However, a few studies only are present in literature to draw definite conclusions. Further studies focusing on gene × gene and gene × environment interactions are needed to more deeply dissect the genetic role in the modulation of BPD.
Collapse
Affiliation(s)
- Raffaella Calati
- IRCCS Centro S. Giovanni di Dio, Fatebenefratelli, Brescia, Italy
| | | | | | | |
Collapse
|
21
|
Gueret G, Guillouet M, Vermeersch V, Guillard E, Talarmin H, Nguyen BV, Rannou F, Giroux-Metges MA, Pennec JP, Ozier Y. [ICU acquired neuromyopathy]. ACTA ACUST UNITED AC 2013; 32:580-91. [PMID: 23958176 DOI: 10.1016/j.annfar.2013.05.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2012] [Accepted: 05/08/2013] [Indexed: 12/19/2022]
Abstract
ICU acquired neuromyopathy (IANM) is the most frequent neurological pathology observed in ICU. Nerve and muscle defects are merged with neuromuscular junction abnormalities. Its physiopathology is complex. The aim is probably the redistribution of nutriments and metabolism towards defense against sepsis. The main risk factors are sepsis, its severity and its duration of evolution. IANM is usually diagnosed in view of difficulties in weaning from mechanical ventilation, but electrophysiology may allow an earlier diagnosis. There is no curative therapy, but early treatment of sepsis, glycemic control as well as early physiotherapy may decrease its incidence. The outcomes of IANM are an increase in morbi-mortality and possibly long-lasting neuromuscular abnormalities as far as tetraplegia.
Collapse
Affiliation(s)
- G Gueret
- Pôle anesthésie réanimations soins intensifs blocs opératoires urgences (ARSIBOU), CHRU de Brest, boulevard Tanguy-Prigent, 29200 Brest, France; Laboratoire de physiologie, faculté de médecine et des sciences de la santé, EA 1274 (mouvement, sport santé), université de Bretagne-Occidentale, 22, avenue Camille-Desmoulins, 29200 Brest, France; Université européenne de Bretagne, 5, boulevard Laennec, 35000 Rennes, France.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Parsons WH, Du Bois J. Maleimide conjugates of saxitoxin as covalent inhibitors of voltage-gated sodium channels. J Am Chem Soc 2013; 135:10582-5. [PMID: 23855513 DOI: 10.1021/ja4019644] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
(+)-Saxitoxin, a naturally occurring guanidinium poison, functions as a potent, selective, and reversible inhibitor of voltage-gated sodium ion channels (NaVs). Modified forms of this toxin bearing cysteine-reactive maleimide groups are available through total synthesis and are found to irreversibly inhibit sodium ion conductance in recombinantly expressed wild-type sodium channels and in hippocampal nerve cells. Our findings support a mechanism for covalent protein modification in which toxin binding to the channel pore precedes maleimide alkylation of a nucleophilic amino acid. Second-generation maleimide-toxin conjugates, which include bioorthogonal reactive groups, are also found to block channel function irreversibly; such compounds have potential as reagents for selective labeling of NaVs for live cell imaging and/or proteomics experiments.
Collapse
Affiliation(s)
- William H Parsons
- Department of Chemistry, Stanford University, 333 Campus Drive, Stanford, California 94305-5080, USA
| | | |
Collapse
|
23
|
Nemoto T, Yanagita T, Maruta T, Sugita C, Satoh S, Kanai T, Wada A, Murakami M. Endothelin-1-induced down-regulation of NaV1.7 expression in adrenal chromaffin cells: attenuation of catecholamine secretion and tau dephosphorylation. FEBS Lett 2013; 587:898-905. [PMID: 23434582 DOI: 10.1016/j.febslet.2013.02.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2012] [Revised: 01/18/2013] [Accepted: 02/06/2013] [Indexed: 10/27/2022]
Abstract
Endothelin-1 and voltage-dependent sodium channels are involved in control and suppression of neuropathological factors, which contribute to sculpting the neuronal network. We previously demonstrated that veratridine-induced NaV1.7 sodium channel activation caused intracellular calcium elevation, catecholamine secretion and tau dephosphorylation in adrenal chromaffin cells. The aim of this study was to examine whether endothelin-1 could modulate NaV1.7. Our results indicated that endothelin-1 decreased the protein level of NaV1.7 and the veratridine-induced increase in intracellular calcium. In addition, it also abolished the veratridine-induced dephosphorylation of tau and the phosphorylation of glycogen synthase kinase-3β and extracellular signal-regulated kinase. These findings suggest that the endothelin-1-induced down-regulation of NaV1.7 diminishes NaV1.7-related catecholamine secretion and dephosphorylation of tau.
Collapse
Affiliation(s)
- Takayuki Nemoto
- Department of Pharmacology, Miyazaki Medical College, University of Miyazaki, Miyazaki, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Ichikawa H, Kim HJ, Shuprisha A, Shikano T, Tsumura M, Shibukawa Y, Tazaki M. Voltage-dependent sodium channels and calcium-activated potassium channels in human odontoblasts in vitro. J Endod 2012; 38:1355-62. [PMID: 22980177 DOI: 10.1016/j.joen.2012.06.015] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2012] [Revised: 06/12/2012] [Accepted: 06/14/2012] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Transmembrane ionic signaling regulates many cellular processes in both physiological and pathologic settings. In this study, the biophysical properties of voltage-dependent Na(+) channels in odontoblasts derived from human dental pulp (HOB cells) were investigated together with the effect of bradykinin on intracellular Ca(2+) signaling and expression of Ca(2+)-activated K(+) channels. METHODS Ionic channel activity was characterized by using whole-cell patch-clamp recording and fura-2 fluorescence. RESULTS Mean resting membrane potential in the HOB cells was -38 mV. Depolarizing steps from a holding potential of -80 mV activated transient voltage-dependent inward currents with rapid activation/inactivation properties. At a holding potential of -50 mV, no inward current was recorded. Fast-activation kinetics exhibited dependence on membrane potential, whereas fast-inactivation kinetics did not. Steady-state inactivation was described by a Boltzmann function with a half-maximal inactivation potential of -70 mV, indicating that whereas the channels were completely inactivated at physiological resting membrane potential, they could be activated when the cells were hyperpolarized. Inward currents disappeared in Na(+)-free extracellular solution. Bradykinin activated intracellular Ca(2+)-releasing and influx pathways. When the HOB cells were clamped at a holding potential of -50 mV, outward currents were recorded at positive potentials, indicating sensitivity to inhibitors of intermediate-conductance Ca(2+)-activated K(+) channels. CONCLUSIONS Human odontoblasts expressed voltage-dependent Na(+) channels, bradykinin receptors, and Ca(2+)-activated K(+) channels, which play an important role in driving cellular functions by channel-receptor signal interaction and membrane potential regulation.
Collapse
Affiliation(s)
- Hideki Ichikawa
- Department of Physiology, Tokyo Dental College, Chiba, Japan
| | | | | | | | | | | | | |
Collapse
|
25
|
Ren CT, Li DM, Ou SW, Wang YJ, Lin Y, Zong ZH, Kameyama M, Kameyama A. Cloning and expression of the two new variants of Nav1.5/SCN5A in rat brain. Mol Cell Biochem 2012; 365:139-48. [PMID: 22331407 DOI: 10.1007/s11010-012-1253-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2011] [Accepted: 01/13/2012] [Indexed: 12/19/2022]
Abstract
The α-subunit of tetrodotoxin-resistant (TTX-R) voltage-gated sodium channel (VSGC, Nav1.5/SCN5A) has been found from the rat heart and human neuroblastoma cell line NB-1, but its expression in rat brain has not been identified radically. In this study, a reverse transcriptase-polymerase chain reaction was used to clone the full sequence of Nav1.5 (designated as rN1) α-subunit in rat brain and compared the distribution in different lobe of brain in different developmental stages. The open reading frame of rN1 encodes 2,016 amino acid residues and sequence analysis indicated that rN1 is highly homologous with 96.53% amino acids identity to rat cardiac Nav1.5 (rH1) and 96.13% amino acids identity to human neuroblastoma Nav1.5 (hNbR1). It has all the structural features of a VSGC and the presence of a cysteine residue (C373) in the pore loop region of domain I suggests that this channel is resistant to TTX. A new exon (exon6A) that is distinct from rH1 was found in DI-S3-S4, meanwhile an isomer of alternative splicing that deleted 53 amino acids (exon18) was found for the first time in domain DII-III in rN1. (designated as rN1-2). Distribution results demonstrated that rN1 expressed discrepancy in different ages and lobe in brain. The expression level of rN1 was gradually more stable in adult than in neonatal; these results suggest that rN1 has a newly identified exon for alternative splicing that is differentfrom rat heart and is more widely expressed in rat brain than previously thought.
Collapse
Affiliation(s)
- Cheng-Tao Ren
- Department of Neurosurgery, The Affiliated Municipal Hospital, Medical College, Qingdao University, Qingdao, China
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Moore AR, Zhou WL, Jakovcevski I, Zecevic N, Antic SD. Physiological Properties of Human Fetal Cortex In Vitro. ISOLATED CENTRAL NERVOUS SYSTEM CIRCUITS 2012. [DOI: 10.1007/978-1-62703-020-5_3] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
27
|
Byers MR, Westenbroek RE. Odontoblasts in developing, mature and ageing rat teeth have multiple phenotypes that variably express all nine voltage-gated sodium channels. Arch Oral Biol 2011; 56:1199-220. [DOI: 10.1016/j.archoralbio.2011.04.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2011] [Revised: 04/20/2011] [Accepted: 04/21/2011] [Indexed: 12/11/2022]
|
28
|
O'Brien JE, Drews VL, Jones JM, Dugas JC, Barres BA, Meisler MH. Rbfox proteins regulate alternative splicing of neuronal sodium channel SCN8A. Mol Cell Neurosci 2011; 49:120-6. [PMID: 22044765 DOI: 10.1016/j.mcn.2011.10.005] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2011] [Revised: 10/10/2011] [Accepted: 10/14/2011] [Indexed: 12/17/2022] Open
Abstract
The SCN8A gene encodes the voltage-gated sodium channel Na(v)1.6, a major channel in neurons of the CNS and PNS. SCN8A contains two alternative exons,18N and 18A, that exhibit tissue specific splicing. In brain, the major SCN8A transcript contains exon 18A and encodes the full-length sodium channel. In other tissues, the major transcript contains exon 18N and encodes a truncated protein, due to the presence of an in-frame stop codon. Selection of exon 18A is therefore essential for generation of a functional channel protein, but the proteins involved in this selection have not been identified. Using a 2.6 kb Scn8a minigene containing exons 18N and 18A, we demonstrate that co-transfection with Fox-1 or Fox-2 initiates inclusion of exon 18A. This effect is dependent on the consensus Fox binding site located 28 bp downstream of exon 18A. We examined the alternative splicing of human SCN8A and found that the postnatal switch to exon 18A is completed later than 10 months of age. In purified cell populations, transcripts containing exon 18A predominate in neurons but are not present in oligodendrocytes or astrocytes. Transcripts containing exon 18N appear to be degraded by nonsense-mediated decay in HEK cells. Our data indicate that RBFOX proteins contribute to the cell-specific expression of Na(v)1.6 channels in mature neurons.
Collapse
Affiliation(s)
- Janelle E O'Brien
- Department of Human Genetics, University of Michigan, Ann Arbor, MI 48109-5618, USA
| | | | | | | | | | | |
Collapse
|
29
|
Ion channels and schizophrenia: a gene set-based analytic approach to GWAS data for biological hypothesis testing. Hum Genet 2011; 131:373-91. [PMID: 21866342 DOI: 10.1007/s00439-011-1082-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2011] [Accepted: 08/08/2011] [Indexed: 01/11/2023]
Abstract
Schizophrenia is a complex genetic disorder. Gene set-based analytic (GSA) methods have been widely applied for exploratory analyses of large, high-throughput datasets, but less commonly employed for biological hypothesis testing. Our primary hypothesis is that variation in ion channel genes contribute to the genetic susceptibility to schizophrenia. We applied Exploratory Visual Analysis (EVA), one GSA application, to analyze European-American (EA) and African-American (AA) schizophrenia genome-wide association study datasets for statistical enrichment of ion channel gene sets, comparing GSA results derived under three SNP-to-gene mapping strategies: (1) GENIC; (2) 500-Kb; (3) 2.5-Mb and three complimentary SNP-to-gene statistical reduction methods: (1) minimum p value (pMIN); (2) a novel method, proportion of SNPs per Gene with p values below a pre-defined α-threshold (PROP); and (3) the truncated product method (TPM). In the EA analyses, ion channel gene set(s) were enriched under all mapping and statistical approaches. In the AA analysis, ion channel gene set(s) were significantly enriched under pMIN for all mapping strategies and under PROP for broader mapping strategies. Less extensive enrichment in the AA sample may reflect true ethnic differences in susceptibility, sampling or case ascertainment differences, or higher dimensionality relative to sample size of the AA data. More consistent findings under broader mapping strategies may reflect enhanced power due to increased SNP inclusion, enhanced capture of effects over extended haplotypes or significant contributions from regulatory regions. While extensive pMIN findings may reflect gene size bias, the extent and significance of PROP and TPM findings suggest that common variation at ion channel genes may capture some of the heritability of schizophrenia.
Collapse
|
30
|
Young A, Machacek DW, Dhara SK, Macleish PR, Benveniste M, Dodla MC, Sturkie CD, Stice SL. Ion channels and ionotropic receptors in human embryonic stem cell derived neural progenitors. Neuroscience 2011; 192:793-805. [PMID: 21672611 DOI: 10.1016/j.neuroscience.2011.04.039] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2011] [Revised: 04/14/2011] [Accepted: 04/15/2011] [Indexed: 11/24/2022]
Abstract
Human neural progenitor cells differentiated from human embryonic stem cells offer a potential cell source for studying neurodegenerative diseases and for drug screening assays. Previously, we demonstrated that human neural progenitors could be maintained in a proliferative state with the addition of leukemia inhibitory factor and basic fibroblast growth factor. Here we demonstrate that 96 h after removal of basic fibroblast growth factor the neural progenitor cell culture was significantly altered and cell replication halted. Fourteen days after the removal of basic fibroblast growth factor, most cells expressed microtubule-associated protein 2 and TUJ1, markers characterizing a post-mitotic neuronal phenotype as well as neural developmental markers Cdh2 and Gbx2. Real-time PCR was performed to determine the ionotropic receptor subunit expression profile. Differentiated neural progenitors express subunits of glutamatergic, GABAergic, nicotinic, purinergic and transient receptor potential receptors. In addition, sodium and calcium channel subunits were also expressed. Functionally, virtually all the hNP cells tested under whole-cell voltage clamp exhibited delayed rectifier potassium channel currents and some differentiated cells exhibited tetrodotoxin-sensitive, voltage-dependent sodium channel current. Action potentials could also be elicited by currents injection under whole-cell current clamp in a minority of cells. These results indicate that removing basic fibroblast growth factor from the neural progenitor cell cultures leads to a post-mitotic state, and has the capability to produce excitable cells that can generate action potentials, a landmark characteristic of a neuronal phenotype. This is the first report of an efficient and simple means of generating human neuronal cells for ionotropic receptor assays and ultimately for electrically active human neural cell assays for drug discovery.
Collapse
Affiliation(s)
- A Young
- Regenerative Bioscience Center, 425 River Road Room 450, Athens, GA 30602, USA
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Westphalen RI, Kwak NB, Daniels K, Hemmings HC. Regional differences in the effects of isoflurane on neurotransmitter release. Neuropharmacology 2011; 61:699-706. [PMID: 21651920 DOI: 10.1016/j.neuropharm.2011.05.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2010] [Revised: 02/04/2011] [Accepted: 05/16/2011] [Indexed: 11/18/2022]
Abstract
Stimulus evoked neurotransmitter release requires that Na(+) channel-dependent nerve terminal depolarization be transduced into synaptic vesicle exocytosis. Inhaled anesthetics block presynaptic Na(+) channels and selectively inhibit glutamate over GABA release from isolated nerve terminals, indicating mechanistic differences between excitatory and inhibitory transmitter release. We compared the effects of isoflurane on depolarization-evoked [(3)H]glutamate and [(14)C]GABA release from isolated nerve terminals prepared from four regions of rat CNS evoked by 4-aminopyridine (4AP), veratridine (VTD), or elevated K(+). These mechanistically distinct secretegogues distinguished between Na(+) channel- and/or Ca(2+) channel-mediated presynaptic effects. Isoflurane completely inhibited total 4AP-evoked glutamate release (IC(50) = 0.42 ± 0.03 mM) more potently than GABA release (IC(50) = 0.56 ± 0.02 mM) from cerebral cortex (1.3-fold greater potency), hippocampus and striatum, but inhibited glutamate and GABA release from spinal cord terminals equipotently. Na(+) channel-specific VTD-evoked glutamate release from cortex was also significantly more sensitive to inhibition by isoflurane than was GABA release. Na(+) channel-independent K(+)-evoked release was insensitive to isoflurane at clinical concentrations in all four regions, consistent with a target upstream of Ca(2+) entry. Isoflurane inhibited Na(+) channel-mediated (tetrodotoxin-sensitive) 4AP-evoked glutamate release (IC(50) = 0.30 ± 0.03 mM) more potently than GABA release (IC(50) = 0.67 ± 0.04 mM) from cortex (2.2-fold greater potency). The magnitude of inhibition of Na(+) channel-mediated 4AP-evoked release by a single clinical concentration of isoflurane (0.35 mM) varied by region and transmitter: Inhibition of glutamate release from spinal cord was greater than from the three brain regions and greater than GABA release for each CNS region. These findings indicate that isoflurane selectively inhibits glutamate release compared to GABA release via Na(+) channel-mediated transduction in the four CNS regions tested, and that differences in presynaptic Na(+) channel involvement determine differences in anesthetic pharmacology.
Collapse
Affiliation(s)
- Robert I Westphalen
- Department of Anesthesiology, Weill Cornell Medical College, New York, NY 10065, United States
| | | | | | | |
Collapse
|
32
|
TNF-α contributes to up-regulation of Nav1.3 and Nav1.8 in DRG neurons following motor fiber injury. Pain 2010; 151:266-279. [PMID: 20638792 DOI: 10.1016/j.pain.2010.06.005] [Citation(s) in RCA: 138] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2009] [Revised: 04/07/2010] [Accepted: 06/10/2010] [Indexed: 12/13/2022]
Abstract
A large body of evidence has demonstrated that the ectopic discharges of action potentials in primary afferents, resulted from the abnormal expression of voltage gated sodium channels (VGSCs) in dorsal root ganglion (DRG) neurons following peripheral nerve injury are important for the development of neuropathic pain. However, how nerve injury affects the expression of VGSCs is largely unknown. Here, we reported that selective injury of motor fibers by L5 ventral root transection (L5-VRT) up-regulated Nav1.3 and Nav1.8 at both mRNA and protein level and increased current densities of TTX-S and TTX-R channels in DRG neurons, suggesting that nerve injury may up-regulate functional VGSCs in sensory neurons indirectly. As the up-regulated Nav1.3 and Nav1.8 were highly co-localized with TNF-α, we tested the hypothesis that the increased TNF-α may lead to over-expression of the sodium channels. Indeed, we found that peri-sciatic administration of recombinant rat TNF-α (rrTNF) without any nerve injury, which produced lasting mechanical allodynia, also up-regulated Nav1.3 and Nav1.8 in DRG neurons in vivo and that rrTNF enhanced the expression of Nav1.3 and Nav1.8 in cultured adult rat DRG neurons in a dose-dependent manner. Furthermore, inhibition of TNF-α synthesis, which prevented neuropathic pain, strongly inhibited the up-regulation of Nav1.3 and Nav1.8. The up-regulation of the both channels following L5-VRT was significantly lower in TNF receptor 1 knockout mice than that in wild type mice. These data suggest that increased TNF-α may be responsible for up-regulation of Nav1.3 and Nav1.8 in uninjured DRG neurons following nerve injury.
Collapse
|
33
|
Fast- or slow-inactivated state preference of Na+ channel inhibitors: a simulation and experimental study. PLoS Comput Biol 2010; 6:e1000818. [PMID: 20585544 PMCID: PMC2887460 DOI: 10.1371/journal.pcbi.1000818] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2009] [Accepted: 05/14/2010] [Indexed: 12/02/2022] Open
Abstract
Sodium channels are one of the most intensively studied drug targets. Sodium channel inhibitors (e.g., local anesthetics, anticonvulsants, antiarrhythmics and analgesics) exert their effect by stabilizing an inactivated conformation of the channels. Besides the fast-inactivated conformation, sodium channels have several distinct slow-inactivated conformational states. Stabilization of a slow-inactivated state has been proposed to be advantageous for certain therapeutic applications. Special voltage protocols are used to evoke slow inactivation of sodium channels. It is assumed that efficacy of a drug in these protocols indicates slow-inactivated state preference. We tested this assumption in simulations using four prototypical drug inhibitory mechanisms (fast or slow-inactivated state preference, with either fast or slow binding kinetics) and a kinetic model for sodium channels. Unexpectedly, we found that efficacy in these protocols (e.g., a shift of the “steady-state slow inactivation curve”), was not a reliable indicator of slow-inactivated state preference. Slowly associating fast-inactivated state-preferring drugs were indistinguishable from slow-inactivated state-preferring drugs. On the other hand, fast- and slow-inactivated state-preferring drugs tended to preferentially affect onset and recovery, respectively. The robustness of these observations was verified: i) by performing a Monte Carlo study on the effects of randomly modifying model parameters, ii) by testing the same drugs in a fundamentally different model and iii) by an analysis of the effect of systematically changing drug-specific parameters. In patch clamp electrophysiology experiments we tested five sodium channel inhibitor drugs on native sodium channels of cultured hippocampal neurons. For lidocaine, phenytoin and carbamazepine our data indicate a preference for the fast-inactivated state, while the results for fluoxetine and desipramine are inconclusive. We suggest that conclusions based on voltage protocols that are used to detect slow-inactivated state preference are unreliable and should be re-evaluated. Sodium channels are the key proteins for action potential firing in most excitable cells. Inhibitor drugs prevent excitation (local anesthetics), regulate excitability (antiarrhythmics), or prevent overexcitation (antiepileptic, antispastic and neuroprotective drugs) by binding to the channel and keeping it in one of the inactivated channel conformations. Sodium channels have one fast- and several slow-inactivated conformations (states). The specific stabilization of slow-inactivated states have been proposed to be advantageous in certain therapeutic applications. The question of whether individual drugs stabilize the fast or the slow-inactivated state is studied using specific voltage protocols. We tested the reliability of conclusions based on these protocols in simulation experiments using a model of sodium channels, and we found that fast- and slow-inactivated state-stabilizing drugs could not be differentiated. We suggested a method by which the state preference of at least a subset of individual drugs could be determined and tried the method in electrophysiology experiments with five individual drugs. Three of the drugs (lidocaine, phenytoin and carbamazepine) were classified as fast-inactivated state-stabilizers, while the state preference of fluoxetine and desipramine was found to be undeterminable by this method.
Collapse
|
34
|
Wang Y, Brittain JM, Jarecki BW, Park KD, Wilson SM, Wang B, Hale R, Meroueh SO, Cummins TR, Khanna R. In silico docking and electrophysiological characterization of lacosamide binding sites on collapsin response mediator protein-2 identifies a pocket important in modulating sodium channel slow inactivation. J Biol Chem 2010; 285:25296-307. [PMID: 20538611 DOI: 10.1074/jbc.m110.128801] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The anti-epileptic drug (R)-lacosamide ((2R)-2-(acetylamino)-N-benzyl-3-methoxypropanamide (LCM)) modulates voltage-gated sodium channels (VGSCs) by preferentially interacting with slow inactivated sodium channels, but the observation that LCM binds to collapsin response mediator protein 2 (CRMP-2) suggests additional mechanisms of action for LCM. We postulated that CRMP-2 levels affects the actions of LCM on VGSCs. CRMP-2 labeling by LCM analogs was competitively displaced by excess LCM in rat brain lysates. Manipulation of CRMP-2 levels in the neuronal model system CAD cells affected slow inactivation of VGSCs without any effects on other voltage-dependent properties. In silico docking was performed to identify putative binding sites in CRMP-2 that may modulate the effects of LCM on VGSCs. These studies identified five cavities in CRMP-2 that can accommodate LCM. CRMP-2 alanine mutants of key residues within these cavities were functionally similar to wild-type CRMP-2 as assessed by similar levels of enhancement in dendritic complexity of cortical neurons. Next, we examined the effects of expression of wild-type and mutant CRMP-2 constructs on voltage-sensitive properties of VGSCs in CAD cells: 1) steady-state voltage-dependent activation and fast-inactivation properties were not affected by LCM, 2) CRMP-2 single alanine mutants reduced the LCM-mediated effects on the ability of endogenous Na(+) channels to transition to a slow inactivated state, and 3) a quintuplicate CRMP-2 alanine mutant further decreased this slow inactivated fraction. Collectively, these results identify key CRMP-2 residues that can coordinate LCM binding thus making it more effective on its primary clinical target.
Collapse
Affiliation(s)
- Yuying Wang
- Department of Pharmacology and Toxicology, Paul and Carole Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Yan L, Wang Q, Fu Q, Ye Q, Xiao H, Wan Q. Amitriptyline inhibits currents and decreases the mRNA expression of voltage-gated sodium channels in cultured rat cortical neurons. Brain Res 2010; 1336:1-9. [DOI: 10.1016/j.brainres.2010.04.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2009] [Revised: 02/28/2010] [Accepted: 04/07/2010] [Indexed: 12/19/2022]
|
36
|
Westphalen RI, Yu J, Krivitski M, Jih TY, Hemmings HC. Regional differences in nerve terminal Na+ channel subtype expression and Na+ channel-dependent glutamate and GABA release in rat CNS. J Neurochem 2010; 113:1611-20. [PMID: 20374421 DOI: 10.1111/j.1471-4159.2010.06722.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
We tested the hypothesis that expression of pre-synaptic voltage-gated sodium channel (Na(v)) subtypes coupled to neurotransmitter release differs between transmitter types and CNS regions in a nerve terminal-specific manner. Na(v) coupling to transmitter release was determined by measuring the sensitivity of 4-aminopyridine (4AP)-evoked [(3)H]glutamate and [(14)C]GABA release to the specific Na(v) blocker tetrodotoxin (TTX) for nerve terminals isolated from rat cerebral cortex, hippocampus, striatum and spinal cord. Expression of various Na(v) subtypes was measured by immunoblotting using subtype-specific antibodies. Potencies of TTX for inhibition of glutamate and GABA release varied between CNS regions. However, the efficacies of TTX for inhibition of 4AP-evoked glutamate release were greater than for inhibition of GABA release in all regions except spinal cord. The relative nerve terminal expression of total Na(v) subtypes as well as of specific subtypes varied considerably between CNS regions. The region-specific potencies of TTX for inhibition of 4AP-evoked glutamate release correlated with greater relative expression of total nerve terminal Na(v) and Na(v)1.2. Nerve terminal-specific differences in the expression of specific Na(v) subtypes contribute to transmitter-specific and regional differences in pharmacological sensitivities of transmitter release.
Collapse
Affiliation(s)
- Robert I Westphalen
- Department of Anesthesiology, Weill Cornell Medical College, New York, New York 10065, USA
| | | | | | | | | |
Collapse
|
37
|
Dai A, Temporal S, Schulz DJ. Cell-specific patterns of alternative splicing of voltage-gated ion channels in single identified neurons. Neuroscience 2010; 168:118-29. [PMID: 20211705 DOI: 10.1016/j.neuroscience.2010.03.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2009] [Revised: 02/25/2010] [Accepted: 03/02/2010] [Indexed: 11/28/2022]
Abstract
CbNa(v) and CbIH encode channels that carry voltage-gated sodium and hyperpolarization activated cation currents respectively in the crab, Cancer borealis. We cloned and sequenced full length cDNAs for both CbNa(v) and CbIH and found nine different regions of alternative splicing for the CbNa(v) gene and four regions of alternative splicing for CbIH. We used RT-PCR to determine tissue-specific differences in splicing of both channel genes among cardiac muscle, skeletal muscle, brain, and stomatogastric ganglion (STG) tissue. We then examined the splice variant isoforms present in single, unambiguously identified neurons of the STG. We found cell-type specific patterns of alternative splicing for CbNa(v), indicating unique cell-specific pattern of post-transcriptional modification. Furthermore, we detected possible differences in cellular localization of alternatively spliced CbNa(v) transcripts; distinct mRNA isoforms are present between the cell somata and the axons of the neurons. In contrast, we found no qualitative differences among different cell types for CbIH variants present, although this analysis did not represent the full spectrum of all possible CbIH variants. CbIH mRNA was not detected in axon samples. Finally, although cell-type specific patterns of splicing were detected for CbNa(v), the same cell type within and between animals also displayed variability in which splice forms were detected. These results indicate that channel splicing is differentially regulated at the level of single neurons of the same neural network, providing yet another mechanism by which cell-specific neuronal output can be achieved.
Collapse
Affiliation(s)
- A Dai
- Department of Biological Sciences, University of Missouri, Columbia, MO, USA
| | | | | |
Collapse
|
38
|
Singh NA, Pappas C, Dahle EJ, Claes LRF, Pruess TH, De Jonghe P, Thompson J, Dixon M, Gurnett C, Peiffer A, White HS, Filloux F, Leppert MF. A role of SCN9A in human epilepsies, as a cause of febrile seizures and as a potential modifier of Dravet syndrome. PLoS Genet 2009; 5:e1000649. [PMID: 19763161 PMCID: PMC2730533 DOI: 10.1371/journal.pgen.1000649] [Citation(s) in RCA: 173] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2009] [Accepted: 08/14/2009] [Indexed: 11/18/2022] Open
Abstract
A follow-up study of a large Utah family with significant linkage to chromosome 2q24 led us to identify a new febrile seizure (FS) gene, SCN9A encoding Na(v)1.7. In 21 affected members, we uncovered a potential mutation in a highly conserved amino acid, p.N641Y, in the large cytoplasmic loop between transmembrane domains I and II that was absent from 586 ethnically matched population control chromosomes. To establish a functional role for this mutation in seizure susceptibility, we introduced the orthologous mutation into the murine Scn9a ortholog using targeted homologous recombination. Compared to wild-type mice, homozygous Scn9a(N641Y/N641Y) knockin mice exhibit significantly reduced thresholds to electrically induced clonic and tonic-clonic seizures, and increased corneal kindling acquisition rates. Together, these data strongly support the SCN9A p.N641Y mutation as disease-causing in this family. To confirm the role of SCN9A in FS, we analyzed a collection of 92 unrelated FS patients and identified additional highly conserved Na(v)1.7 missense variants in 5% of the patients. After one of these children with FS later developed Dravet syndrome (severe myoclonic epilepsy of infancy), we sequenced the SCN1A gene, a gene known to be associated with Dravet syndrome, and identified a heterozygous frameshift mutation. Subsequent analysis of 109 Dravet syndrome patients yielded nine Na(v)1.7 missense variants (8% of the patients), all in highly conserved amino acids. Six of these Dravet syndrome patients with SCN9A missense variants also harbored either missense or splice site SCN1A mutations and three had no SCN1A mutations. This study provides evidence for a role of SCN9A in human epilepsies, both as a cause of FS and as a partner with SCN1A mutations.
Collapse
Affiliation(s)
- Nanda A Singh
- Department of Human Genetics, University of Utah, Salt Lake City, Utah, United States of America.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Byers MR, Rafie MM, Westenbroek RE. Dexamethasone effects on Na(v)1.6 in tooth pulp, dental nerves, and alveolar osteoclasts of adult rats. Cell Tissue Res 2009; 338:217-26. [PMID: 19763626 DOI: 10.1007/s00441-009-0842-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2009] [Accepted: 07/07/2009] [Indexed: 02/02/2023]
Abstract
Dexamethasone causes extensive physiologic reactions including the reduction of inflammation and pain. Here, we asked whether it also affected dental or periodontal cells or dental innervation by altering voltage-gated sodium channel Na(v)1.6 immunoreactivity (IR) or neural synaptophysin. Daily dexamethasone (0.2 mg/kg) given for 1 week to rats caused 12-fold increased intensity of Na(v)1.6-IR in dendritic pulpal cells of normal molars and incisors compared with vehicle treatment. These cells also co-localized monocyte (ED-1) or dendritic cell (CD11b/Ox42) markers, and their location in molars expanded during dexamethasone treatment to include deeper pulp. Furthermore, dexamethasone caused a 10-fold decrease in the number of Na(v)1.6-immunoreactive multinucleate osteoclasts along the alveolar bone of molar root sockets. No changes occurred for neural Na(v)1.6 at axonal nodes of Ranvier, even though IR for calcitonin gene-related peptide was greatly decreased, as expected, and neural synaptophysin-IR was decreased 59% by dexamethasone. At 4 days after tooth injury, pulpal vasodilation and increased Na(v)1.6-immunoreactive pulp cells were similar for all groups. Thus, dexamethasone changes dental pulp cell and alveolar osteoclast Na(v)1.6-IR in normal teeth, but different mechanisms occur after tooth injury when tissue reactions were similar for dexamethasone- and vehicle-treated rats. Steroid-induced alterations of dental pain and inflammation coincide with altered exocytic capability in dental nerve fibers as shown by synaptophysin-IR and with altered pulp cell Na(v)1.6-IR and osteoclast number, but not with any changes in Na(v)1.6-IR for nodes of Ranvier in myelinated dental axons.
Collapse
Affiliation(s)
- Margaret R Byers
- Department of Anesthesiology, University of Washington, Box 356540, Seattle, WA 98195-6540, USA.
| | | | | |
Collapse
|
40
|
Xiang H, Wang L, Cui J, Du J, Wang K, Xu A. Effects of recombinant neurotoxins on single Na(+) channels in isolated rat hippocampal neurons. J Biochem Mol Toxicol 2009; 23:244-55. [PMID: 19705351 DOI: 10.1002/jbt.20285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Four recombinant neurotoxins Hk2a, Hk7a, Hk8a, Hk16a, originally from a sea anemone species Anthopleura sp., were obtained by fusion expression of their genes in Escherichia coli. These neurotoxins were composed of 47 amino acid residues, among which the differences were found at positions 14, 22, 25, and 37, respectively. The effects of the four neurotoxins on single-channel current of sodium in rat hippocampal neurons were studied by cell-attached patch clamp. Each neurotoxin 2 microM could modulate the sodium channel by prolonging the opening dwell time and increasing the open probability, but did not change the amplitude of sodium channel currents. Based on the studies of the structure-function relationship, we found that Hk7a displayed the biggest increase of the open probability because His14 (from Arg14) makes its structure seem more compact in comparison with the other three toxins and Ap-A. Phe25 (Hk8a, Hk16a), which varied from Ala25 (Hk2a, Hk7a), showed that phenyl group might interfere with other key amino acid residue to decrease the activity of toxins. Arg37 (from His37) in Hk8a contributed to decrease of open probability. In our work, it was shown that these important amino acid sites might provide a reliable proof for the future pharmaceutical design.
Collapse
Affiliation(s)
- Hui Xiang
- Department of Biological Science and Technology, Sun Yat-sen, Zhongshan University, Guangzhou, Guangdong Province, People's Republic of China.
| | | | | | | | | | | |
Collapse
|
41
|
Wang J, Ou SW, Wang YJ, Kameyama M, Kameyama A, Zong ZH. Analysis of four novel variants of Nav1.5/SCN5A cloned from the brain. Neurosci Res 2009; 64:339-47. [DOI: 10.1016/j.neures.2009.04.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2008] [Revised: 04/05/2009] [Accepted: 04/07/2009] [Indexed: 11/26/2022]
|
42
|
Wang J, Ou SW, Wang YJ, Zong ZH, Lin L, Kameyama M, Kameyama A. New Variants of Nav1.5/SCN5A Encode Na+Channels in the Brain. J Neurogenet 2009; 22:57-75. [DOI: 10.1080/01677060701672077] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
43
|
Gao N, Lu M, Echeverri F, Laita B, Kalabat D, Williams ME, Hevezi P, Zlotnik A, Moyer BD. Voltage-gated sodium channels in taste bud cells. BMC Neurosci 2009; 10:20. [PMID: 19284629 PMCID: PMC2660338 DOI: 10.1186/1471-2202-10-20] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2008] [Accepted: 03/12/2009] [Indexed: 12/19/2022] Open
Abstract
Background Taste bud cells transmit information regarding the contents of food from taste receptors embedded in apical microvilli to gustatory nerve fibers innervating basolateral membranes. In particular, taste cells depolarize, activate voltage-gated sodium channels, and fire action potentials in response to tastants. Initial cell depolarization is attributable to sodium influx through TRPM5 in sweet, bitter, and umami cells and an undetermined cation influx through an ion channel in sour cells expressing PKD2L1, a candidate sour taste receptor. The molecular identity of the voltage-gated sodium channels that sense depolarizing signals and subsequently initiate action potentials coding taste information to gustatory nerve fibers is unknown. Results We describe the molecular and histological expression profiles of cation channels involved in electrical signal transmission from apical to basolateral membrane domains. TRPM5 was positioned immediately beneath tight junctions to receive calcium signals originating from sweet, bitter, and umami receptor activation, while PKD2L1 was positioned at the taste pore. Using mouse taste bud and lingual epithelial cells collected by laser capture microdissection, SCN2A, SCN3A, and SCN9A voltage-gated sodium channel transcripts were expressed in taste tissue. SCN2A, SCN3A, and SCN9A were expressed beneath tight junctions in subsets of taste cells. SCN3A and SCN9A were expressed in TRPM5 cells, while SCN2A was expressed in TRPM5 and PKD2L1 cells. HCN4, a gene previously implicated in sour taste, was expressed in PKD2L1 cells and localized to cell processes beneath the taste pore. Conclusion SCN2A, SCN3A and SCN9A voltage-gated sodium channels are positioned to sense initial depolarizing signals stemming from taste receptor activation and initiate taste cell action potentials. SCN2A, SCN3A and SCN9A gene products likely account for the tetrodotoxin-sensitive sodium currents in taste receptor cells.
Collapse
Affiliation(s)
- Na Gao
- Senomyx, Inc, 4767 Nexus Centre Drive, San Diego, CA 92121, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Gao N, Lu M, Echeverri F, Laita B, Kalabat D, Williams ME, Hevezi P, Zlotnik A, Moyer BD. Voltage-gated sodium channels in taste bud cells. BMC Neurosci 2009. [PMID: 19284629 DOI: 10.1186/1471‐2202‐10‐20] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Taste bud cells transmit information regarding the contents of food from taste receptors embedded in apical microvilli to gustatory nerve fibers innervating basolateral membranes. In particular, taste cells depolarize, activate voltage-gated sodium channels, and fire action potentials in response to tastants. Initial cell depolarization is attributable to sodium influx through TRPM5 in sweet, bitter, and umami cells and an undetermined cation influx through an ion channel in sour cells expressing PKD2L1, a candidate sour taste receptor. The molecular identity of the voltage-gated sodium channels that sense depolarizing signals and subsequently initiate action potentials coding taste information to gustatory nerve fibers is unknown. RESULTS We describe the molecular and histological expression profiles of cation channels involved in electrical signal transmission from apical to basolateral membrane domains. TRPM5 was positioned immediately beneath tight junctions to receive calcium signals originating from sweet, bitter, and umami receptor activation, while PKD2L1 was positioned at the taste pore. Using mouse taste bud and lingual epithelial cells collected by laser capture microdissection, SCN2A, SCN3A, and SCN9A voltage-gated sodium channel transcripts were expressed in taste tissue. SCN2A, SCN3A, and SCN9A were expressed beneath tight junctions in subsets of taste cells. SCN3A and SCN9A were expressed in TRPM5 cells, while SCN2A was expressed in TRPM5 and PKD2L1 cells. HCN4, a gene previously implicated in sour taste, was expressed in PKD2L1 cells and localized to cell processes beneath the taste pore. CONCLUSION SCN2A, SCN3A and SCN9A voltage-gated sodium channels are positioned to sense initial depolarizing signals stemming from taste receptor activation and initiate taste cell action potentials. SCN2A, SCN3A and SCN9A gene products likely account for the tetrodotoxin-sensitive sodium currents in taste receptor cells.
Collapse
Affiliation(s)
- Na Gao
- Senomyx, Inc, 4767 Nexus Centre Drive, San Diego, CA 92121, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Pathways-based analyses of whole-genome association study data in bipolar disorder reveal genes mediating ion channel activity and synaptic neurotransmission. Hum Genet 2008; 125:63-79. [PMID: 19052778 DOI: 10.1007/s00439-008-0600-y] [Citation(s) in RCA: 110] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2008] [Accepted: 11/21/2008] [Indexed: 01/10/2023]
Abstract
Despite known heritability, the complex genetic architecture of bipolar disorder (likely including trait, locus and allelic heterogeneity, as well as genetic interactions) has confounded genetic discovery for many years. Even modern day whole genome association studies (WGAS) using over half a million common SNPs have implicated only a handful of genes at the genomewide level. Temporally coincident with this series of WGAS, a host of pathways-based analyses (PBAs) have emerged as novel computational approaches in the examination of large-scale datasets, but thus far rarely have been applied to WGAS data in psychiatric disorders. Here, we report a series of PBAs conducted using exploratory visual analysis, an analytic and visualization software tool for examining genomic data, to examine results from the National Institutes of Mental Health and Wellcome-Trust Case Control Consortium WGAS in bipolar disorder. Consistent with a host of prior linkage findings, some candidate gene association studies, and recent WGAS, our strongest findings suggest involvement of ion channel structural and regulatory genes, including voltage-gated ion channels and the broader ion channel group that comprises both voltage- and ligand-gated channels. Moreover, we found only modest overlap in the particular genes driving the significance of these gene sets across the analyses. This observation strongly suggests that variation in ion channel genes, as a class of genes, may contribute to the susceptibility of bipolar disorder and that heterogeneity may figure prominently in the genetic architecture of this susceptibility.
Collapse
|
46
|
Wang M, Liu Q, Luo H, Li J, Tang J, Xiao Y, Liang S. Jingzhaotoxin-II, a novel tarantula toxin preferentially targets rat cardiac sodium channel. Biochem Pharmacol 2008; 76:1716-27. [DOI: 10.1016/j.bcp.2008.09.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2008] [Revised: 09/04/2008] [Accepted: 09/05/2008] [Indexed: 11/26/2022]
|
47
|
Tadić A, Baskaya O, Victor A, Lieb K, Höppner W, Dahmen N. Association analysis of SCN9A gene variants with borderline personality disorder. J Psychiatr Res 2008; 43:155-63. [PMID: 18439623 DOI: 10.1016/j.jpsychires.2008.03.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2007] [Revised: 03/06/2008] [Accepted: 03/07/2008] [Indexed: 10/22/2022]
Abstract
Borderline personality disorder (BPD) is a serious psychiatric disorder affecting about 1-2% of the general population. Key features of BPD are emotional instability, strong impulsivity, repeated self-injurious behavior (SIB) and dissociation. In the etiology of BPD and its predominant symptoms, genetic factors have been suggested. The voltage-gated sodium channel Nav1.7 is expressed in sensory neurons and in the hippocampus, a key region of the limbic system probably dysfunctional in BPD and dissociative disorders. The alpha-subunit of Nav1.7 is encoded by the SCN9A gene on chromosome 2 and variations of SCN9A can lead to complete inability to sense pain. The aim of the present study was to test for associations between SCN9A gene variants and BPD as well as BPD-related phenotypes. We genotyped ten tagging single nucleotide polymorphisms (SNPs) within the SCN9A gene in 161 well-defined Caucasian BPD patients and 156 healthy controls. We found no globally significant association of SCN9A markers with BPD at level 5%. However, in the female and in the male subsample, different SCN9A markers and individual haplotypes showed uncorrected p-values<0.05. In addition, p-values<0.05 were observed in the analysis of associations between SCN9A markers and dissociative symptoms. Although our results were largely negative, replication studies in an independent sample are warranted to follow up on the potential role of SCN9A gene variants in BPD and dissociative symptoms, paying special attention to a possible gender different etiology.
Collapse
Affiliation(s)
- André Tadić
- Department of Psychiatry, University of Mainz, Untere Zahlbacher Strasse 8, 55131 Mainz, Germany.
| | | | | | | | | | | |
Collapse
|
48
|
Seda M, Pinto FM, Wray S, Cintado CG, Noheda P, Buschmann H, Candenas L. Functional and molecular characterization of voltage-gated sodium channels in uteri from nonpregnant rats. Biol Reprod 2007; 77:855-63. [PMID: 17671266 DOI: 10.1095/biolreprod.107.063016] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
We investigated the function and expression of voltage-gated Na(+) channels (VGSC) in the uteri of nonpregnant rats using organ bath techniques, intracellular [Ca(2+)] fluorescence measurements, and RT-PCR. In longitudinally arranged whole-tissue uterine strips, veratridine, a VGSC activator, caused the rapid appearance of phasic contractions of irregular frequency and amplitude. After 50-60 min in the continuous presence of veratridine, rhythmic contractions of very regular frequency and slightly increasing amplitude occurred and were sustained for up to 12 h. Both the early and late components of the contractile response to veratridine were inhibited in a concentration-dependent manner by tetrodotoxin (TTX). In small strips dissected from the uterine longitudinal smooth muscle layer and loaded with Fura-2, veratridine also caused rhythmic contractions, accompanied by transient increases in [Ca(2+)](i), which were abolished by treatment with 0.1 microM TTX. Using end-point and real-time quantitative RT-PCR, we detected the presence of the VGSC alpha subunits Scn2a1, Scn3a, Scn5a, and Scn8a in the cDNA from longitudinal muscle. The mRNAs of the auxiliary beta subunits Scbn1b, Scbn2b, Scbn4b, and traces of Scn3b were also present. These data show for the first time that Scn2a1, Scn3a, Scn5a, and Scn8a, as well as all VGSC beta subunits are expressed in the longitudinal smooth muscle layer of the rat myometrium. In addition, our data show that TTX-sensitive VGSC are able to mediate phasic contractions maintained over long periods of time in the uteri of nonpregnant rats.
Collapse
Affiliation(s)
- Marian Seda
- Instituto de Investigaciones Químicas, 41092 Sevilla, Spain
| | | | | | | | | | | | | |
Collapse
|
49
|
Brugeaud A, Travo C, Demêmes D, Lenoir M, Llorens J, Puel JL, Chabbert C. Control of hair cell excitability by vestibular primary sensory neurons. J Neurosci 2007; 27:3503-11. [PMID: 17392466 PMCID: PMC1994966 DOI: 10.1523/jneurosci.5185-06.2007] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
In the rat utricle, synaptic contacts between hair cells and the nerve fibers arising from the vestibular primary neurons form during the first week after birth. During that period, the sodium-based excitability that characterizes neonate utricle sensory cells is switched off. To investigate whether the establishment of synaptic contacts was responsible for the modulation of the hair cell excitability, we used an organotypic culture of rat utricle in which the setting of synapses was prevented. Under this condition, the voltage-gated sodium current and the underlying action potentials persisted in a large proportion of nonafferented hair cells. We then studied whether impairment of nerve terminals in the utricle of adult rats may also affect hair cell excitability. We induced selective and transient damages of afferent terminals using glutamate excitotoxicity in vivo. The efficiency of the excitotoxic injury was attested by selective swellings of the terminals and underlying altered vestibular behavior. Under this condition, the sodium-based excitability transiently recovered in hair cells. These results indicate that the modulation of hair cell excitability depends on the state of the afferent terminals. In adult utricle hair cells, this property may be essential to set the conditions required for restoration of the sensory network after damage. This is achieved via re-expression of a biological process that occurs during synaptogenesis.
Collapse
Affiliation(s)
- Aurore Brugeaud
- Institut National de la Santé et de la Recherche Médicale Unité 583, 34091 Montpellier, France, and
| | - Cécile Travo
- Institut National de la Santé et de la Recherche Médicale Unité 583, 34091 Montpellier, France, and
| | - Danielle Demêmes
- Institut National de la Santé et de la Recherche Médicale Unité 583, 34091 Montpellier, France, and
| | - Marc Lenoir
- Institut National de la Santé et de la Recherche Médicale Unité 583, 34091 Montpellier, France, and
| | - Jordi Llorens
- Departament de Ciencies Fisiologiques II, Universitat de Barcelona, l'Hospitalet de Llobregat, 08907 Barcelona, Spain
| | - Jean-Luc Puel
- Institut National de la Santé et de la Recherche Médicale Unité 583, 34091 Montpellier, France, and
| | - Christian Chabbert
- Institut National de la Santé et de la Recherche Médicale Unité 583, 34091 Montpellier, France, and
| |
Collapse
|
50
|
Loucif AJC, Bonnavion P, Macri B, Golmard JL, Boni C, Melfort M, Leonard G, Lesch KP, Adrien J, Jacquin TD. Gender-dependent regulation of G-protein-gated inwardly rectifying potassium current in dorsal raphe neurons in knock-out mice devoid of the 5-hydroxytryptamine transporter. ACTA ACUST UNITED AC 2007; 66:1475-88. [PMID: 17013926 DOI: 10.1002/neu.20321] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Agonists at G-protein-coupled receptors in neurons of the dorsal raphe nucleus (DRN) of knock-out mice devoid of the serotonin transporter (5-HTT(-/-)) exhibit lower efficacy to inhibit cellular discharge than in wild-type counterparts. Using patch-clamp whole-cell recordings, we found that a G-protein-gated inwardly rectifying potassium (GIRK) current is involved in the inhibition of spike discharge induced by 5-HT1A agonists (5-carboxamidotryptamine (5-CT) and (+/-)-2-dipropylamino-8-hydroxy-1,2,3,4-tetrahydronaphthalene hydrobromide (8-OH-DPAT); 50 nM-30 microM) in both wild-type and 5-HTT(-/-) female and male mice. These effects were mimicked by 5'-guanylyl-imido-diphosphate (Gpp(NH)p; 400 microM) dialysis into cells with differences between genders. The 5-HTT(-/-) knock-out mutation reduced the current density induced by Gpp(NH)p in females but not in males. These data suggest that the decreased response of 5-HT1A receptors to agonists in 5-HTT(-/-) mutants reflects notably alteration in the coupling between G-proteins and GIRK channels in females but not in males. Accordingly, gender differences in central 5-HT neurotransmission appear to depend-at least in part-on sex-related variations in corresponding receptor-G protein signaling mechanisms.
Collapse
Affiliation(s)
- Alexandre Julien Châu Loucif
- UMR 677, INSERM/UPMC, NeuroPsychoPharmacologie, Faculté de Médecine Pierre et Marie Curie, Site Pitié-Salpêtrière, 91 Boulevard de l'Hôpital, 75634 Paris Cedex 13, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|