1
|
Yohn SE, Harvey PD, Brannan SK, Horan WP. The potential of muscarinic M 1 and M 4 receptor activators for the treatment of cognitive impairment associated with schizophrenia. Front Psychiatry 2024; 15:1421554. [PMID: 39483736 PMCID: PMC11525114 DOI: 10.3389/fpsyt.2024.1421554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 09/03/2024] [Indexed: 11/03/2024] Open
Abstract
Cognitive impairment is a core symptom of schizophrenia and a major determinant of poor long-term functional outcomes. Despite considerable efforts, we do not yet have any approved pharmacological treatments for cognitive impairment associated with schizophrenia (CIAS). A combination of advances in pre-clinical research and recent clinical trial findings have led to a resurgence of interest in the cognition-enhancing potential of novel muscarinic acetylcholine receptor (mAChR) agonists in schizophrenia. This article provides an overview of the scientific rationale for targeting M1 and M4 mAChRs. We describe the evolution of neuroscience research on these receptors since early drug discovery efforts focused on the mAChR agonist xanomeline. This work has revealed that M1 and M4 mAChRs are highly expressed in brain regions that are implicated in cognition. The functional significance of M1 and M4 mAChRs has been extensively characterized in animal models via use of selective receptor subtype compounds through neuronal and non-neuronal mechanisms. Recent clinical trials of a dual M1/M4 mAChR agonist show promising, replicable evidence of potential pro-cognitive effects in schizophrenia, with several other mAChR agonists in clinical development.
Collapse
Affiliation(s)
| | - Phillip D. Harvey
- Division of Psychology, University of Miami, Miami, FL, United States
| | | | - William P. Horan
- Bristol Myers Squibb, Princeton, NJ, United States
- Department of Psychiatry & Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
2
|
Früh S, Boudkkazi S, Koppensteiner P, Sereikaite V, Chen LY, Fernandez-Fernandez D, Rem PD, Ulrich D, Schwenk J, Chen Z, Le Monnier E, Fritzius T, Innocenti SM, Besseyrias V, Trovò L, Stawarski M, Argilli E, Sherr EH, van Bon B, Kamsteeg EJ, Iascone M, Pilotta A, Cutrì MR, Azamian MS, Hernández-García A, Lalani SR, Rosenfeld JA, Zhao X, Vogel TP, Ona H, Scott DA, Scheiffele P, Strømgaard K, Tafti M, Gassmann M, Fakler B, Shigemoto R, Bettler B. Monoallelic de novo AJAP1 loss-of-function variants disrupt trans-synaptic control of neurotransmitter release. SCIENCE ADVANCES 2024; 10:eadk5462. [PMID: 38985877 PMCID: PMC11235169 DOI: 10.1126/sciadv.adk5462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 06/04/2024] [Indexed: 07/12/2024]
Abstract
Adherens junction-associated protein 1 (AJAP1) has been implicated in brain diseases; however, a pathogenic mechanism has not been identified. AJAP1 is widely expressed in neurons and binds to γ-aminobutyric acid type B receptors (GBRs), which inhibit neurotransmitter release at most synapses in the brain. Here, we show that AJAP1 is selectively expressed in dendrites and trans-synaptically recruits GBRs to presynaptic sites of neurons expressing AJAP1. We have identified several monoallelic AJAP1 variants in individuals with epilepsy and/or neurodevelopmental disorders. Specifically, we show that the variant p.(W183C) lacks binding to GBRs, resulting in the inability to recruit them. Ultrastructural analysis revealed significantly decreased presynaptic GBR levels in Ajap1-/- and Ajap1W183C/+ mice. Consequently, these mice exhibited reduced GBR-mediated presynaptic inhibition at excitatory and inhibitory synapses, along with impaired synaptic plasticity. Our study reveals that AJAP1 enables the postsynaptic neuron to regulate the level of presynaptic GBR-mediated inhibition, supporting the clinical relevance of loss-of-function AJAP1 variants.
Collapse
Affiliation(s)
- Simon Früh
- Department of Biomedicine, Pharmazentrum, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland
| | - Sami Boudkkazi
- Institute of Physiology II, University of Freiburg, Hermann-Herderstrasse 7, 79104 Freiburg, Germany
| | - Peter Koppensteiner
- Institute of Science and Technology Austria (IST Austria), Klosterneuburg, Austria
| | - Vita Sereikaite
- Center for Biopharmaceuticals, Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Li-Yuan Chen
- Department of Biomedical Sciences, Faculty of Biology and Medicine, University of Lausanne, Rue du Bugnon 7, 1005 Lausanne, Switzerland
| | - Diego Fernandez-Fernandez
- Department of Biomedicine, Pharmazentrum, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland
| | - Pascal D. Rem
- Department of Biomedicine, Pharmazentrum, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland
| | - Daniel Ulrich
- Department of Biomedicine, Pharmazentrum, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland
| | - Jochen Schwenk
- Institute of Physiology II, University of Freiburg, Hermann-Herderstrasse 7, 79104 Freiburg, Germany
| | - Ziyang Chen
- Center for Biopharmaceuticals, Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Elodie Le Monnier
- Institute of Science and Technology Austria (IST Austria), Klosterneuburg, Austria
| | - Thorsten Fritzius
- Department of Biomedicine, Pharmazentrum, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland
| | | | - Valérie Besseyrias
- Department of Biomedicine, Pharmazentrum, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland
| | - Luca Trovò
- Department of Biomedicine, Pharmazentrum, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland
| | - Michal Stawarski
- Department of Biomedicine, Pharmazentrum, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland
| | - Emanuela Argilli
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94158, USA
- Institute of Human Genetics and Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Elliott H. Sherr
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94158, USA
- Institute of Human Genetics and Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Bregje van Bon
- Department of Human Genetics, Radboud University Medical Center, Nijmegen 6525, Netherlands
| | - Erik-Jan Kamsteeg
- Department of Human Genetics, Radboud University Medical Center, Nijmegen 6525, Netherlands
| | - Maria Iascone
- Laboratorio Genetica Medica, ASST Papa Giovanni XXIII, Bergamo, Italy
| | | | | | - Mahshid S. Azamian
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Andrés Hernández-García
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Seema R. Lalani
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jill A. Rosenfeld
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Xiaonan Zhao
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
- Baylor Genetics, Houston, TX 77021, USA
| | - Tiphanie P. Vogel
- Division of Rheumatology, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
- Center for Human Immunobiology, Texas Children's Hospital, Houston, TX 77030, USA
| | - Herda Ona
- Division of Rheumatology, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
- Center for Human Immunobiology, Texas Children's Hospital, Houston, TX 77030, USA
| | - Daryl A. Scott
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Peter Scheiffele
- Biocenter, University of Basel, Spitalstrasse 41, 4056 Basel, Switzerland
| | - Kristian Strømgaard
- Center for Biopharmaceuticals, Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Mehdi Tafti
- Department of Biomedical Sciences, Faculty of Biology and Medicine, University of Lausanne, Rue du Bugnon 7, 1005 Lausanne, Switzerland
| | - Martin Gassmann
- Department of Biomedicine, Pharmazentrum, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland
| | - Bernd Fakler
- Institute of Physiology II, University of Freiburg, Hermann-Herderstrasse 7, 79104 Freiburg, Germany
| | - Ryuichi Shigemoto
- Institute of Science and Technology Austria (IST Austria), Klosterneuburg, Austria
| | - Bernhard Bettler
- Department of Biomedicine, Pharmazentrum, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland
| |
Collapse
|
3
|
Charlick JN, Bozadzhieva D, Butler AS, Wilkinson KA, Marrion NV. A single coiled-coil domain mutation in hIKCa channel subunits disrupts preferential formation of heteromeric hSK1:hIKCa channels. Eur J Neurosci 2024; 59:3-16. [PMID: 38018635 PMCID: PMC10952195 DOI: 10.1111/ejn.16189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 09/22/2023] [Accepted: 10/25/2023] [Indexed: 11/30/2023]
Abstract
The expression of IKCa (SK4) channel subunits overlaps with that of SK channel subunits, and it has been proposed that the two related subunits prefer to co-assemble to form heteromeric hSK1:hIKCa channels. This implicates hSK1:hIKCa heteromers in physiological roles that might have been attributed to activation of SK channels. We have used a mutation approach to confirm formation of heterometric hSK1:hIKCa channels. Introduction of residues within hSK1 that were predicted to impart sensitivity to the hIKCa current blocker TRAM-34 changed the pharmacology of functional heteromers. Heteromeric channels formed between wildtype hIKCa and mutant hSK1 subunits displayed a significantly higher sensitivity and maximum block to addition of TRAM-34 than heteromers formed between wildtype subunits. Heteromer formation was disrupted by a single point mutation within one COOH-terminal coiled-coil domain of the hIKCa channel subunit. This mutation only disrupted the formation of hSK1:hIKCa heteromeric channels, without affecting the formation of homomeric hIKCa channels. Finally, the Ca2+ gating sensitivity of heteromeric hSK1:hIKCa channels was found to be significantly lower than the Ca2+ gating sensitivity of homomeric hIKCa channels. These data confirmed the preferred formation of heteromeric channels that results from COOH-terminal interactions between subunits. The distinct sensitivity of the heteromer to activation by Ca2+ suggests that heteromeric channels fulfil a distinct function within those neurons that express both subunits.
Collapse
Affiliation(s)
- James N. Charlick
- School of Physiology, Pharmacology and NeuroscienceUniversity of BristolBristolUK
| | - Daniella Bozadzhieva
- School of Physiology, Pharmacology and NeuroscienceUniversity of BristolBristolUK
| | - Andrew S. Butler
- School of Physiology, Pharmacology and NeuroscienceUniversity of BristolBristolUK
| | - Kevin A. Wilkinson
- School of Physiology, Pharmacology and NeuroscienceUniversity of BristolBristolUK
| | - Neil V. Marrion
- School of Physiology, Pharmacology and NeuroscienceUniversity of BristolBristolUK
| |
Collapse
|
4
|
Nomura T, Taniguchi S, Wang YZ, Yeh NH, Wilen AP, Castillon CCM, Foote KM, Xu J, Armstrong JN, Savas JN, Swanson GT, Contractor A. A Pathogenic Missense Mutation in Kainate Receptors Elevates Dendritic Excitability and Synaptic Integration through Dysregulation of SK Channels. J Neurosci 2023; 43:7913-7928. [PMID: 37802657 PMCID: PMC10669804 DOI: 10.1523/jneurosci.1259-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 09/12/2023] [Accepted: 09/16/2023] [Indexed: 10/08/2023] Open
Abstract
Numerous rare variants that cause neurodevelopmental disorders (NDDs) occur within genes encoding synaptic proteins, including ionotropic glutamate receptors. However, in many cases, it remains unclear how damaging missense variants affect brain function. We determined the physiological consequences of an NDD causing missense mutation in the GRIK2 kainate receptor (KAR) gene, that results in a single amino acid change p.Ala657Thr in the GluK2 receptor subunit. We engineered this mutation in the mouse Grik2 gene, yielding a GluK2(A657T) mouse, and studied mice of both sexes to determine how hippocampal neuronal function is disrupted. Synaptic KAR currents in hippocampal CA3 pyramidal neurons from heterozygous A657T mice exhibited slow decay kinetics, consistent with incorporation of the mutant subunit into functional receptors. Unexpectedly, CA3 neurons demonstrated elevated action potential spiking because of downregulation of the small-conductance Ca2+ activated K+ channel (SK), which mediates the post-spike afterhyperpolarization. The reduction in SK activity resulted in increased CA3 dendritic excitability, increased EPSP-spike coupling, and lowered the threshold for the induction of LTP of the associational-commissural synapses in CA3 neurons. Pharmacological inhibition of SK channels in WT mice increased dendritic excitability and EPSP-spike coupling, mimicking the phenotype in A657T mice and suggesting a causative role for attenuated SK activity in aberrant excitability observed in the mutant mice. These findings demonstrate that a disease-associated missense mutation in GRIK2 leads to altered signaling through neuronal KARs, pleiotropic effects on neuronal and dendritic excitability, and implicate these processes in neuropathology in patients with genetic NDDs.SIGNIFICANCE STATEMENT Damaging mutations in genes encoding synaptic proteins have been identified in various neurodevelopmental disorders, but the functional consequences at the cellular and circuit level remain elusive. By generating a novel knock-in mutant mouse, this study examined the role of a pathogenic mutation in the GluK2 kainate receptor (KAR) subunit, a subclass of ionotropic glutamate receptors. Analyses of hippocampal CA3 pyramidal neurons determined elevated action potential firing because of an increase in dendritic excitability. Increased dendritic excitability was attributable to reduced activity of a Ca2+ activated K+ channel. These results indicate that a pathogenic KAR mutation results in dysregulation of dendritic K+ channels, which leads to an increase in synaptic integration and backpropagation of action potentials into distal dendrites.
Collapse
Affiliation(s)
- Toshihiro Nomura
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| | - Sakiko Taniguchi
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| | - Yi-Zhi Wang
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| | - Nai-Hsing Yeh
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| | - Anika P Wilen
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| | - Charlotte C M Castillon
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| | - Kendall M Foote
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| | - Jian Xu
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| | - John N Armstrong
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| | - Jeffrey N Savas
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| | - Geoffrey T Swanson
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
- Department of Neurobiology, Weinberg College of Arts and Sciences Northwestern University, Chicago, Illinois 60611
| | - Anis Contractor
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
- Department of Psychiatry and Behavioral Sciences Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
- Department of Neurobiology, Weinberg College of Arts and Sciences Northwestern University, Chicago, Illinois 60611
| |
Collapse
|
5
|
Feng B, Liu H, Mishra I, Duerrschmid C, Gao P, Xu P, Wang C, He Y. Asprosin promotes feeding through SK channel-dependent activation of AgRP neurons. SCIENCE ADVANCES 2023; 9:eabq6718. [PMID: 36812308 PMCID: PMC9946352 DOI: 10.1126/sciadv.abq6718] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 01/20/2023] [Indexed: 05/08/2023]
Abstract
Asprosin, a recently identified adipokine, activates agouti-related peptide (AgRP) neurons in the arcuate nucleus of the hypothalamus (ARH) via binding to protein tyrosine phosphatase receptor δ (Ptprd) to increase food intake. However, the intracellular mechanisms responsible for asprosin/Ptprd-mediated activation of AgRPARH neurons remain unknown. Here, we demonstrate that the small-conductance calcium-activated potassium (SK) channel is required for the stimulatory effects of asprosin/Ptprd on AgRPARH neurons. Specifically, we found that deficiency or elevation of circulating asprosin increased or decreased the SK current in AgRPARH neurons, respectively. AgRPARH-specific deletion of SK3 (an SK channel subtype highly expressed in AgRPARH neurons) blocked asprosin-induced AgRPARH activation and overeating. Furthermore, pharmacological blockade, genetic knockdown, or knockout of Ptprd abolished asprosin's effects on the SK current and AgRPARH neuronal activity. Therefore, our results demonstrated an essential asprosin-Ptprd-SK3 mechanism in asprosin-induced AgRPARH activation and hyperphagia, which is a potential therapeutic target for the treatment of obesity.
Collapse
Affiliation(s)
- Bing Feng
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, USA
| | - Hesong Liu
- USDA-ARS Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Ila Mishra
- Harrington Discovery Institute, Cleveland, OH, USA
| | - Clemens Duerrschmid
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Peiyu Gao
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, USA
| | - Pingwen Xu
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, The University of Illinois at Chicago, Chicago, IL, USA
| | - Chunmei Wang
- USDA-ARS Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Yanlin He
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, USA
| |
Collapse
|
6
|
Wu X, Yan Q, Liu L, Xue X, Yao W, Li X, Li W, Ding S, Xia Y, Zhang D, Zhu F. Domesticated HERV-W env contributes to the activation of the small conductance Ca 2+-activated K + type 2 channels via decreased 5-HT4 receptor in recent-onset schizophrenia. Virol Sin 2023; 38:9-22. [PMID: 36007838 PMCID: PMC10006216 DOI: 10.1016/j.virs.2022.08.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 08/17/2022] [Indexed: 11/25/2022] Open
Abstract
The human endogenous retroviruses type W family envelope (HERV-W env) gene is located on chromosome 7q21-22. Our previous studies show that HERV-W env is elevated in schizophrenia and HERV-W env can increase calcium influx. Additionally, the 5-HTergic system and particularly 5-hydroxytryptamine (5-HT) receptors play a prominent role in the pathogenesis and treatment of schizophrenia. 5-hydroxytryptamine receptor 4 (5-HT4R) agonist can block calcium channels. However, the underlying relationship between HERV-W env and 5-HT4R in the etiology of schizophrenia has not been revealed. Here, we used enzyme-linked immunosorbent assay to detect the concentration of HERV-W env and 5-HT4R in the plasma of patients with schizophrenia and we found that there were decreased levels of 5-HT4R and a negative correlation between 5-HT4R and HERV-W env in schizophrenia. Overexpression of HERV-W env decreased the transcription and protein levels of 5-HT4R but increased small conductance Ca2+-activated K+ type 2 channels (SK2) expression levels. Further studies revealed that HERV-W env could interact with 5-HT4R. Additionally, luciferase assay showed that an essential region (-364 to -176 from the transcription start site) in the SK2 promoter was required for HERV-W env-induced SK2 expression. Importantly, 5-HT4R participated in the regulation of SK2 expression and promoter activity. Electrophysiological recordings suggested that HERV-W env could increase SK2 channel currents and the increase of SK2 currents was inhibited by 5-HT4R. In conclusion, HERV-W env could activate SK2 channels via decreased 5-HT4R, which might exhibit a novel mechanism for HERV-W env to influence neuronal activity in schizophrenia.
Collapse
Affiliation(s)
- Xiulin Wu
- State Key Laboratory of Virology and Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Qiujin Yan
- State Key Laboratory of Virology and Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | | | - Xing Xue
- State Key Laboratory of Virology and Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Wei Yao
- State Key Laboratory of Virology and Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Xuhang Li
- State Key Laboratory of Virology and Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Wenshi Li
- State Key Laboratory of Virology and Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Shuang Ding
- State Key Laboratory of Virology and Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Yaru Xia
- State Key Laboratory of Virology and Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Dongyan Zhang
- State Key Laboratory of Virology and Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Fan Zhu
- State Key Laboratory of Virology and Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China; Hubei Province Key Laboratory of Allergy & Immunology, Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
7
|
Butler AS, Hancox JC, Marrion NV. Preferential formation of human heteromeric SK2:SK3 channels limits homomeric SK channel assembly and function. J Biol Chem 2022; 299:102783. [PMID: 36502918 PMCID: PMC9841042 DOI: 10.1016/j.jbc.2022.102783] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 11/29/2022] [Accepted: 12/04/2022] [Indexed: 12/13/2022] Open
Abstract
Three isoforms of small conductance, calcium-activated potassium (SK) channel subunits have been identified (SK1-3) that exhibit a broad and overlapping tissue distribution. SK channels have been implicated in several disease states including hypertension and atrial fibrillation, but therapeutic targeting of SK channels is hampered by a lack of subtype-selective inhibitors. This is further complicated by studies showing that SK1 and SK2 preferentially form heteromeric channels during co-expression, likely limiting the function of homomeric channels in vivo. Here, we utilized a simplified expression system to investigate functional current produced when human (h) SK2 and hSK3 subunits are co-expressed. When expressed alone, hSK3 subunits were more clearly expressed on the cell surface than hSK2 subunits. hSK3 surface expression was reduced by co-transfection with hSK2. Whole-cell recording showed homomeric hSK3 currents were larger than homomeric hSK2 currents or heteromeric hSK2:hSK3 currents. The smaller amplitude of hSK2:hSK3-mediated current when compared with homomeric hSK3-mediated current suggests hSK2 subunits regulate surface expression of heteromers. Co-expression of hSK2 and hSK3 subunits produced a current that arose from a single population of heteromeric channels as exhibited by an intermediate sensitivity to the inhibitors apamin and UCL1684. Co-expression of the apamin-sensitive hSK2 subunit and a mutant, apamin-insensitive hSK3 subunit [hSK3(H485N)], produced an apamin-sensitive current. Concentration-inhibition relationships were best fit by a monophasic Hill equation, confirming preferential formation of heteromers. These data show that co-expressed hSK2 and hSK3 preferentially form heteromeric channels and suggest that the hSK2 subunit acts as a chaperone, limiting membrane expression of hSK2:hSK3 heteromeric channels.
Collapse
Affiliation(s)
- Andrew S Butler
- School of Physiology, Pharmacology and Neuroscience, Medical Sciences Building, University of Bristol, University Walk, Bristol, United Kingdom
| | - Jules C Hancox
- School of Physiology, Pharmacology and Neuroscience, Medical Sciences Building, University of Bristol, University Walk, Bristol, United Kingdom.
| | - Neil V Marrion
- School of Physiology, Pharmacology and Neuroscience, Medical Sciences Building, University of Bristol, University Walk, Bristol, United Kingdom.
| |
Collapse
|
8
|
Wang J, Wang Y, Du X, Zhang H. Potassium Channel Conductance Is Involved in Phenylephrine-Induced Spontaneous Firing of Serotonergic Neurons in the Dorsal Raphe Nucleus. Front Cell Neurosci 2022; 16:891912. [PMID: 35734219 PMCID: PMC9207280 DOI: 10.3389/fncel.2022.891912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 04/22/2022] [Indexed: 11/13/2022] Open
Abstract
The serotonergic (5-HT) network from the dorsal raphe nucleus (DRN) of the brain has been demonstrated to regulate cognition, emotion, and behaviors, including learning and the sleep-wake cycle. Dysregulation of the activity of 5-HT neurons in the DRN is thought to play an important role in emotional disorders. The activity of 5-HT neurons is regulated by norepinephrine (NE) released from the projection terminals of noradrenergic input from the locus coeruleus (LC) via activation of the α1-adrenoceptor. However, insight into the molecular mechanism underlying this NE-induced regulation of 5-HT neuron activity is not clear. In this study, using the agonist of α1-adrenoceptor phenylephrine (PE), brain slices, and patch clamp, we found that A-type, Kv7/KCNQ, and calcium-activated low-conductance K+ channels (SK) underlie PE-induced spontaneous firing in DRN 5-HT neurons. Using single-cell PCR and immunofluorescence, we also identified the isoforms of these K+ channel families that might contribute to the NE/PE-induced spontaneous firing of DRN 5-HT neurons.
Collapse
Affiliation(s)
- Jing Wang
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, China
- Department of Pharmacochemistry, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Yingzi Wang
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, China
| | - Xiaona Du
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, China
| | - Hailin Zhang
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, China
- *Correspondence: Hailin Zhang,
| |
Collapse
|
9
|
Yamawaki Y, Wada Y, Matsui S, Ohtsuki G. Microglia-triggered hypoexcitability plasticity of pyramidal neurons in the rat medial prefrontal cortex. CURRENT RESEARCH IN NEUROBIOLOGY 2022; 3:100028. [DOI: 10.1016/j.crneur.2022.100028] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 01/14/2022] [Accepted: 01/30/2022] [Indexed: 12/16/2022] Open
|
10
|
Klemz A, Wildner F, Tütüncü E, Gerevich Z. Regulation of Hippocampal Gamma Oscillations by Modulation of Intrinsic Neuronal Excitability. Front Neural Circuits 2022; 15:778022. [PMID: 35177966 PMCID: PMC8845518 DOI: 10.3389/fncir.2021.778022] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 12/21/2021] [Indexed: 11/16/2022] Open
Abstract
Ion channels activated around the subthreshold membrane potential determine the likelihood of neuronal firing in response to synaptic inputs, a process described as intrinsic neuronal excitability. Long-term plasticity of chemical synaptic transmission is traditionally considered the main cellular mechanism of information storage in the brain; however, voltage- and calcium-activated channels modulating the inputs or outputs of neurons are also subjects of plastic changes and play a major role in learning and memory formation. Gamma oscillations are associated with numerous higher cognitive functions such as learning and memory, but our knowledge of their dependence on intrinsic plasticity is by far limited. Here we investigated the roles of potassium and calcium channels activated at near subthreshold membrane potentials in cholinergically induced persistent gamma oscillations measured in the CA3 area of rat hippocampal slices. Among potassium channels, which are responsible for the afterhyperpolarization in CA3 pyramidal cells, we found that blockers of SK (KCa2) and KV7.2/7.3 (KCNQ2/3), but not the BK (KCa1.1) and IK (KCa3.1) channels, increased the power of gamma oscillations. On the contrary, activators of these channels had an attenuating effect without affecting the frequency. Pharmacological blockade of the low voltage-activated T-type calcium channels (CaV3.1–3.3) reduced gamma power and increased the oscillation peak frequency. Enhancement of these channels also inhibited the peak power without altering the frequency of the oscillations. The presented data suggest that voltage- and calcium-activated ion channels involved in intrinsic excitability strongly regulate the power of hippocampal gamma oscillations. Targeting these channels could represent a valuable pharmacological strategy against cognitive impairment.
Collapse
|
11
|
Nashed MG, Waye S, Hasan SMN, Nguyen D, Wiseman M, Zhang J, Lau H, Dinesh OC, Raymond R, Greig IR, Bambico FR, Nobrega JN. Antidepressant activity of pharmacological and genetic deactivation of the small-conductance calcium-activated potassium channel subtype-3. Psychopharmacology (Berl) 2022; 239:253-266. [PMID: 34982171 DOI: 10.1007/s00213-021-06045-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 12/13/2021] [Indexed: 11/25/2022]
Abstract
RATIONALE The voltage-insensitive, small-conductance calcium-activated potassium (SK) channel is a key regulator of neuronal depolarization and is implicated in the pathophysiology of depressive disorders. OBJECTIVE We ascertained whether the SK channel is impaired in the chronic unpredictable stress (CUS) model and whether it can serve as a molecular target of antidepressant action. METHODS We assessed the depressive-like behavioral phenotype of CUS-exposed rats and performed post-mortem SK channel binding and activity-dependent zif268 mRNA analyses on their brains. To begin an assessment of SK channel subtypes involved, we examined the effects of genetic and pharmacological inhibition of the SK3 channel using conditional knockout mice and selective SK3 channel negative allosteric modulators (NAMs). RESULTS We found that [125I]apamin binding to SK channels is increased in the prefrontal cortex and decreased in the hippocampus, an effect that was associated with reciprocal levels of zif268 mRNA transcripts indicating abnormal regional cell activity in this model. We found that genetic and pharmacological manipulations significantly decreased immobility in the forced swim test without altering general locomotor activity, a hallmark of antidepressant-like activity. CONCLUSIONS Taken together, these findings link depression-related neural and behavioral pathophysiology with abnormal SK channel functioning and suggest that this can be reversed by the selective inhibition of SK3 channels.
Collapse
Affiliation(s)
- Mina G Nashed
- Behavioural Neurobiology Laboratory, Research Imaging Center, Centre for Addiction and Mental Health (CAMH), Toronto, ON, M5T 1R8, Canada
| | - Shannon Waye
- Department of Psychology, Memorial University of Newfoundland, St. John's, Newfoundland & Labrador, A1B 3X9, Canada
| | - S M Nageeb Hasan
- Department of Psychology, Memorial University of Newfoundland, St. John's, Newfoundland & Labrador, A1B 3X9, Canada
| | - Diana Nguyen
- Behavioural Neurobiology Laboratory, Research Imaging Center, Centre for Addiction and Mental Health (CAMH), Toronto, ON, M5T 1R8, Canada
| | - Micaela Wiseman
- Behavioural Neurobiology Laboratory, Research Imaging Center, Centre for Addiction and Mental Health (CAMH), Toronto, ON, M5T 1R8, Canada
| | - Jing Zhang
- Behavioural Neurobiology Laboratory, Research Imaging Center, Centre for Addiction and Mental Health (CAMH), Toronto, ON, M5T 1R8, Canada
| | - Harry Lau
- Behavioural Neurobiology Laboratory, Research Imaging Center, Centre for Addiction and Mental Health (CAMH), Toronto, ON, M5T 1R8, Canada
| | - O Chandani Dinesh
- Department of Psychology, Memorial University of Newfoundland, St. John's, Newfoundland & Labrador, A1B 3X9, Canada
| | - Roger Raymond
- Behavioural Neurobiology Laboratory, Research Imaging Center, Centre for Addiction and Mental Health (CAMH), Toronto, ON, M5T 1R8, Canada
| | - Iain R Greig
- Institute of Medical Sciences, University of Aberdeen, King's College, Aberdeen, AB25 2ZD, Scotland
| | - Francis Rodriguez Bambico
- Behavioural Neurobiology Laboratory, Research Imaging Center, Centre for Addiction and Mental Health (CAMH), Toronto, ON, M5T 1R8, Canada. .,Department of Psychology, Memorial University of Newfoundland, St. John's, Newfoundland & Labrador, A1B 3X9, Canada.
| | - José N Nobrega
- Behavioural Neurobiology Laboratory, Research Imaging Center, Centre for Addiction and Mental Health (CAMH), Toronto, ON, M5T 1R8, Canada
| |
Collapse
|
12
|
Proulx É, Power SK, Oliver DK, Sargin D, McLaurin J, Lambe EK. Apamin Improves Prefrontal Nicotinic Impairment in Mouse Model of Alzheimer's Disease. Cereb Cortex 2021; 30:563-574. [PMID: 31188425 DOI: 10.1093/cercor/bhz107] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 04/29/2019] [Accepted: 05/01/2019] [Indexed: 12/11/2022] Open
Abstract
Disruption of attention is an early and disabling symptom of Alzheimer's disease (AD). The underlying cellular mechanisms are poorly understood and treatment options for patients are limited. These early attention deficits are evident in the TgCRND8 mouse, a well-established murine model of AD that recapitulates several features of the disease. Here, we report severe impairment of the nicotinic receptor-mediated excitation of prefrontal attentional circuitry in TgCRND8 mice relative to wild-type littermate controls. We demonstrate that this impairment can be remedied by apamin, a bee venom neurotoxin peptide that acts as a selective antagonist to the SK family of calcium-sensitive potassium channels. We probe this seeming upregulation of calcium-sensitive inhibition and find that the attenuated nicotinic firing rates in TgCRND8 attention circuits are mediated neither by greater cellular calcium signals nor by elevated SK channel expression. Instead, we find that TgCRND8 mice show enhanced functional coupling of nicotinic calcium signals to inhibition. This SK-mediated inhibition exerts a powerful negative feedback on nicotinic excitation, dampening attention-relevant signaling in the TgCRND8 brain. These mechanistic findings identify a new cellular target involved in the modulation of attention and a novel therapeutic target for early attention deficits in AD.
Collapse
Affiliation(s)
- É Proulx
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada, M5S 1A8
| | - S K Power
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada, M5S 1A8
| | - D K Oliver
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada, M5S 1A8
| | - D Sargin
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada, M5S 1A8
| | - J McLaurin
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada M5S 1A8.,Biological Sciences and Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Toronto, Ontario, Canada M4N 3M5
| | - E K Lambe
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada, M5S 1A8.,Department of Obstetrics and Gynaecology, University of Toronto, Toronto, Ontario, Canada M5G 1E2.,Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada M5T 1R8
| |
Collapse
|
13
|
Dwivedi D, Bhalla US. Physiology and Therapeutic Potential of SK, H, and M Medium AfterHyperPolarization Ion Channels. Front Mol Neurosci 2021; 14:658435. [PMID: 34149352 PMCID: PMC8209339 DOI: 10.3389/fnmol.2021.658435] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 04/13/2021] [Indexed: 12/19/2022] Open
Abstract
SK, HCN, and M channels are medium afterhyperpolarization (mAHP)-mediating ion channels. The three channels co-express in various brain regions, and their collective action strongly influences cellular excitability. However, significant diversity exists in the expression of channel isoforms in distinct brain regions and various subcellular compartments, which contributes to an equally diverse set of specific neuronal functions. The current review emphasizes the collective behavior of the three classes of mAHP channels and discusses how these channels function together although they play specialized roles. We discuss the biophysical properties of these channels, signaling pathways that influence the activity of the three mAHP channels, various chemical modulators that alter channel activity and their therapeutic potential in treating various neurological anomalies. Additionally, we discuss the role of mAHP channels in the pathophysiology of various neurological diseases and how their modulation can alleviate some of the symptoms.
Collapse
Affiliation(s)
- Deepanjali Dwivedi
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, GKVK Campus, Bengaluru, India.,Department of Neurobiology, Harvard Medical School, Boston, MA, United States.,Stanley Center at the Broad, Cambridge, MA, United States
| | - Upinder S Bhalla
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, GKVK Campus, Bengaluru, India
| |
Collapse
|
14
|
Nikitin ES, Vinogradova LV. Potassium channels as prominent targets and tools for the treatment of epilepsy. Expert Opin Ther Targets 2021; 25:223-235. [PMID: 33754930 DOI: 10.1080/14728222.2021.1908263] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
INTRODUCTION K+ channels are of great interest to epilepsy research as mutations in their genes are found in humans with inherited epilepsy. At the level of cellular physiology, K+ channels control neuronal intrinsic excitability and are the main contributors to membrane repolarization of active neurons. Recently, a genetically modified voltage-dependent K+ channel has been patented as a remedy for epileptic seizures. AREAS COVERED We review the role of potassium channels in excitability, clinical and experimental evidence for the association of potassium channelopathies with epilepsy, the targeting of K+ channels by drugs, and perspectives of gene therapy in epilepsy with the expression of extra K+ channels in the brain. EXPERT OPINION Control over K+ conductance is of great potential benefit for the treatment of epilepsy. Nowadays, gene therapy affecting K+ channels is one of the most promising approaches to treat pharmacoresistant focal epilepsy.
Collapse
Affiliation(s)
- E S Nikitin
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, Moscow, Russia
| | - L V Vinogradova
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
15
|
Kadam M, Perveen S, Kushwah N, Prasad D, Panjwani U, Kumar B, Khan N. Elucidating the role of hypoxia/reoxygenation in hippocampus-dependent memory impairment: do SK channels play role? Exp Brain Res 2021; 239:1747-1763. [PMID: 33779792 DOI: 10.1007/s00221-021-06095-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 03/19/2021] [Indexed: 10/21/2022]
Abstract
Professionals and mountaineers often face the problem of reperfusion injury due to re-oxygenation, upon their return to sea-level after sojourn at high altitude. Small conductance calcium-activated potassium channels (SK channels) have a role in regulating hippocampal synaptic plasticity. However, the role of SK channels under hypoxia-reoxygenation (H/R) is unknown. The present study hypothesized that SK channels play a significant role in H/R induced cognitive dysfunction. Sprague-Dawley rats were exposed to simulated HH (25,000 ft) continuously for 7 days followed by reoxygenation periods 3, 6, 24, 48, 72 and 120 h. It was observed that H/R exposure caused impairment in spatial memory as indicated by increased latency (p < 0.001) and pathlength (p < 0.001). The SK1 channel expression increased upon HH exposure (102.89 ± 7.055), which abrogated upon reoxygenation. HH exposure results in an increase in SK2 (CA3, 297.67 ± 6.69) and SK3 (CA1, 246 ± 5.13) channels which continued to increase gradually upon reoxygenation. The number of pyknotic cells (24 ± 2.03) (p < 0.01) and the expression of caspase-3 increased with HH exposure, which continued in the reoxygenation group (177.795 ± 1.264). Similar pattern was observed in lipid peroxidation (p < 0.001), LDH activity (p < 0.001) and ROS production (p < 0.001). A positive correlation of memory, cell death and oxidative stress indicates that H/R exposure increases oxidative stress coupled with SK channel expression, which may play a role in H/R-induced cognitive decline and neurodegeneration.
Collapse
Affiliation(s)
- Manisha Kadam
- Neurobiology Division, Defence Institute of Physiology and Allied Sciences (DIPAS), Defence Research and Developmental Organization (DRDO), Lucknow Road, Timarpur, Delhi, 110054, India
| | - Saba Perveen
- Neurobiology Division, Defence Institute of Physiology and Allied Sciences (DIPAS), Defence Research and Developmental Organization (DRDO), Lucknow Road, Timarpur, Delhi, 110054, India
| | - Neetu Kushwah
- Neurobiology Division, Defence Institute of Physiology and Allied Sciences (DIPAS), Defence Research and Developmental Organization (DRDO), Lucknow Road, Timarpur, Delhi, 110054, India
| | - Dipti Prasad
- Neurobiology Division, Defence Institute of Physiology and Allied Sciences (DIPAS), Defence Research and Developmental Organization (DRDO), Lucknow Road, Timarpur, Delhi, 110054, India
| | - Usha Panjwani
- Neurobiology Division, Defence Institute of Physiology and Allied Sciences (DIPAS), Defence Research and Developmental Organization (DRDO), Lucknow Road, Timarpur, Delhi, 110054, India
| | - Bhuvnesh Kumar
- Neurobiology Division, Defence Institute of Physiology and Allied Sciences (DIPAS), Defence Research and Developmental Organization (DRDO), Lucknow Road, Timarpur, Delhi, 110054, India
| | - Nilofar Khan
- Neurobiology Division, Defence Institute of Physiology and Allied Sciences (DIPAS), Defence Research and Developmental Organization (DRDO), Lucknow Road, Timarpur, Delhi, 110054, India.
| |
Collapse
|
16
|
Mishra P, Narayanan R. Ion-channel regulation of response decorrelation in a heterogeneous multi-scale model of the dentate gyrus. CURRENT RESEARCH IN NEUROBIOLOGY 2021; 2:100007. [PMID: 33997798 PMCID: PMC7610774 DOI: 10.1016/j.crneur.2021.100007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Heterogeneities in biological neural circuits manifest in afferent connectivity as well as in local-circuit components such as neuronal excitability, neural structure and local synaptic strengths. The expression of adult neurogenesis in the dentate gyrus (DG) amplifies local-circuit heterogeneities and guides heterogeneities in afferent connectivity. How do neurons and their networks endowed with these distinct forms of heterogeneities respond to perturbations to individual ion channels, which are known to change under several physiological and pathophysiological conditions? We sequentially traversed the ion channels-neurons-network scales and assessed the impact of eliminating individual ion channels on conductance-based neuronal and network models endowed with disparate local-circuit and afferent heterogeneities. We found that many ion channels differentially contributed to specific neuronal or network measurements, and the elimination of any given ion channel altered several functional measurements. We then quantified the impact of ion-channel elimination on response decorrelation, a well-established metric to assess the ability of neurons in a network to convey complementary information, in DG networks endowed with different forms of heterogeneities. Notably, we found that networks constructed with structurally immature neurons exhibited functional robustness, manifesting as minimal changes in response decorrelation in the face of ion-channel elimination. Importantly, the average change in output correlation was dependent on the eliminated ion channel but invariant to input correlation. Our analyses suggest that neurogenesis-driven structural heterogeneities could assist the DG network in providing functional resilience to molecular perturbations. Perturbations at one scale result in a cascading impact on physiology across scales. Heterogeneous multi-scale models used to assess the impact of ion-channel deletion. Mapping of structural components to functional outcomes is many-to-many. Differential & variable impact of ion channel deletion on response decorrelation. Neurogenesis-induced structural heterogeneity confers resilience to perturbations.
Collapse
Affiliation(s)
- Poonam Mishra
- Cellular Neurophysiology Laboratory, Molecular Biophysics Unit, Indian Institute of Science, Bangalore 560012, India
| | - Rishikesh Narayanan
- Cellular Neurophysiology Laboratory, Molecular Biophysics Unit, Indian Institute of Science, Bangalore 560012, India
| |
Collapse
|
17
|
Sun J, Liu Y, Baudry M, Bi X. SK2 channel regulation of neuronal excitability, synaptic transmission, and brain rhythmic activity in health and diseases. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2020; 1867:118834. [PMID: 32860835 PMCID: PMC7541745 DOI: 10.1016/j.bbamcr.2020.118834] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 08/13/2020] [Accepted: 08/19/2020] [Indexed: 11/20/2022]
Abstract
Small conductance calcium-activated potassium channels (SKs) are solely activated by intracellular Ca2+ and their activation leads to potassium efflux, thereby repolarizing/hyperpolarizing membrane potential. Thus, these channels play a critical role in synaptic transmission, and consequently in information transmission along the neuronal circuits expressing them. SKs are widely but not homogeneously distributed in the central nervous system (CNS). Activation of SKs requires submicromolar cytoplasmic Ca2+ concentrations, which are reached following either Ca2+ release from intracellular Ca2+ stores or influx through Ca2+ permeable membrane channels. Both Ca2+ sensitivity and synaptic levels of SKs are regulated by protein kinases and phosphatases, and degradation pathways. SKs in turn control the activity of multiple Ca2+ channels. They are therefore critically involved in coordinating diverse Ca2+ signaling pathways and controlling Ca2+ signal amplitude and duration. This review highlights recent advances in our understanding of the regulation of SK2 channels and of their roles in normal brain functions, including synaptic plasticity, learning and memory, and rhythmic activities. It will also discuss how alterations in their expression and regulation might contribute to various brain disorders such as Angelman Syndrome, Alzheimer's disease and Parkinson's disease.
Collapse
Affiliation(s)
- Jiandong Sun
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, United States of America; Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766, United States of America
| | - Yan Liu
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, United States of America; Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766, United States of America
| | - Michel Baudry
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, United States of America; Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766, United States of America
| | - Xiaoning Bi
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, United States of America; Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766, United States of America.
| |
Collapse
|
18
|
Kessi M, Chen B, Peng J, Tang Y, Olatoutou E, He F, Yang L, Yin F. Intellectual Disability and Potassium Channelopathies: A Systematic Review. Front Genet 2020; 11:614. [PMID: 32655623 PMCID: PMC7324798 DOI: 10.3389/fgene.2020.00614] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 05/20/2020] [Indexed: 01/15/2023] Open
Abstract
Intellectual disability (ID) manifests prior to adulthood as severe limitations to intellectual function and adaptive behavior. The role of potassium channelopathies in ID is poorly understood. Therefore, we aimed to evaluate the relationship between ID and potassium channelopathies. We hypothesized that potassium channelopathies are strongly associated with ID initiation, and that both gain- and loss-of-function mutations lead to ID. This systematic review explores the burden of potassium channelopathies, possible mechanisms, advancements using animal models, therapies, and existing gaps. The literature search encompassed both PubMed and Embase up to October 2019. A total of 75 articles describing 338 cases were included in this review. Nineteen channelopathies were identified, affecting the following genes: KCNMA1, KCNN3, KCNT1, KCNT2, KCNJ10, KCNJ6, KCNJ11, KCNA2, KCNA4, KCND3, KCNH1, KCNQ2, KCNAB1, KCNQ3, KCNQ5, KCNC1, KCNB1, KCNC3, and KCTD3. Twelve of these genes presented both gain- and loss-of-function properties, three displayed gain-of-function only, three exhibited loss-of-function only, and one had unknown function. How gain- and loss-of-function mutations can both lead to ID remains largely unknown. We identified only a few animal studies that focused on the mechanisms of ID in relation to potassium channelopathies and some of the few available therapeutic options (channel openers or blockers) appear to offer limited efficacy. In conclusion, potassium channelopathies contribute to the initiation of ID in several instances and this review provides a comprehensive overview of which molecular players are involved in some of the most prominent disease phenotypes.
Collapse
Affiliation(s)
- Miriam Kessi
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China.,Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China.,Kilimanjaro Christian Medical University College, Moshi, Tanzania.,Mawenzi Regional Referral Hospital, Moshi, Tanzania
| | - Baiyu Chen
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China.,Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
| | - Jing Peng
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China.,Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
| | - Yulin Tang
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China.,Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
| | - Eleonore Olatoutou
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China.,Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
| | - Fang He
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China.,Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
| | - Lifen Yang
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China.,Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
| | - Fei Yin
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China.,Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
| |
Collapse
|
19
|
PKA and Ube3a regulate SK2 channel trafficking to promote synaptic plasticity in hippocampus: Implications for Angelman Syndrome. Sci Rep 2020; 10:9824. [PMID: 32555345 PMCID: PMC7299966 DOI: 10.1038/s41598-020-66790-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 05/04/2020] [Indexed: 12/29/2022] Open
Abstract
The ubiquitin ligase, Ube3a, plays important roles in brain development and functions, since its deficiency results in Angelman Syndrome (AS) while its over-expression increases the risk for autism. We previously showed that the lack of Ube3a-mediated ubiquitination of the Ca2+-activated small conductance potassium channel, SK2, contributes to impairment of synaptic plasticity and learning in AS mice. Synaptic SK2 levels are also regulated by protein kinase A (PKA), which phosphorylates SK2 in its C-terminal domain, facilitating its endocytosis. Here, we report that PKA activation restores theta burst stimulation (TBS)-induced long-term potentiation (LTP) in hippocampal slices from AS mice by enhancing SK2 internalization. While TBS-induced SK2 endocytosis is facilitated by PKA activation, SK2 recycling to synaptic membranes after TBS is inhibited by Ube3a. Molecular and cellular studies confirmed that phosphorylation of SK2 in the C-terminal domain increases its ubiquitination and endocytosis. Finally, PKA activation increases SK2 phosphorylation and ubiquitination in Ube3a-overexpressing mice. Our results indicate that, although both Ube3a-mediated ubiquitination and PKA-induced phosphorylation reduce synaptic SK2 levels, phosphorylation is mainly involved in TBS-induced endocytosis, while ubiquitination predominantly inhibits SK2 recycling. Understanding the complex interactions between PKA and Ube3a in the regulation of SK2 synaptic levels might provide new platforms for developing treatments for AS and various forms of autism.
Collapse
|
20
|
Gu H, Han SM, Park KK. Therapeutic Effects of Apamin as a Bee Venom Component for Non-Neoplastic Disease. Toxins (Basel) 2020; 12:E195. [PMID: 32204567 PMCID: PMC7150898 DOI: 10.3390/toxins12030195] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 03/15/2020] [Accepted: 03/17/2020] [Indexed: 02/06/2023] Open
Abstract
Bee venom is a natural toxin produced by honeybees and plays an important role in defending bee colonies. Bee venom has several kinds of peptides, including melittin, apamin, adolapamine, and mast cell degranulation peptides. Apamin accounts for about 2%-3% dry weight of bee venom and is a peptide neurotoxin that contains 18 amino acid residues that are tightly crosslinked by two disulfide bonds. It is well known for its pharmacological functions, which irreversibly block Ca2+-activated K+ (SK) channels. Apamin regulates gene expression in various signal transduction pathways involved in cell development. The aim of this study was to review the current understanding of apamin in the treatment of apoptosis, fibrosis, and central nervous system diseases, which are the pathological processes of various diseases. Apamin's potential therapeutic and pharmacological applications are also discussed.
Collapse
Affiliation(s)
- Hyemin Gu
- Department of Pathology, College of Medicine, Catholic University of Daegu, Daegu 42472, Korea;
| | - Sang Mi Han
- National Academy of Agricultural Science, Jeonjusi, Jeonbuk 54875, Korea;
| | - Kwan-Kyu Park
- Department of Pathology, College of Medicine, Catholic University of Daegu, Daegu 42472, Korea;
| |
Collapse
|
21
|
Nimitvilai S, Lopez MF, Woodward JJ. Sex-dependent differences in ethanol inhibition of mouse lateral orbitofrontal cortex neurons. Addict Biol 2020; 25:e12698. [PMID: 30468275 DOI: 10.1111/adb.12698] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 09/21/2018] [Accepted: 10/23/2018] [Indexed: 01/27/2023]
Abstract
Biological differences between males and females likely influence responses to alcohol and the propensity to engage in excessive drinking. In both humans and rodents, females escalate alcohol use and develop addiction-like behaviors faster than males, while males exhibit more severe withdrawal symptoms during abstinence. The mechanisms underlying these differences are not yet known but may reflect fundamental differences in the ethanol sensitivity of neurons in reward and control areas of the brain. To address this question, we recorded current-evoked spiking of lateral orbitofrontal cortex (lOFC) neurons in male and female C57BL/6J mice following acute and chronic exposure to ethanol. Ethanol (11-66 mM) reduced firing of lOFC neurons but produced less inhibition in neurons from female mice. As previously reported for male mice, the glycine receptor blocker strychnine blocked ethanol inhibition of spiking of lOFC neurons from female mice and prevented the ethanol-induced increase in tonic current. Following chronic intermittent ethanol (CIE) exposure, current-evoked spiking of lOFC neurons was significantly enhanced with a greater effect observed in males. After CIE treatment, acute ethanol had no effect on spiking in neurons from male mice, while it produced a slight but significant decrease in firing in females. Finally, like male mice, the inhibitory effect of the glycine transport inhibitor sarcosine was blunted in CIE-exposed female mice. Together, these results suggest that while lOFC neurons in male and female mice are similarly affected by ethanol, there are significant differences in sensitivity that may contribute to differences in alcohol actions between males and females.
Collapse
Affiliation(s)
- Sudarat Nimitvilai
- Department of NeuroscienceMedical University of South Carolina Charleston South Carolina
| | - Marcelo F. Lopez
- Department of Psychiatry and Behavioral SciencesMedical University of South Carolina Charleston South Carolina
| | - John J. Woodward
- Department of NeuroscienceMedical University of South Carolina Charleston South Carolina
- Department of Psychiatry and Behavioral SciencesMedical University of South Carolina Charleston South Carolina
| |
Collapse
|
22
|
Al Dera H, Alassiri M, Eleawa SM, AlKhateeb MA, Hussein AM, Dallak M, Sakr HF, Alqahtani S, Khalil MA. Melatonin Improves Memory Deficits in Rats with Cerebral Hypoperfusion, Possibly, Through Decreasing the Expression of Small-Conductance Ca 2+-Activated K + Channels. Neurochem Res 2019; 44:1851-1868. [PMID: 31187398 DOI: 10.1007/s11064-019-02820-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Revised: 05/02/2019] [Accepted: 05/27/2019] [Indexed: 12/18/2022]
Abstract
This study investigated the expression pattern, regulation of expression, and the role of hippocampal small-conductance Ca2+-activated K+ (SK) channels in memory deficits after cerebral hypoperfusion (CHP) with or without melatonin treatment, in rats. Adults male Wistar rats (n = 20/group) were divided into (1) a sham (2) a sham + melatonin (3) a two-vessel occlusion (2-VO) model, and (4) a 2-VO + melatonin. Melatonin was administered (i.p.) to all rats at a daily dose of 10 mg kg-1 for 7 days starting at the time of 2-VO-induction. In contrast to 2-VO rats, melatonin increased the latency of the passive avoidance learning test and decreased time to find the hidden platform in Water Morris Test in all tested rats. In addition, it concomitantly downregulated SK1, SK2, and SK3 channels, downregulated mRNA levels of TNFα and IL-1β, enhanced BDNF levels and activity of PKA levels, and restored the levels of cholinergic markers in the hippocampi of the treated-rats. Mechanistically, melatonin significantly prevented CHP-induced activation of ERK1/2, JNK, and P38 MAPK at least by inhibiting ROS generation and enhancing the total antioxidant potential. In cultured hypoxic hippocampal neurons, individual blockage of MAPK signaling by the MEK1/2 inhibitor (U0126), but not by the P38 inhibitor (SB203580) or JNK inhibitor (SP600125), completely prevented the upregulation of all three kinds of SK channels. These data clearly confirm that upregulation of SK channels plays a role in CHP-induced memory loss and indicate that melatonin reverses memory deficits after CHP in rats, at least by, downregulation of SK1, SK2, and SK3 channels in their hippocampi.
Collapse
Affiliation(s)
- Hussain Al Dera
- Department of Basic Medical Sciences, College of Medicine At King Saud, Abdulaziz University for Health Sciences (KSAU-HS), Riyadh, Kingdom of Saudi Arabia. .,King Abdullah International Medical Research Center (KAIMRC), Riyadh, Kingdom of Saudi Arabia.
| | - Mohammed Alassiri
- Department of Basic Medical Sciences, College of Medicine At King Saud, Abdulaziz University for Health Sciences (KSAU-HS), Riyadh, Kingdom of Saudi Arabia.,King Abdullah International Medical Research Center (KAIMRC), Riyadh, Kingdom of Saudi Arabia
| | - Samy M Eleawa
- Department of Applied Medical Sciences, College of Health Sciences, Dept., PAAET, Adailiyah, Kuwait
| | - Mahmoud A AlKhateeb
- Department of Basic Medical Sciences, College of Medicine At King Saud, Abdulaziz University for Health Sciences (KSAU-HS), Riyadh, Kingdom of Saudi Arabia
| | - Abdelaziz M Hussein
- Department of Medical Physiology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Mohammad Dallak
- Department of Medical Physiology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Hussein F Sakr
- Department of Medical Physiology, Faculty of Medicine, Mansoura University, Mansoura, Egypt.,Department of Medical Physiology, College of Medicine, King Khalid University, Abha, Kingdom of Saudi Arabia
| | - Sultan Alqahtani
- Department of Basic Medical Sciences, College of Medicine At King Saud, Abdulaziz University for Health Sciences (KSAU-HS), Riyadh, Kingdom of Saudi Arabia.,King Abdullah International Medical Research Center (KAIMRC), Riyadh, Kingdom of Saudi Arabia
| | - Mohammad A Khalil
- Department of Basic Medical Sciences, College of Medicine, King Fahid Medical City, Riyadh, Kingdom of Saudi Arabia
| |
Collapse
|
23
|
Autuori E, Sedlak P, Xu L, C Ridder M, Tedoldi A, Sah P. rSK1 in Rat Neurons: A Controller of Membrane rSK2? Front Neural Circuits 2019; 13:21. [PMID: 31001092 PMCID: PMC6456674 DOI: 10.3389/fncir.2019.00021] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 03/11/2019] [Indexed: 11/13/2022] Open
Abstract
In mammalian neurons, small conductance calcium-activated potassium channels (SK channels) are activated by calcium influx and contribute to the afterhyperpolarization (AHP) that follows action potentials. Three types of SK channel, SK1, SK2 and SK3 are recognized and encoded by separate genes that are widely expressed in overlapping distributions in the mammalian brain. Expression of the rat genes, rSK2 and rSK3 generates functional ion channels that traffic to the membrane as homomeric and heteromeric complexes. However, rSK1 is not trafficked to the plasma membrane, appears not to form functional channels, and the role of rSK1 in neurons is not clear. Here, we show that rSK1 co-assembles with rSK2. rSK1 is not trafficked to the membrane but is retained in a cytoplasmic compartment. When rSK2 is present, heteromeric rSK1-rSK2 channels are also retained in the cytosolic compartment, reducing the total SK channel content on the plasma membrane. Thus, rSK1 appears to act as chaperone for rSK2 channels and expression of rSK1 may control the level of functional SK current in rat neurons.
Collapse
Affiliation(s)
- Eleonora Autuori
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Petra Sedlak
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Li Xu
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Margreet C Ridder
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Angelo Tedoldi
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Pankaj Sah
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
24
|
Qu L, Wang Y, Ge SN, Li N, Fu J, Zhang Y, Wang X, Jing JP, Li Y, Wang Q, Gao GD, He SM, Wang XL. Altered Activity of SK Channel Underpins Morphine Withdrawal Relevant Psychiatric Deficiency in Infralimbic to Accumbens Shell Pathway. Front Psychiatry 2019; 10:240. [PMID: 31031665 PMCID: PMC6470400 DOI: 10.3389/fpsyt.2019.00240] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 03/29/2019] [Indexed: 12/17/2022] Open
Abstract
Drug addiction can be viewed as a chronic psychiatric disorder that is related to dysfunction of neural circuits, including reward deficits, stress surfeits, craving changes, and compromised executive function. The nucleus accumbens (NAc) plays a crucial role in regulating craving and relapse, while the medial prefrontal cortex (mPFC) represents a higher cortex projecting into the NAc that is active in the management of executive function. In this study, we investigated the role of the small conductance calcium-activated potassium channels (SK channels) in NAc and mPFC after morphine withdrawal. Action potential (AP) firing of neurons in the NAc shell was enhanced via the downregulations of the SK channels after morphine withdrawal. Furthermore, the expression of SK2 and SK3 subunits in the NAc was significantly reduced after 3 weeks of morphine withdrawal, but was not altered in the dorsal striatum. In mPFC, the SK channel subunits were differentially expressed. To be specific, the expression of SK3 was upregulated, while the expression of SK2 was unchanged. Furthermore, the AP firing in layer 5 pyramidal neurons of the infralimbic (IL) cortex was decreased via the upregulations of the SK channel-related tail current after 3 weeks of morphine withdrawal. These results suggest that the SK channel plays a specific role in reward circuits following morphine exposure and a period of drug withdrawal, making it a potential target for the prevention of relapse.
Collapse
Affiliation(s)
- Liang Qu
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Yuan Wang
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Shun-Nan Ge
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Nan Li
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Jian Fu
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Yue Zhang
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Xin Wang
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Jiang-Peng Jing
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Yang Li
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Qiang Wang
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Guo-Dong Gao
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Shi-Ming He
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Xue-Lian Wang
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| |
Collapse
|
25
|
Luján R, Aguado C, Ciruela F, Arus XM, Martín-Belmonte A, Alfaro-Ruiz R, Martínez-Gómez J, de la Ossa L, Watanabe M, Adelman JP, Shigemoto R, Fukazawa Y. SK2 Channels Associate With mGlu 1α Receptors and Ca V2.1 Channels in Purkinje Cells. Front Cell Neurosci 2018; 12:311. [PMID: 30283304 PMCID: PMC6156379 DOI: 10.3389/fncel.2018.00311] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 08/28/2018] [Indexed: 11/13/2022] Open
Abstract
The small-conductance, Ca2+-activated K+ (SK) channel subtype SK2 regulates the spike rate and firing frequency, as well as Ca2+ transients in Purkinje cells (PCs). To understand the molecular basis by which SK2 channels mediate these functions, we analyzed the exact location and densities of SK2 channels along the neuronal surface of the mouse cerebellar PCs using SDS-digested freeze-fracture replica labeling (SDS-FRL) of high sensitivity combined with quantitative analyses. Immunogold particles for SK2 were observed on post- and pre-synaptic compartments showing both scattered and clustered distribution patterns. We found an axo-somato-dendritic gradient of the SK2 particle density increasing 12-fold from soma to dendritic spines. Using two different immunogold approaches, we also found that SK2 immunoparticles were frequently adjacent to, but never overlap with, the postsynaptic density of excitatory synapses in PC spines. Co-immunoprecipitation analysis demonstrated that SK2 channels form macromolecular complexes with two types of proteins that mobilize Ca2+: CaV2.1 channels and mGlu1α receptors in the cerebellum. Freeze-fracture replica double-labeling showed significant co-clustering of particles for SK2 with those for CaV2.1 channels and mGlu1α receptors. SK2 channels were also detected at presynaptic sites, mostly at the presynaptic active zone (AZ), where they are close to CaV2.1 channels, though they are not significantly co-clustered. These data demonstrate that SK2 channels located in different neuronal compartments can associate with distinct proteins mobilizing Ca2+, and suggest that the ultrastructural association of SK2 with CaV2.1 and mGlu1α provides the mechanism that ensures voltage (excitability) regulation by distinct intracellular Ca2+ transients in PCs.
Collapse
Affiliation(s)
- Rafael Luján
- Synaptic Structure Laboratory, Instituto de Investigación en Discapacidades Neurológicas (IDINE), Departamento de Ciencias Médicas, Facultad de Medicina, Campus Biosanitario, Universidad Castilla-La Mancha, Albacete, Spain
| | - Carolina Aguado
- Synaptic Structure Laboratory, Instituto de Investigación en Discapacidades Neurológicas (IDINE), Departamento de Ciencias Médicas, Facultad de Medicina, Campus Biosanitario, Universidad Castilla-La Mancha, Albacete, Spain
| | - Francisco Ciruela
- Unitat de Farmacologia, Departament de Patologia i Terapèutica Experimental, Facultat de Medicina i Ciències de la Salut, IDIBELL, Universitat de Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain.,Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain.,Department of Biochemistry and Microbiology, Faculty of Sciences, University of Ghent, Ghent, Belgium
| | - Xavier Morató Arus
- Unitat de Farmacologia, Departament de Patologia i Terapèutica Experimental, Facultat de Medicina i Ciències de la Salut, IDIBELL, Universitat de Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Alejandro Martín-Belmonte
- Synaptic Structure Laboratory, Instituto de Investigación en Discapacidades Neurológicas (IDINE), Departamento de Ciencias Médicas, Facultad de Medicina, Campus Biosanitario, Universidad Castilla-La Mancha, Albacete, Spain
| | - Rocío Alfaro-Ruiz
- Synaptic Structure Laboratory, Instituto de Investigación en Discapacidades Neurológicas (IDINE), Departamento de Ciencias Médicas, Facultad de Medicina, Campus Biosanitario, Universidad Castilla-La Mancha, Albacete, Spain
| | - Jesús Martínez-Gómez
- Departamento de Sistemas Informáticos, Escuela Superior de Ingeniería Informática, Universidad de Castilla-La Mancha, Albacete, Spain
| | - Luis de la Ossa
- Departamento de Sistemas Informáticos, Escuela Superior de Ingeniería Informática, Universidad de Castilla-La Mancha, Albacete, Spain
| | - Masahiko Watanabe
- Department of Anatomy, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - John P Adelman
- Vollum Institute, Oregon Health and Science University, Portland, OR, United States
| | - Ryuichi Shigemoto
- Institute of Science and Technology (IST Austria), Klosterneuburg, Austria
| | - Yugo Fukazawa
- Division of Brain Structure and Function, Research Center for Child Mental Development, Life Science Advancement Program, Faculty of Medical Science, University of Fukui, Fukui, Japan
| |
Collapse
|
26
|
Bajorat R, Porath K, Kuhn J, Goßla E, Goerss D, Sellmann T, Köhling R, Kirschstein T. Oral administration of the casein kinase 2 inhibitor TBB leads to persistent K Ca2.2 channel up-regulation in the epileptic CA1 area and cortex, but lacks anti-seizure efficacy in the pilocarpine epilepsy model. Epilepsy Res 2018; 147:42-50. [PMID: 30219695 DOI: 10.1016/j.eplepsyres.2018.08.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 07/31/2018] [Accepted: 08/29/2018] [Indexed: 02/04/2023]
Abstract
Temporal lobe epilepsy (TLE) is the most common epileptic syndrome in adults and often presents with seizures that prove intractable with currently available anticonvulsants. Thus, there is still a need for new anti-seizure drugs in this condition. Recently, we found that the casein kinase 2 inhibitor 4,5,6,7-tetrabromotriazole (TBB) prevented the emergence of spontaneous epileptic discharges in an acute in vitro epilepsy model. This prompted us to study the anti-seizure effects of TBB in the pilocarpine model of chronic epilepsy in vivo. To this end, we performed long-term video-EEG monitoring lasting 78-167 days of nine chronically epileptic rats and obtained a baseline seizure rate of 3.3 ± 1.3 per day (baseline of 27-80 days). We found a significant age effect with more pronounced seizure rates in older animals as compared to younger ones. However, the seizure rate increased to 6.3 ± 2.2 per day during the oral TBB administration (treatment period of 21-50 days), and following discontinuation of TBB, this rate remained stable with 5.2 ± 1.4 seizures per day (follow-up of 30-55 days). After completing the video-EEG during the follow-up the hippocampal tissue was prepared and studied for the expression of the Ca2+-activated K+ channel KCa2.2. We found a significant up-regulation of KCa2.2 in the epileptic CA1 region and in the neocortex, but in no other hippocampal subfield. Hence, our findings indicate that oral administration of TBB leads to persistent up-regulation of KCa2.2 in the epileptic CA1 subfield and in the neocortex, but lacks anti-seizure efficacy in the pilocarpine epilepsy model.
Collapse
Affiliation(s)
- Rika Bajorat
- Oscar Langendorff Institute of Physiology, Rostock University Medical Centre, Rostock, Germany; Department of Anesthesiology and Intensive Care Medicine, Rostock University Medical Centre, Rostock, Germany.
| | - Katrin Porath
- Oscar Langendorff Institute of Physiology, Rostock University Medical Centre, Rostock, Germany.
| | - Johannes Kuhn
- Oscar Langendorff Institute of Physiology, Rostock University Medical Centre, Rostock, Germany.
| | - Elke Goßla
- Oscar Langendorff Institute of Physiology, Rostock University Medical Centre, Rostock, Germany.
| | - Doreen Goerss
- Oscar Langendorff Institute of Physiology, Rostock University Medical Centre, Rostock, Germany; Department of Psychosomatic and Psychotherapeutic Medicine, Rostock University Medical Centre, Rostock, Germany.
| | - Tina Sellmann
- Oscar Langendorff Institute of Physiology, Rostock University Medical Centre, Rostock, Germany.
| | - Rüdiger Köhling
- Oscar Langendorff Institute of Physiology, Rostock University Medical Centre, Rostock, Germany.
| | - Timo Kirschstein
- Oscar Langendorff Institute of Physiology, Rostock University Medical Centre, Rostock, Germany.
| |
Collapse
|
27
|
Hypobaric Hypoxia-Induced Learning and Memory Impairment: Elucidating the Role of Small Conductance Ca2+-Activated K+ Channels. Neuroscience 2018; 388:418-429. [DOI: 10.1016/j.neuroscience.2018.07.026] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 07/11/2018] [Accepted: 07/13/2018] [Indexed: 11/19/2022]
|
28
|
Skarsfeldt MA, Bomholtz SH, Lundegaard PR, Lopez-Izquierdo A, Tristani-Firouzi M, Bentzen BH. Atrium-specific ion channels in the zebrafish-A role of I KACh in atrial repolarization. Acta Physiol (Oxf) 2018; 223:e13049. [PMID: 29412518 DOI: 10.1111/apha.13049] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 01/26/2018] [Accepted: 01/29/2018] [Indexed: 12/16/2022]
Abstract
AIM The zebrafish has emerged as a novel model for investigating cardiac physiology and pathology. The aim of this study was to investigate the atrium-specific ion channels responsible for shaping the atrial cardiac action potential in zebrafish. METHODS Using quantitative polymerase chain reaction, we assessed the expression level of atrium-specific potassium channels. The functional role of these channels was studied by patch clamp experiments on isolated atrial and ventricular cardiomyocytes and by optical mapping of explanted adult zebrafish hearts. Finally, surface ECGs were recorded to establish possible in vivo roles of atrial ion channels. RESULTS In isolated adult zebrafish hearts, we identified the expression of kcnk3, kcnk9, kcnn1, kcnn2, kcnn3, kcnj3 and kcnj5, the genes that encode the atrium-specific K2P , KCa 2.x and Kir 3.1/4 (KACh ) ion channels. The electrophysiological data indicate that the acetylcholine-activated inward-rectifying current, IKACh, plays a major role in the zebrafish atrium, whereas K2P 3.1/9.1 and KCa 2.x channels do not appear to be involved in regulating the action potential in the zebrafish heart. CONCLUSION We demonstrate that the acetylcholine-activated inward-rectifying current (IKACh ) current plays a major role in the zebrafish atrium and that the zebrafish could potentially be a cost-effective and reliable model for pharmacological testing of atrium-specific IKACh modulating compounds.
Collapse
Affiliation(s)
- M. A. Skarsfeldt
- Department of Biomedical Sciences; Faculty of Health and Medical Sciences; University of Copenhagen; Copenhagen N Denmark
- Nora Eccles Harrison Cardiovascular Research and Training Institute; University of Utah; Salt Lake City UT USA
| | - S. H. Bomholtz
- Department of Biomedical Sciences; Faculty of Health and Medical Sciences; University of Copenhagen; Copenhagen N Denmark
| | - P. R. Lundegaard
- Department of Biomedical Sciences; Faculty of Health and Medical Sciences; University of Copenhagen; Copenhagen N Denmark
| | - A. Lopez-Izquierdo
- Nora Eccles Harrison Cardiovascular Research and Training Institute; University of Utah; Salt Lake City UT USA
| | - M. Tristani-Firouzi
- Nora Eccles Harrison Cardiovascular Research and Training Institute; University of Utah; Salt Lake City UT USA
| | - B. H. Bentzen
- Department of Biomedical Sciences; Faculty of Health and Medical Sciences; University of Copenhagen; Copenhagen N Denmark
| |
Collapse
|
29
|
Abiraman K, Tzingounis AV, Lykotrafitis G. K Ca2 channel localization and regulation in the axon initial segment. FASEB J 2018; 32:1794-1805. [PMID: 29180442 DOI: 10.1096/fj.201700605r] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Small conductance calcium-activated potassium (KCa2) channels are expressed throughout the CNS and play a critical role in synaptic and neuronal excitability. KCa2 channels have a somatodendritic distribution with their highest expression in distal dendrites. It is unclear whether KCa2 channels are specifically present on the axon initial segment (AIS), the site at which action potentials are initiated in neurons. Through a powerful combination of toxin pharmacology, single-molecule atomic force microscopy, and dual-color fluorescence microscopy, we report here that KCa2 channels-predominantly the KCa2.3 subtype-are indeed present on the AIS. We also report that cAMP-PKA controls the axonal KCa2 channel surface expression. Surprisingly, and in contrast to KCa2 channels that were observed in the soma and dendrites, the inhibition of cAMP-PKA increased the surface expression of KCa2 channels without promoting nanoclustering. Lastly, we found that axonal KCa2 channels seem to undergo endocytosis in a dynamin-independent manner, unlike KCa2 channels in the soma and dendrites. Together, these novel results demonstrate that the distribution and membrane recycling of KCa2 channels differs among various neuronal subcompartments.-Abiraman, K., Tzingounis, A. V., Lykotrafitis, G. KCa2 channel localization and regulation in the axon initial segment.
Collapse
Affiliation(s)
- Krithika Abiraman
- Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut, USA
| | - Anastasios V Tzingounis
- Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut, USA.,Department of Physiology and Neurobiology, University of Connecticut, Storrs, Connecticut, USA
| | - George Lykotrafitis
- Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut, USA.,Department of Mechanical Engineering, University of Connecticut, Storrs, Connecticut, USA
| |
Collapse
|
30
|
A non-synaptic mechanism of complex learning: Modulation of intrinsic neuronal excitability. Neurobiol Learn Mem 2017; 154:30-36. [PMID: 29196146 DOI: 10.1016/j.nlm.2017.11.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Revised: 11/06/2017] [Accepted: 11/27/2017] [Indexed: 11/24/2022]
Abstract
Training rats in a particularly difficult olfactory discrimination task initiates a period of accelerated learning of other odors, manifested as a dramatic increase in the rats' capacity to acquire memories for new odors once they have learned the first discrimination task, implying that rule learning has taken place. At the cellular level, pyramidal neurons in the piriform cortex, hippocampus and bsolateral amygdala of olfactory-discrimination trained rats show enhanced intrinsic neuronal excitability that lasts for several days after rule learning. Such enhanced intrinsic excitability is mediated by long-term reduction in the post-burst after-hyperpolarization (AHP) which is generated by repetitive spike firing, and is maintained by persistent activation of key second messenger systems. Much like late-LTP, the induction of long-term modulation of intrinsic excitability is protein synthesis dependent. Learning-induced modulation of intrinsic excitability can be bi-directional, pending of the valance of the outcome of the learned task. In this review we describe the physiological and molecular mechanisms underlying the rule learning-induced long-term enhancement in neuronal excitability and discuss the functional significance of such a wide spread modulation of the neurons' ability to sustain repetitive spike generation.
Collapse
|
31
|
Bröker-Lai J, Kollewe A, Schindeldecker B, Pohle J, Nguyen Chi V, Mathar I, Guzman R, Schwarz Y, Lai A, Weißgerber P, Schwegler H, Dietrich A, Both M, Sprengel R, Draguhn A, Köhr G, Fakler B, Flockerzi V, Bruns D, Freichel M. Heteromeric channels formed by TRPC1, TRPC4 and TRPC5 define hippocampal synaptic transmission and working memory. EMBO J 2017; 36:2770-2789. [PMID: 28790178 DOI: 10.15252/embj.201696369] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 07/01/2017] [Accepted: 07/07/2017] [Indexed: 12/30/2022] Open
Abstract
Canonical transient receptor potential (TRPC) channels influence various neuronal functions. Using quantitative high-resolution mass spectrometry, we demonstrate that TRPC1, TRPC4, and TRPC5 assemble into heteromultimers with each other, but not with other TRP family members in the mouse brain and hippocampus. In hippocampal neurons from Trpc1/Trpc4/Trpc5-triple-knockout (Trpc1/4/5-/-) mice, lacking any TRPC1-, TRPC4-, or TRPC5-containing channels, action potential-triggered excitatory postsynaptic currents (EPSCs) were significantly reduced, whereas frequency, amplitude, and kinetics of quantal miniature EPSC signaling remained unchanged. Likewise, evoked postsynaptic responses in hippocampal slice recordings and transient potentiation after tetanic stimulation were decreased. In vivo, Trpc1/4/5-/- mice displayed impaired cross-frequency coupling in hippocampal networks and deficits in spatial working memory, while spatial reference memory was unaltered. Trpc1/4/5-/- animals also exhibited deficiencies in adapting to a new challenge in a relearning task. Our results indicate the contribution of heteromultimeric channels from TRPC1, TRPC4, and TRPC5 subunits to the regulation of mechanisms underlying spatial working memory and flexible relearning by facilitating proper synaptic transmission in hippocampal neurons.
Collapse
Affiliation(s)
- Jenny Bröker-Lai
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany
| | - Astrid Kollewe
- Institute of Physiology, University of Freiburg, Freiburg, Germany
| | - Barbara Schindeldecker
- Center for Integrative Physiology and Molecular Medicine, Saarland University, Homburg, Germany
| | - Jörg Pohle
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany.,Physiology of Neural Networks, Psychiatry/Psychopharmacology, Central Institute of Mental Health, J5, Heidelberg University, Mannheim, Germany
| | - Vivan Nguyen Chi
- Institute of Physiology and Pathophysiology, Heidelberg University, Heidelberg, Germany
| | - Ilka Mathar
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany
| | - Raul Guzman
- Center for Integrative Physiology and Molecular Medicine, Saarland University, Homburg, Germany
| | - Yvonne Schwarz
- Center for Integrative Physiology and Molecular Medicine, Saarland University, Homburg, Germany
| | - Alan Lai
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany
| | - Petra Weißgerber
- Experimental and Clinical Pharmacology and Toxicology, Saarland University, Homburg, Germany
| | | | - Alexander Dietrich
- Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University München, München, Germany
| | - Martin Both
- Institute of Physiology and Pathophysiology, Heidelberg University, Heidelberg, Germany
| | - Rolf Sprengel
- Max Planck Research Group of the Max Planck Institute for Medical Research at the Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Andreas Draguhn
- Institute of Physiology and Pathophysiology, Heidelberg University, Heidelberg, Germany
| | - Georg Köhr
- Physiology of Neural Networks, Psychiatry/Psychopharmacology, Central Institute of Mental Health, J5, Heidelberg University, Mannheim, Germany
| | - Bernd Fakler
- Institute of Physiology, University of Freiburg, Freiburg, Germany.,BIOSS, Center for Biological Signaling Studies, University of Freiburg, Freiburg, Germany‡
| | - Veit Flockerzi
- Experimental and Clinical Pharmacology and Toxicology, Saarland University, Homburg, Germany
| | - Dieter Bruns
- Center for Integrative Physiology and Molecular Medicine, Saarland University, Homburg, Germany
| | - Marc Freichel
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
32
|
Cadet JL, Brannock C, Krasnova IN, Jayanthi S, Ladenheim B, McCoy MT, Walther D, Godino A, Pirooznia M, Lee RS. Genome-wide DNA hydroxymethylation identifies potassium channels in the nucleus accumbens as discriminators of methamphetamine addiction and abstinence. Mol Psychiatry 2017; 22:1196-1204. [PMID: 27046646 PMCID: PMC7405865 DOI: 10.1038/mp.2016.48] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Revised: 01/28/2016] [Accepted: 02/18/2016] [Indexed: 12/17/2022]
Abstract
Epigenetic consequences of exposure to psychostimulants are substantial but the relationship of these changes to compulsive drug taking and abstinence is not clear. Here, we used a paradigm that helped to segregate rats that reduce or stop their methamphetamine (METH) intake (nonaddicted) from those that continue to take the drug compulsively (addicted) in the presence of footshocks. We used that model to investigate potential alterations in global DNA hydroxymethylation in the nucleus accumbens (NAc) because neuroplastic changes in the NAc may participate in the development and maintenance of drug-taking behaviors. We found that METH-addicted rats did indeed show differential DNA hydroxymethylation in comparison with both control and nonaddicted rats. Nonaddicted rats also showed differences from control rats. Differential DNA hydroxymethylation observed in addicted rats occurred mostly at intergenic sites located on long and short interspersed elements. Interestingly, differentially hydroxymethylated regions in genes encoding voltage (Kv1.1, Kv1.2, Kvb1 and Kv2.2)- and calcium (Kcnma1, Kcnn1 and Kcnn2)-gated potassium channels observed in the NAc of nonaddicted rats were accompanied by increased mRNA levels of these potassium channels when compared with mRNA expression in METH-addicted rats. These observations indicate that changes in differentially hydroxymethylated regions and increased expression of specific potassium channels in the NAc may promote abstinence from drug-taking behaviors. Thus, activation of specific subclasses of voltage- and/or calcium-gated potassium channels may provide an important approach to the beneficial treatment for METH addiction.
Collapse
Affiliation(s)
- Jean Lud Cadet
- Molecular Neuropsychiatry Research Branch, NIDA Intramural Research Program Baltimore, MD, USA
| | - Christie Brannock
- Molecular Neuropsychiatry Research Branch, NIDA Intramural Research Program Baltimore, MD, USA
| | - Irina N. Krasnova
- Molecular Neuropsychiatry Research Branch, NIDA Intramural Research Program Baltimore, MD, USA
| | - Subramaniam Jayanthi
- Molecular Neuropsychiatry Research Branch, NIDA Intramural Research Program Baltimore, MD, USA
| | - Bruce Ladenheim
- Molecular Neuropsychiatry Research Branch, NIDA Intramural Research Program Baltimore, MD, USA
| | - Michael T. McCoy
- Molecular Neuropsychiatry Research Branch, NIDA Intramural Research Program Baltimore, MD, USA
| | - Donna Walther
- Molecular Neuropsychiatry Research Branch, NIDA Intramural Research Program Baltimore, MD, USA
| | - Arthur Godino
- Département de Biologie, École Normale Supérieure de Lyon, Lyon, France
| | - Mehdi Pirooznia
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Richard S. Lee
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
| |
Collapse
|
33
|
Kaczmarek LK, Aldrich RW, Chandy KG, Grissmer S, Wei AD, Wulff H. International Union of Basic and Clinical Pharmacology. C. Nomenclature and Properties of Calcium-Activated and Sodium-Activated Potassium Channels. Pharmacol Rev 2017; 69:1-11. [PMID: 28267675 PMCID: PMC11060434 DOI: 10.1124/pr.116.012864] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2024] Open
Abstract
A subset of potassium channels is regulated primarily by changes in the cytoplasmic concentration of ions, including calcium, sodium, chloride, and protons. The eight members of this subfamily were originally all designated as calcium-activated channels. More recent studies have clarified the gating mechanisms for these channels and have documented that not all members are sensitive to calcium. This article describes the molecular relationships between these channels and provides an introduction to their functional properties. It also introduces a new nomenclature that differentiates between calcium- and sodium-activated potassium channels.
Collapse
Affiliation(s)
- Leonard K Kaczmarek
- Departments of Pharmacology and Cellular and Molecular Physiology, Yale School of Medicine, New Haven, Connecticut (L.K.K.); Center for Learning and Memory and Department of Neuroscience, University of Texas at Austin, Austin, Texas (R.W.A.); Laboratory of Molecular Physiology in the Infection and Immunity Theme, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore (K.G.C.); Institute of Applied Physiology, Ulm University, Ulm, Germany (S.G.); Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington (A.D.W.); and Department of Pharmacology, School of Medicine, University of California, Davis, California (H.W.)
| | - Richard W Aldrich
- Departments of Pharmacology and Cellular and Molecular Physiology, Yale School of Medicine, New Haven, Connecticut (L.K.K.); Center for Learning and Memory and Department of Neuroscience, University of Texas at Austin, Austin, Texas (R.W.A.); Laboratory of Molecular Physiology in the Infection and Immunity Theme, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore (K.G.C.); Institute of Applied Physiology, Ulm University, Ulm, Germany (S.G.); Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington (A.D.W.); and Department of Pharmacology, School of Medicine, University of California, Davis, California (H.W.)
| | - K George Chandy
- Departments of Pharmacology and Cellular and Molecular Physiology, Yale School of Medicine, New Haven, Connecticut (L.K.K.); Center for Learning and Memory and Department of Neuroscience, University of Texas at Austin, Austin, Texas (R.W.A.); Laboratory of Molecular Physiology in the Infection and Immunity Theme, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore (K.G.C.); Institute of Applied Physiology, Ulm University, Ulm, Germany (S.G.); Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington (A.D.W.); and Department of Pharmacology, School of Medicine, University of California, Davis, California (H.W.)
| | - Stephan Grissmer
- Departments of Pharmacology and Cellular and Molecular Physiology, Yale School of Medicine, New Haven, Connecticut (L.K.K.); Center for Learning and Memory and Department of Neuroscience, University of Texas at Austin, Austin, Texas (R.W.A.); Laboratory of Molecular Physiology in the Infection and Immunity Theme, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore (K.G.C.); Institute of Applied Physiology, Ulm University, Ulm, Germany (S.G.); Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington (A.D.W.); and Department of Pharmacology, School of Medicine, University of California, Davis, California (H.W.)
| | - Aguan D Wei
- Departments of Pharmacology and Cellular and Molecular Physiology, Yale School of Medicine, New Haven, Connecticut (L.K.K.); Center for Learning and Memory and Department of Neuroscience, University of Texas at Austin, Austin, Texas (R.W.A.); Laboratory of Molecular Physiology in the Infection and Immunity Theme, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore (K.G.C.); Institute of Applied Physiology, Ulm University, Ulm, Germany (S.G.); Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington (A.D.W.); and Department of Pharmacology, School of Medicine, University of California, Davis, California (H.W.)
| | - Heike Wulff
- Departments of Pharmacology and Cellular and Molecular Physiology, Yale School of Medicine, New Haven, Connecticut (L.K.K.); Center for Learning and Memory and Department of Neuroscience, University of Texas at Austin, Austin, Texas (R.W.A.); Laboratory of Molecular Physiology in the Infection and Immunity Theme, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore (K.G.C.); Institute of Applied Physiology, Ulm University, Ulm, Germany (S.G.); Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington (A.D.W.); and Department of Pharmacology, School of Medicine, University of California, Davis, California (H.W.)
| |
Collapse
|
34
|
Abstract
Small-conductance Ca2+-activated potassium (SK) channels are relative newcomers within the field of cardiac electrophysiology. In recent years, an increased focus has been given to these channels because they might constitute a relatively atrial-selective target. This review will give a general introduction to SK channels followed by their proposed function in the heart under normal and pathophysiological conditions. It is revealed how antiarrhythmic effects can be obtained by SK channel inhibition in a number of species in situations of atrial fibrillation. On the contrary, the beneficial effects of SK channel inhibition in situations of heart failure are questionable and still needs investigation. The understanding of cardiac SK channels is rapidly increasing these years, and it is hoped that this will clarify whether SK channel inhibition has potential as a new anti–atrial fibrillation principle.
Collapse
|
35
|
Willis M, Trieb M, Leitner I, Wietzorrek G, Marksteiner J, Knaus HG. Small-conductance calcium-activated potassium type 2 channels (SK2, KCa2.2) in human brain. Brain Struct Funct 2016; 222:973-979. [PMID: 27357310 PMCID: PMC5334391 DOI: 10.1007/s00429-016-1258-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 06/20/2016] [Indexed: 12/02/2022]
Abstract
SK2 (KCa2.2) channels are voltage-independent Ca2+-activated K+ channels that regulate neuronal excitability in brain regions important for memory formation. In this study, we investigated the distribution and expression of SK2 channels in human brain by Western blot analysis and immunohistochemistry. Immunoblot analysis of human brain indicated expression of four distinct SK2 channel isoforms: the standard, the long and two short isoforms. Immunohistochemistry in paraffin-embedded post-mortem brain sections was performed in the hippocampal formation, amygdala and neocortex. In hippocampus, SK2-like immunoreactivity could be detected in strata oriens and radiatum of area CA1-CA2 and in the molecular layer. In the amygdala, SK2-like immunoreactivity was highest in the basolateral nuclei, while in neocortex, staining was mainly found enriched in layer V. Activation of SK2 channels is thought to regulate neuronal excitability in brain by contributing to the medium afterhyperpolarization. However, SK2 channels are blocked by apamin with a sensitivity that suggests heteromeric channels. The herein first shown expression of SK2 human isoform b in brain could explain the variability of electrophysiological findings observed with SK2 channels.
Collapse
Affiliation(s)
- Michael Willis
- Department of Psychiatry, Psychotherapy and Psychosomatics, Medical University Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria.
| | - Maria Trieb
- Division of Molecular and Cellular Pharmacology, Medical University Innsbruck, Peter Mayr Strasse 1, 6020, Innsbruck, Austria
| | - Irmgard Leitner
- Division of Molecular and Cellular Pharmacology, Medical University Innsbruck, Peter Mayr Strasse 1, 6020, Innsbruck, Austria
| | - Georg Wietzorrek
- Division of Molecular and Cellular Pharmacology, Medical University Innsbruck, Peter Mayr Strasse 1, 6020, Innsbruck, Austria
| | - Josef Marksteiner
- Department of Psychiatry and Psychotherapy A, Landeskrankenhaus Hall in Tirol, Milser Strasse 10, 6060, Hall in Tirol, Austria
| | - Hans-Günther Knaus
- Division of Molecular and Cellular Pharmacology, Medical University Innsbruck, Peter Mayr Strasse 1, 6020, Innsbruck, Austria
| |
Collapse
|
36
|
Abiraman K, Sah M, Walikonis RS, Lykotrafitis G, Tzingounis AV. Tonic PKA Activity Regulates SK Channel Nanoclustering and Somatodendritic Distribution. J Mol Biol 2016; 428:2521-2537. [PMID: 27107637 DOI: 10.1016/j.jmb.2016.04.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 03/28/2016] [Accepted: 04/07/2016] [Indexed: 01/02/2023]
Abstract
Small-conductance calcium-activated potassium (SK) channels mediate a potassium conductance in the brain and are involved in synaptic plasticity, learning, and memory. SK channels show a distinct subcellular localization that is crucial for their neuronal functions. However, the mechanisms that control this spatial distribution are unknown. We imaged SK channels labeled with fluorophore-tagged apamin and monitored SK channel nanoclustering at the single molecule level by combining atomic force microscopy and toxin (i.e., apamin) pharmacology. Using these two complementary approaches, we found that native SK channel distribution in pyramidal neurons, across the somatodendritic domain, depends on ongoing cyclic adenosine monophosphate (cAMP)-protein kinase A (PKA) levels, strongly limiting SK channel expression at the pyramidal neuron soma. Furthermore, tonic cAMP-PKA levels also controlled whether SK channels were expressed in nanodomains as single entities or as a group of multiple channels. Our study reveals a new level of regulation of SK channels by cAMP-PKA and suggests that ion channel topography and nanoclustering might be under the control of second messenger cascades.
Collapse
Affiliation(s)
- Krithika Abiraman
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA
| | - Megha Sah
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT 06269, USA
| | - Randall S Walikonis
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT 06269, USA
| | - George Lykotrafitis
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA; Department of Mechanical Engineering, University of Connecticut, Storrs, CT 06269, USA.
| | - Anastasios V Tzingounis
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA; Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT 06269, USA.
| |
Collapse
|
37
|
Yap FC, Weber DS, Taylor MS, Townsley MI, Comer BS, Maylie J, Adelman JP, Lin MT. Endothelial SK3 channel-associated Ca2+ microdomains modulate blood pressure. Am J Physiol Heart Circ Physiol 2016; 310:H1151-63. [PMID: 26945080 DOI: 10.1152/ajpheart.00787.2015] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Accepted: 02/22/2016] [Indexed: 11/22/2022]
Abstract
Activation of vascular endothelial small- (KCa2.3, SK3) or intermediate- (KCa3.1, IK1) conductance Ca(2+)-activated potassium channels induces vasorelaxation via an endothelium-derived hyperpolarization (EDH) pathway. Although the activation of SK3 and IK1 channels converges on EDH, their subcellular effects on signal transduction are different and not completely clear. In this study, a novel endothelium-specific SK3 knockout (SK3(-/-)) mouse model was utilized to specifically examine the contribution of SK3 channels to mesenteric artery vasorelaxation, endothelial Ca(2+) dynamics, and blood pressure. The absence of SK3 expression was confirmed using real-time quantitative PCR and Western blot analysis. Functional studies showed impaired EDH-mediated vasorelaxation in SK3(-/-) small mesenteric arteries. Immunostaining results from SK3(-/-) vessels confirmed the absence of SK3 and further showed altered distribution of transient receptor potential channels, type 4 (TRPV4). Electrophysiological recordings showed a lack of SK3 channel activity, while TRPV4-IK1 channel coupling remained intact in SK3(-/-) endothelial cells. Moreover, Ca(2+) imaging studies in SK3(-/-) endothelium showed increased Ca(2+) transients with reduced amplitude and duration under basal conditions. Importantly, SK3(-/-) endothelium lacked a distinct type of Ca(2+) dynamic that is sensitive to TRPV4 activation. Blood pressure measurements showed that the SK3(-/-) mice were hypertensive, and the blood pressure increase was further enhanced during the 12-h dark cycle when animals are most active. Taken together, our results reveal a previously unappreciated SK3 signaling microdomain that modulates endothelial Ca(2+) dynamics, vascular tone, and blood pressure.
Collapse
Affiliation(s)
- Fui C Yap
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, Alabama
| | - David S Weber
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, Alabama
| | - Mark S Taylor
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, Alabama
| | - Mary I Townsley
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, Alabama
| | - Brian S Comer
- Department of Cellular and Integrative Physiology, Indiana University, Indianapolis, Indiana
| | - James Maylie
- Department of Obstetrics and Gynecology, Oregon Health & Science University, Portland, Oregon; and
| | - John P Adelman
- Vollum Institute, Oregon Health & Science University, Portland, Oregon
| | - Mike T Lin
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, Alabama;
| |
Collapse
|
38
|
Bock T, Stuart GJ. Impact of calcium-activated potassium channels on NMDA spikes in cortical layer 5 pyramidal neurons. J Neurophysiol 2016; 115:1740-8. [PMID: 26936985 DOI: 10.1152/jn.01047.2015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 01/20/2016] [Indexed: 01/01/2023] Open
Abstract
Active electrical events play an important role in shaping signal processing in dendrites. As these events are usually associated with an increase in intracellular calcium, they are likely to be under the control of calcium-activated potassium channels. Here, we investigate the impact of calcium-activated potassium channels onN-methyl-d-aspartate (NMDA) receptor-dependent spikes, or NMDA spikes, evoked by glutamate iontophoresis onto basal dendrites of cortical layer 5 pyramidal neurons. We found that small-conductance calcium-activated potassium channels (SK channels) act to reduce NMDA spike amplitude but at the same time, also decrease the iontophoretic current required for their generation. This SK-mediated decrease in NMDA spike threshold was dependent on R-type voltage-gated calcium channels and indicates a counterintuitive, excitatory effect of SK channels on NMDA spike generation, whereas the capacity of SK channels to suppress NMDA spike amplitude is in line with the expected inhibitory action of potassium channels on dendritic excitability. Large-conductance calcium-activated potassium channels had no significant impact on NMDA spikes, indicating that these channels are either absent from basal dendrites or not activated by NMDA spikes. These experiments reveal complex and opposing interactions among NMDA receptors, SK channels, and voltage-gated calcium channels in basal dendrites of cortical layer 5 pyramidal neurons during NMDA spike generation, which are likely to play an important role in regulating the way these neurons integrate the thousands of synaptic inputs they receive.
Collapse
Affiliation(s)
- Tobias Bock
- Eccles Institute of Neuroscience and Australian Research Council Centre of Excellence for Integrative Brain Function, John Curtin School of Medical Research, Australian National University, Canberra, Australia
| | - Greg J Stuart
- Eccles Institute of Neuroscience and Australian Research Council Centre of Excellence for Integrative Brain Function, John Curtin School of Medical Research, Australian National University, Canberra, Australia
| |
Collapse
|
39
|
Nimitvilai S, Lopez MF, Mulholland PJ, Woodward JJ. Chronic Intermittent Ethanol Exposure Enhances the Excitability and Synaptic Plasticity of Lateral Orbitofrontal Cortex Neurons and Induces a Tolerance to the Acute Inhibitory Actions of Ethanol. Neuropsychopharmacology 2016; 41:1112-27. [PMID: 26286839 PMCID: PMC4748436 DOI: 10.1038/npp.2015.250] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2015] [Revised: 08/14/2015] [Accepted: 08/15/2015] [Indexed: 02/07/2023]
Abstract
Alcoholism is associated with changes in brain reward and control systems, including the prefrontal cortex. In prefrontal areas, the orbitofrontal cortex (OFC) has been suggested to have an important role in the development of alcohol-abuse disorders and studies from this laboratory demonstrate that OFC-mediated behaviors are impaired in alcohol-dependent animals. However, it is not known whether chronic alcohol (ethanol) exposure alters the fundamental properties of OFC neurons. In this study, mice were exposed to repeated cycles of chronic intermittent ethanol (CIE) exposure to induce dependence and whole-cell patch-clamp electrophysiology was used to examine the effects of CIE treatment on lateral OFC (lOFC) neuron excitability, synaptic transmission, and plasticity. Repeated cycles of CIE exposure and withdrawal enhanced current-evoked action potential (AP) spiking and this was accompanied by a reduction in the after-hyperpolarization and a decrease in the functional activity of SK channels. CIE mice also showed an increase in the AMPA/NMDA ratio, and this was associated with an increase in GluA1/GluA2 AMPA receptor expression and a decrease in GluN2B NMDA receptor subunits. Following CIE treatment, lOFC neurons displayed a persistent long-term potentiation of glutamatergic synaptic transmission following a spike-timing-dependent protocol. Lastly, CIE treatment diminished the inhibitory effect of acute ethanol on AP spiking of lOFC neurons and reduced expression of the GlyT1 transporter. Taken together, these results suggest that chronic exposure to ethanol leads to enhanced intrinsic excitability and glutamatergic synaptic signaling of lOFC neurons. These alterations may contribute to the impairment of OFC-dependent behaviors in alcohol-dependent individuals.
Collapse
Affiliation(s)
- Sudarat Nimitvilai
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Marcelo F Lopez
- Addiction Sciences Division, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Patrick J Mulholland
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
- Addiction Sciences Division, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - John J Woodward
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
- Addiction Sciences Division, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
40
|
Wang K, Mateos-Aparicio P, Hönigsperger C, Raghuram V, Wu WW, Ridder MC, Sah P, Maylie J, Storm JF, Adelman JP. IK1 channels do not contribute to the slow afterhyperpolarization in pyramidal neurons. eLife 2016; 5:e11206. [PMID: 26765773 PMCID: PMC4733036 DOI: 10.7554/elife.11206] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 12/01/2015] [Indexed: 11/16/2022] Open
Abstract
In pyramidal neurons such as hippocampal area CA1 and basolateral amygdala, a slow afterhyperpolarization (sAHP) follows a burst of action potentials, which is a powerful regulator of neuronal excitability. The sAHP amplitude increases with aging and may underlie age related memory decline. The sAHP is due to a Ca2+-dependent, voltage-independent K+ conductance, the molecular identity of which has remained elusive until a recent report suggested the Ca2+-activated K+ channel, IK1 (KCNN4) as the sAHP channel in CA1 pyramidal neurons. The signature pharmacology of IK1, blockade by TRAM-34, was reported for the sAHP and underlying current. We have examined the sAHP and find no evidence that TRAM-34 affects either the current underling the sAHP or excitability of CA1 or basolateral amygdala pyramidal neurons. In addition, CA1 pyramidal neurons from IK1 null mice exhibit a characteristic sAHP current. Our results indicate that IK1 channels do not mediate the sAHP in pyramidal neurons. DOI:http://dx.doi.org/10.7554/eLife.11206.001 Neurons carry signals in the form of electrical impulses called action potentials. These nerve impulses result from ions flowing through proteins called ion channels in the neuron’s membrane, and they determine how the neuron communicates with neighboring neurons. The number of action potentials a neuron can produce can vary over a wide range. In the brain, a particular kind of ion channel limits the number of action potentials that many neurons produce via a negative feedback mechanism. That is to say, nerve impulses activate this ion channel and the activated channel then makes the neuron less able to send further nerve impulses for a while.The activity of this ion channel increases with age and it may be responsible for some forms of age-related decline in cognitive abilities. However, the exact identity of the ion channel responsible was unclear. Recent research has suggested the ion channel in question was a protein called IK1. This conclusion was largely based on how this ion channel responded to drugs in the laboratory. Wang, Materos-Aparico et al. sought to verify this conclusion and, in contrast with the previous reports, found that the IK1 ion channel did not respond to these drugs in the same way when it was in neurons in the brains of mice. In further experiments, mice that had been engineered to lack the IK1 ion channel still showed the characteristic negative feedback that regulates the firing of action potentials. Thus, Wang, Materos-Aparico et al. found no evidence to support the previous conclusion, and instead conclude that the exact identity of this important ion channel in the brain has yet to be defined. DOI:http://dx.doi.org/10.7554/eLife.11206.002
Collapse
Affiliation(s)
- Kang Wang
- Vollum Institute, Oregon Health and Science University, Portland, United States
| | - Pedro Mateos-Aparicio
- Department of Physiology, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Christoph Hönigsperger
- Department of Physiology, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Vijeta Raghuram
- Vollum Institute, Oregon Health and Science University, Portland, United States
| | - Wendy W Wu
- Department of Obstetrics and Gynecology, Oregon Health and Science University, Portland, United States
| | - Margreet C Ridder
- Queensland Brain Institute, The University of Queensland, Brisbane, Australia
| | - Pankaj Sah
- Queensland Brain Institute, The University of Queensland, Brisbane, Australia
| | - Jim Maylie
- Department of Obstetrics and Gynecology, Oregon Health and Science University, Portland, United States
| | - Johan F Storm
- Department of Physiology, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - John P Adelman
- Vollum Institute, Oregon Health and Science University, Portland, United States
| |
Collapse
|
41
|
Thompson JM, Ji G, Neugebauer V. Small-conductance calcium-activated potassium (SK) channels in the amygdala mediate pain-inhibiting effects of clinically available riluzole in a rat model of arthritis pain. Mol Pain 2015; 11:51. [PMID: 26311432 PMCID: PMC4551697 DOI: 10.1186/s12990-015-0055-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 08/21/2015] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Arthritis pain is an important healthcare issue with significant emotional and affective consequences. Here we focus on potentially beneficial effects of activating small-conductance calcium-activated potassium (SK) channels in the amygdala, a brain center of emotions that plays an important role in central pain modulation and processing. SK channels have been reported to regulate neuronal activity in the central amygdala (CeA, output nucleus). We tested the effects of riluzole, a clinically available drug for the treatment of amyotrophic lateral sclerosis, for the following reasons. Actions of riluzole include activation of SK channels. Evidence in the literature suggests that riluzole may have antinociceptive effects through an action in the brain but not the spinal cord. Mechanism and site of action of riluzole remain to be determined. Here we tested the hypothesis that riluzole inhibits pain behaviors by acting on SK channels in the CeA in an arthritis pain model. RESULTS Systemic (intraperitoneal) application of riluzole (8 mg/kg) inhibited audible (nocifensive response) and ultrasonic (averse affective response) vocalizations of adult rats with arthritis (5 h postinduction of a kaolin-carrageenan monoarthritis in the knee) but did not affect spinal withdrawal thresholds, which is consistent with a supraspinal action. Stereotaxic administration of riluzole into the CeA by microdialysis (1 mM, concentration in the microdialysis fiber, 15 min) also inhibited vocalizations, confirming the CeA as a site of action of riluzole. Stereotaxic administration of a selective SK channel blocker (apamin, 1 µM, concentration in the microdialysis fiber, 15 min) into the CeA had no effect by itself but inhibited the effect of systemic riluzole on vocalizations. Off-site administration of apamin into the basolateral amygdala (BLA) as a placement control or stereotaxic application of a selective blocker of large-conductance calcium-activated potassium (BK) channels (charybdotoxin, 1 µM, concentration in the microdialysis fiber, 15 min) into the CeA did not affect the inhibitory effects of systemically applied riluzole. CONCLUSIONS The results suggest that riluzole can inhibit supraspinally organized pain behaviors in an arthritis model by activating SK, but not BK, channels in the amygdala (CeA but not BLA).
Collapse
Affiliation(s)
- Jeremy M Thompson
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, 3601 4th St, Lubbock, TX, 79430-6592, USA.
| | - Guangchen Ji
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, 3601 4th St, Lubbock, TX, 79430-6592, USA.
| | - Volker Neugebauer
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, 3601 4th St, Lubbock, TX, 79430-6592, USA.
- Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, Lubbock, TX, USA.
| |
Collapse
|
42
|
KCNN Genes that Encode Small-Conductance Ca2+-Activated K+ Channels Influence Alcohol and Drug Addiction. Neuropsychopharmacology 2015; 40:1928-39. [PMID: 25662840 PMCID: PMC4839516 DOI: 10.1038/npp.2015.42] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Revised: 01/17/2015] [Accepted: 01/26/2015] [Indexed: 02/02/2023]
Abstract
Small-conductance Ca(2+)-activated K(+) (KCa2) channels control neuronal excitability and synaptic plasticity, and have been implicated in substance abuse. However, it is unknown if genes that encode KCa2 channels (KCNN1-3) influence alcohol and drug addiction. In the present study, an integrative functional genomics approach shows that genetic datasets for alcohol, nicotine, and illicit drugs contain the family of KCNN genes. Alcohol preference and dependence QTLs contain KCNN2 and KCNN3, and Kcnn3 transcript levels in the nucleus accumbens (NAc) of genetically diverse BXD strains of mice predicted voluntary alcohol consumption. Transcript levels of Kcnn3 in the NAc negatively correlated with alcohol intake levels in BXD strains, and alcohol dependence enhanced the strength of this association. Microinjections of the KCa2 channel inhibitor apamin into the NAc increased alcohol intake in control C57BL/6J mice, while spontaneous seizures developed in alcohol-dependent mice following apamin injection. Consistent with this finding, alcohol dependence enhanced the intrinsic excitability of medium spiny neurons in the NAc core and reduced the function and protein expression of KCa2 channels in the NAc. Altogether, these data implicate the family of KCNN genes in alcohol, nicotine, and drug addiction, and identify KCNN3 as a mediator of voluntary and excessive alcohol consumption. KCa2.3 channels represent a promising novel target in the pharmacogenetic treatment of alcohol and drug addiction.
Collapse
|
43
|
Murthy SRK, Sherrin T, Jansen C, Nijholt I, Robles M, Dolga AM, Andreotti N, Sabatier JM, Knaus HG, Penner R, Todorovic C, Blank T. Small-conductance Ca2+-activated potassium type 2 channels regulate the formation of contextual fear memory. PLoS One 2015; 10:e0127264. [PMID: 25938421 PMCID: PMC4418695 DOI: 10.1371/journal.pone.0127264] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Accepted: 04/13/2015] [Indexed: 12/28/2022] Open
Abstract
Small-conductance, Ca2+ activated K+ channels (SK channels) are expressed at high levels in brain regions responsible for learning and memory. In the current study we characterized the contribution of SK2 channels to synaptic plasticity and to different phases of hippocampal memory formation. Selective SK2 antisense-treatment facilitated basal synaptic transmission and theta-burst induced LTP in hippocampal brain slices. Using the selective SK2 antagonist Lei-Dab7 or SK2 antisense probes, we found that hippocampal SK2 channels are critical during two different time windows: 1) blockade of SK2 channels before the training impaired fear memory, whereas, 2) blockade of SK2 channels immediately after the training enhanced contextual fear memory. We provided the evidence that the post-training cleavage of the SK2 channels was responsible for the observed bidirectional effect of SK2 channel blockade on memory consolidation. Thus, Lei-Dab7-injection before training impaired the C-terminal cleavage of SK2 channels, while Lei-Dab7 given immediately after training facilitated the C-terminal cleavage. Application of the synthetic peptide comprising a leucine-zipper domain of the C-terminal fragment to Jurkat cells impaired SK2 channel-mediated currents, indicating that the endogenously cleaved fragment might exert its effects on memory formation by blocking SK2 channel-mediated currents. Our present findings suggest that SK2 channel proteins contribute to synaptic plasticity and memory not only as ion channels but also by additionally generating a SK2 C-terminal fragment, involved in both processes. The modulation of fear memory by down-regulating SK2 C-terminal cleavage might have applicability in the treatment of anxiety disorders in which fear conditioning is enhanced.
Collapse
Affiliation(s)
- Saravana R. K. Murthy
- Section on Cellular Neurobiology, Program on Developmental Neuroscience, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland, United States of America
| | - Tessi Sherrin
- Department of Cell & Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, United States of America
| | - Chad Jansen
- Department of Cell & Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, United States of America
- Laboratory of Cell and Molecular Signaling, The Queen’s Medical Center, Honolulu, Hawaii, United States of America
| | | | - Michael Robles
- Department of Cell & Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, United States of America
| | - Amalia M. Dolga
- Department of Pharmacology and Toxicology, Philipps-University Marburg, Marburg, Germany
| | - Nicolas Andreotti
- Laboratoire INSERM UMR1097, Parc scientifique et technologique de Luminy, Marseille, cedex 09, France
| | - Jean-Marc Sabatier
- Laboratoire INSERM UMR1097, Parc scientifique et technologique de Luminy, Marseille, cedex 09, France
| | - Hans-Guenther Knaus
- Division for Molecular and Cellular Pharmacology, Medical University Innsbruck, Innsbruck, Austria
| | - Reinhold Penner
- Department of Cell & Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, United States of America
- Laboratory of Cell and Molecular Signaling, The Queen’s Medical Center, Honolulu, Hawaii, United States of America
| | - Cedomir Todorovic
- Department of Cell & Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, United States of America
- * E-mail:
| | - Thomas Blank
- Department of Cell & Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, United States of America
- Institute for Neuropathology, University of Freiburg, Freiburg, Germany
| |
Collapse
|
44
|
Aidi-Knani S, Pezard L, Mpari B, Ben Hamida J, Sabatier JM, Mourre C, Regaya I. Correspondences between the binding characteristics of a non-natural peptide, Lei-Dab7, and the distribution of SK subunits in the rat central nervous system. Eur J Pharmacol 2015; 752:106-11. [DOI: 10.1016/j.ejphar.2015.02.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Revised: 02/09/2015] [Accepted: 02/11/2015] [Indexed: 11/26/2022]
|
45
|
Trimmer JS. Subcellular localization of K+ channels in mammalian brain neurons: remarkable precision in the midst of extraordinary complexity. Neuron 2015; 85:238-56. [PMID: 25611506 DOI: 10.1016/j.neuron.2014.12.042] [Citation(s) in RCA: 155] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Potassium channels (KChs) are the most diverse ion channels, in part due to extensive combinatorial assembly of a large number of principal and auxiliary subunits into an assortment of KCh complexes. Their structural and functional diversity allows KChs to play diverse roles in neuronal function. Localization of KChs within specialized neuronal compartments defines their physiological role and also fundamentally impacts their activity, due to localized exposure to diverse cellular determinants of channel function. Recent studies in mammalian brain reveal an exquisite refinement of KCh subcellular localization. This includes axonal KChs at the initial segment, and near/within nodes of Ranvier and presynaptic terminals, dendritic KChs found at sites reflecting specific synaptic input, and KChs defining novel neuronal compartments. Painting the remarkable diversity of KChs onto the complex architecture of mammalian neurons creates an elegant picture of electrical signal processing underlying the sophisticated function of individual neuronal compartments, and ultimately neurotransmission and behavior.
Collapse
Affiliation(s)
- James S Trimmer
- Department of Neurobiology, Physiology, and Behavior, University of California, Davis, Davis, CA 95616, USA; Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA 95616, USA.
| |
Collapse
|
46
|
Ballesteros-Merino C, Martínez-Hernández J, Aguado C, Watanabe M, Adelman JP, Luján R. Localization of SK2 channels relative to excitatory synaptic sites in the mouse developing Purkinje cells. Front Neuroanat 2014; 8:154. [PMID: 25565979 PMCID: PMC4266016 DOI: 10.3389/fnana.2014.00154] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Accepted: 11/25/2014] [Indexed: 11/18/2022] Open
Abstract
Small-conductance, Ca2+-activated K+ (SK) channels regulate neuronal excitability in a variety of ways. To understand their roles in different neuronal subtypes it is important to determine their precise subcellular distribution. Here, we used biochemical, light microscopy immunohistochemical and immunoelectron microscopy techniques, combined with quantitative approaches, to reveal the expression and subcellular localization patterns of SK2 in the developing cerebellum. Using western blots, the SK2 protein showed a progressive increase during postnatal development. At the light microscopic level, SK2 immunoreactivity was very prominent in the developing Purkinje cells (PC), particularly in the molecular layer (ML). Electron microscopy revealed that throughout development SK2 was mostly detected at the extrasynaptic and perisynaptic plasma membrane of dendritic shafts and dendritic spines of PCs. However, there was some localization at axon terminals as well. Quantitative analyses and 3D reconstructions further revealed a progressive developmental change of SK2 on the surface of PCs from dendritic shafts to dendritic spines. Together, these results indicate that SK2 channels undergo dynamic spatial regulation during cerebellar development, and this process is associated with the formation and maturation of excitatory synaptic contacts to PCs.
Collapse
Affiliation(s)
- Carmen Ballesteros-Merino
- Instituto de Investigación en Discapacidades Neurológicas (IDINE), Departamento Ciencias Médicas, Facultad de Medicina, Universidad Castilla-La Mancha Albacete, Albacete, Spain
| | - José Martínez-Hernández
- Instituto de Investigación en Discapacidades Neurológicas (IDINE), Departamento Ciencias Médicas, Facultad de Medicina, Universidad Castilla-La Mancha Albacete, Albacete, Spain
| | - Carolina Aguado
- Instituto de Investigación en Discapacidades Neurológicas (IDINE), Departamento Ciencias Médicas, Facultad de Medicina, Universidad Castilla-La Mancha Albacete, Albacete, Spain
| | - Masahiko Watanabe
- Department of Anatomy, Hokkaido University School of Medicine Sapporo, Japan
| | - John P Adelman
- Vollum Institute, Oregon Health and Science University Portland, OR, USA
| | - Rafael Luján
- Instituto de Investigación en Discapacidades Neurológicas (IDINE), Departamento Ciencias Médicas, Facultad de Medicina, Universidad Castilla-La Mancha Albacete, Albacete, Spain
| |
Collapse
|
47
|
Church TW, Weatherall KL, Corrêa SAL, Prole DL, Brown JT, Marrion NV. Preferential assembly of heteromeric small conductance calcium-activated potassium channels. Eur J Neurosci 2014; 41:305-15. [PMID: 25421315 DOI: 10.1111/ejn.12789] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Revised: 10/16/2014] [Accepted: 10/20/2014] [Indexed: 11/27/2022]
Abstract
The activation of small conductance calcium-dependent (SK) channels regulates membrane excitability by causing membrane hyperpolarization. Three subtypes (SK1-3) have been cloned, with each subtype expressed within the nervous system. The locations of channel subunits overlap, with SK1 and SK2 subunits often expressed in the same brain region. We showed that expressed homomeric rat SK1 subunits did not form functional channels, because subunits accumulated in the Golgi. This raised the question of whether heteromeric channels could form with SK1 subunits. The co-expression of SK1 and SK2 subunits in HEK293 cells preferentially co-assembled to produce heteromeric channels with a fixed stoichiometry of alternating subunits. The expression in hippocampal CA1 neurons of mutant rat SK1 subunits [rat SK1(LV213/4YA)] that produced an apamin-sensitive current changed the amplitude and pharmacology of the medium afterhyperpolarization. The overexpression of rat SK1(LV213/4YA) subunits reduced the sensitivity of the medium afterhyperpolarization to apamin, substantiating the preferential co-assembly of SK1 and SK2 subunits to form heteromeric channels. Species-specific channel assembly occurred as the co-expression of human SK1 with rat SK2 did not form functional heteromeric channels. The replacement of two amino acids within the C-terminus of rat SK2 with those from human SK2 permitted the assembly of heteromeric channels when co-expressed with human SK1. These data showed that species-specific co-assembly was mediated by interaction between the C-termini of SK channel subunits. The finding that SK channels preferentially co-assembled to form heteromeric channels suggested that native heteromeric channels will predominate in cells expressing multiple SK channel subunits.
Collapse
Affiliation(s)
- Timothy W Church
- School of Physiology and Pharmacology, University of Bristol, Bristol, UK
| | | | | | | | | | | |
Collapse
|
48
|
Gymnopoulos M, Cingolani LA, Pedarzani P, Stocker M. Developmental mapping of small-conductance calcium-activated potassium channel expression in the rat nervous system. J Comp Neurol 2014; 522:1072-101. [PMID: 24096910 PMCID: PMC4016743 DOI: 10.1002/cne.23466] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Revised: 09/06/2013] [Accepted: 09/17/2013] [Indexed: 12/22/2022]
Abstract
Early electrical activity and calcium influx regulate crucial aspects of neuronal development. Small-conductance calcium-activated potassium (SK) channels regulate action potential firing and shape calcium influx through feedback regulation in mature neurons. These functions, observed in the adult nervous system, make them ideal candidates to regulate activity-and calcium-dependent processes in neurodevelopment. However, to date little is known about the onset of expression and regions expressing SK channel subunits in the embryonic and postnatal development of the central nervous system (CNS). To allow studies on the contribution of SK channels to different phases of development of single neurons and networks, we have performed a detailed in situ hybridization mapping study, providing comprehensive distribution profiles of all three SK subunits (SK1, SK2, and SK3) in the rat CNS during embryonic and postnatal development. SK channel transcripts are expressed at early stages of prenatal CNS development. The three SK channel subunits display different developmental expression gradients in distinct CNS regions, with time points of expression and up-or downregulation that can be associated with a range of diverse developmental events. Their early expression in embryonic development suggests an involvement of SK channels in the regulation of developmental processes. Additionally, this study shows how the postnatal ontogenetic patterns lead to the adult expression map for each SK channel subunit and how their coexpression in the same regions or neurons varies throughout development. J. Comp. Neurol. 522:1072–1101, 2014. © 2013 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Marco Gymnopoulos
- Department of Molecular Biology of Neuronal Signals, Max Planck Institute for Experimental Medicine, 37075, Göttingen, Germany
| | | | | | | |
Collapse
|
49
|
Egorova PA, Karelina TV, Vlasova OL, Antonov SM, Bezprozvanny IB. The effect of SK channel modulators on the simple spike firing frequency in discharge of cerebellar Purkinje cells in laboratory mice. J EVOL BIOCHEM PHYS+ 2014. [DOI: 10.1134/s0022093014020045] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
50
|
Pachuau J, Li DP, Chen SR, Lee HA, Pan HL. Protein kinase CK2 contributes to diminished small conductance Ca2+-activated K+ channel activity of hypothalamic pre-sympathetic neurons in hypertension. J Neurochem 2014; 130:657-67. [PMID: 24806793 DOI: 10.1111/jnc.12758] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Revised: 05/05/2014] [Accepted: 05/06/2014] [Indexed: 11/29/2022]
Abstract
Small conductance calcium-activated K(+) (SK) channels regulate neuronal excitability. However, little is known about changes in SK channel activity of pre-sympathetic neurons in the hypothalamic paraventricular nucleus (PVN) in essential hypertension. SK channels, calmodulin, and casein kinase II (CK2) form a molecular complex. Because CK2 is up-regulated in the PVN in spontaneously hypertensive rats (SHRs), we hypothesized that CK2 increases calmodulin phosphorylation and contributes to diminished SK channel activity in PVN pre-sympathetic neurons in SHRs. Perforated whole-cell recordings were performed on retrogradely labeled spinally projecting PVN neurons in Wistar-Kyoto (WKY) rats and SHRs. Blocking SK channels with apamin significantly increased the firing rate of PVN neurons in WKY rats but not in SHRs. CK2 inhibition restored the stimulatory effect of apamin on the firing activity of PVN neurons in SHRs. Furthermore, apamin-sensitive SK currents and depolarization-induced medium after-hyperpolarization potentials of PVN neurons were significantly larger in WKY rats than in SHRs. CK2 inhibition significantly increased the SK channel current and medium after-depolarization potential of PVN neurons in SHRs. In addition, CK2-mediated calmodulin phosphorylation level in the PVN was significantly higher in SHRs than in WKY rats. Although SK3 was detected in the PVN, its expression level did not differ significantly between SHRs and WKY rats. Our findings suggest that CK2-mediated calmodulin phosphorylation is increased and contributes to diminished SK channel function of PVN pre-sympathetic neurons in SHRs. This information advances our understanding of the mechanisms underlying hyperactivity of PVN pre-sympathetic neurons and increased sympathetic vasomotor tone in hypertension. Small conductance calcium-activated K(+) (SK) channels, calmodulin, and protein kinase CK2 form a molecular complex and regulate neuronal excitability. Our study suggests that augmented CK2 activity in hypertension can increase calmodulin (CaM) phosphorylation, which leads to diminished SK channel function in pre-sympathetic neurons. Diminished SK channel activity plays a role in hyperactivity of pre-sympathetic neurons in the hypothalamus in hypertension.
Collapse
Affiliation(s)
- Judith Pachuau
- Center for Neuroscience and Pain Research, Department of Anesthesiology and Perioperative Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | | | | | | |
Collapse
|