1
|
Lee SY, Shin J, Kwon MJ, Kim Y, Ho WK, Lee SH. Kv4.2 Regulates Basal Synaptic Strength by Inhibiting R-Type Calcium Channels in the Hippocampus. J Neurosci 2025; 45:e0444242025. [PMID: 39933929 PMCID: PMC11924881 DOI: 10.1523/jneurosci.0444-24.2025] [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: 03/07/2024] [Revised: 01/15/2025] [Accepted: 01/17/2025] [Indexed: 02/13/2025] Open
Abstract
Kv4.2 subunits, which mediate transient A-type K+ current, are crucial in regulating neuronal excitability and synaptic responses within the hippocampus. While their contribution to activity-dependent regulation of synaptic response is well-established, the impact of Kv4.2 on basal synaptic strength remains elusive. To address this gap, we introduced a Kv4.2-specific antibody (anti-Kv4.2) into hippocampal neurons of mice of both sexes to selectively inhibit postsynaptic Kv4.2, enabling direct examination of its impact on excitatory postsynaptic potentials (EPSPs) and currents (EPSCs) during basal synaptic activity. Our results demonstrated that blocking Kv4.2 significantly enhanced the amplitude of EPSPs. This amplification was proportional to the increase in the amplitude of EPSCs, which, in turn, correlated with the expression level of Kv4.2 in the dendritic regions of the hippocampus. Furthermore, the anti-Kv4.2-induced increase in EPSC amplitude was associated with a decrease in the failure rate of EPSCs evoked by minimal stimulation, suggesting that blocking Kv4.2 facilitates the recruitment of AMPA receptors to both silent and functional synapses to enhance synaptic efficacy. The anti-Kv4.2-induced synaptic potentiation was effectively abolished by intracellular 10 mM BAPTA or by blocking R-type calcium channels (RTCCs) and downstream signaling molecules, including protein kinases A and C. Importantly, Kv4.2 inhibition did not occlude further synaptic potentiation induced by high-frequency stimulation, suggesting that anti-Kv4.2-induced synaptic strengthening involves unique mechanisms that are distinct from long-term potentiation pathways. Taken together, these findings underscore the essential role of Kv4.2 in the regulation of basal synaptic strength, which is mediated by the inhibition of RTCCs.
Collapse
Affiliation(s)
- Seung Yeon Lee
- Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Jiwoo Shin
- Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
- Department of Brain and Cognitive Sciences, College of Natural Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Min Jeong Kwon
- Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Yujin Kim
- Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
- Neuroscience Research Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Won-Kyung Ho
- Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Suk-Ho Lee
- Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
- Department of Brain and Cognitive Sciences, College of Natural Sciences, Seoul National University, Seoul 08826, Republic of Korea
- Neuroscience Research Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| |
Collapse
|
2
|
Incontro S, Musella ML, Sammari M, Di Scala C, Fantini J, Debanne D. Lipids shape brain function through ion channel and receptor modulations: physiological mechanisms and clinical perspectives. Physiol Rev 2025; 105:137-207. [PMID: 38990068 DOI: 10.1152/physrev.00004.2024] [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: 01/16/2024] [Revised: 05/28/2024] [Accepted: 07/01/2024] [Indexed: 07/12/2024] Open
Abstract
Lipids represent the most abundant molecular type in the brain, with a fat content of ∼60% of the dry brain weight in humans. Despite this fact, little attention has been paid to circumscribe the dynamic role of lipids in brain function and disease. Membrane lipids such as cholesterol, phosphoinositide, sphingolipids, arachidonic acid, and endocannabinoids finely regulate both synaptic receptors and ion channels that ensure critical neural functions. After a brief introduction on brain lipids and their respective properties, we review here their role in regulating synaptic function and ion channel activity, action potential propagation, neuronal development, and functional plasticity and their contribution in the development of neurological and neuropsychiatric diseases. We also provide possible directions for future research on lipid function in brain plasticity and diseases.
Collapse
Affiliation(s)
| | | | - Malika Sammari
- UNIS, INSERM, Aix-Marseille Université, Marseille, France
| | | | | | | |
Collapse
|
3
|
Sanz-Gálvez R, Falardeau D, Kolta A, Inglebert Y. The role of astrocytes from synaptic to non-synaptic plasticity. Front Cell Neurosci 2024; 18:1477985. [PMID: 39493508 PMCID: PMC11527691 DOI: 10.3389/fncel.2024.1477985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 10/02/2024] [Indexed: 11/05/2024] Open
Abstract
Information storage and transfer in the brain require a high computational power. Neuronal network display various local or global mechanisms to allow information storage and transfer in the brain. From synaptic to intrinsic plasticity, the rules of input-output function modulation have been well characterized in neurons. In the past years, astrocytes have been suggested to increase the computational power of the brain and we are only just starting to uncover their role in information processing. Astrocytes maintain a close bidirectional communication with neurons to modify neuronal network excitability, transmission, axonal conduction, and plasticity through various mechanisms including the release of gliotransmitters or local ion homeostasis. Astrocytes have been significantly studied in the context of long-term or short-term synaptic plasticity, but this is not the only mechanism involved in memory formation. Plasticity of intrinsic neuronal excitability also participates in memory storage through regulation of voltage-gated ion channels or axonal morphological changes. Yet, the contribution of astrocytes to these other forms of non-synaptic plasticity remains to be investigated. In this review, we summarized the recent advances on the role of astrocytes in different forms of plasticity and discuss new directions and ideas to be explored regarding astrocytes-neuronal communication and regulation of plasticity.
Collapse
Affiliation(s)
- Rafael Sanz-Gálvez
- Department of Neurosciences, Université de Montréal, Montréal, QC, Canada
- Centre Interdisciplinaire de Recherche sur le Cerveau et l’Apprentissage (CIRCA), Montréal, QC, Canada
| | - Dominic Falardeau
- Department of Neurosciences, Université de Montréal, Montréal, QC, Canada
- Centre Interdisciplinaire de Recherche sur le Cerveau et l’Apprentissage (CIRCA), Montréal, QC, Canada
| | - Arlette Kolta
- Department of Neurosciences, Université de Montréal, Montréal, QC, Canada
- Centre Interdisciplinaire de Recherche sur le Cerveau et l’Apprentissage (CIRCA), Montréal, QC, Canada
- Department of Stomatology, Université de Montréal, Montréal, QC, Canada
| | - Yanis Inglebert
- Department of Neurosciences, Université de Montréal, Montréal, QC, Canada
- Centre Interdisciplinaire de Recherche sur le Cerveau et l’Apprentissage (CIRCA), Montréal, QC, Canada
| |
Collapse
|
4
|
Esaki H, Izumi S, Nishikawa K, Nagayasu K, Kaneko S, Nishitani N, Deyama S, Kaneda K. Role of medial prefrontal cortex voltage-dependent potassium 4.3 channels in nicotine-induced enhancement of object recognition memory in male mice. Eur J Pharmacol 2024; 978:176790. [PMID: 38942263 DOI: 10.1016/j.ejphar.2024.176790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 06/26/2024] [Indexed: 06/30/2024]
Abstract
Nicotine has been shown to enhance object recognition memory in the novel object recognition (NOR) test by activating excitatory neurons in the medial prefrontal cortex (mPFC). However, the exact neuronal mechanisms underlying the nicotine-induced activation of mPFC neurons and the resultant memory enhancement remain poorly understood. To address this issue, we performed brain-slice electrophysiology and the NOR test in male C57BL/6J mice. Whole-cell patch-clamp recordings from layer V pyramidal neurons in the mPFC revealed that nicotine augments the summation of evoked excitatory postsynaptic potentials (eEPSPs) and that this effect was suppressed by N-[3,5-Bis(trifluoromethyl)phenyl]-N'-[2,4-dibromo-6-(2H-tetrazol-5-yl)phenyl]urea (NS5806), a voltage-dependent potassium (Kv) 4.3 channel activator. In line with these findings, intra-mPFC infusion of NS5806 suppressed systemically administered nicotine-induced memory enhancement in the NOR test. Additionally, miRNA-mediated knockdown of Kv4.3 channels in mPFC pyramidal neurons enhanced object recognition memory. Furthermore, inhibition of A-type Kv channels by intra-mPFC infusion of 4-aminopyridine was found to enhance object recognition memory, while this effect was abrogated by prior intra-mPFC NS5806 infusion. These results suggest that nicotine augments the summation of eEPSPs via the inhibition of Kv4.3 channels in mPFC layer V pyramidal neurons, resulting in the enhancement of object recognition memory.
Collapse
Affiliation(s)
- Hirohito Esaki
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Shoma Izumi
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Keisuke Nishikawa
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Kazuki Nagayasu
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida-Shimoadachi-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Shuji Kaneko
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida-Shimoadachi-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Naoya Nishitani
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Satoshi Deyama
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Katsuyuki Kaneda
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan.
| |
Collapse
|
5
|
Abstract
Repetitive transcranial magnetic stimulation (rTMS) has become an increasingly popular tool to modulate neural excitability and induce neural plasticity in clinical and preclinical models; however, the physiological mechanisms in which it exerts these effects remain largely unknown. To date, studies have primarily focused on characterizing rTMS-induced changes occurring at the synapse, with little attention given to changes in intrinsic membrane properties. However, accumulating evidence suggests that rTMS may induce its effects, in part, via intrinsic plasticity mechanisms, suggesting a new and potentially complementary understanding of how rTMS alters neural excitability and neural plasticity. In this review, we provide an overview of several intrinsic plasticity mechanisms before reviewing the evidence for rTMS-induced intrinsic plasticity. In addition, we discuss a select number of neurological conditions where rTMS-induced intrinsic plasticity has therapeutic potential before speculating on the temporal relationship between rTMS-induced intrinsic and synaptic plasticity.
Collapse
Affiliation(s)
- Emily S King
- Experimental and Regenerative Neurosciences, School of Biological Sciences, The University of Western Australia, Perth, Australia
- Perron Institute for Neurological and Translational Science, Perth, Australia
| | - Alexander D Tang
- Experimental and Regenerative Neurosciences, School of Biological Sciences, The University of Western Australia, Perth, Australia
- Perron Institute for Neurological and Translational Science, Perth, Australia
| |
Collapse
|
6
|
Francis-Oliveira J, Higa GSV, Viana FJC, Cruvinel E, Carlos-Lima E, da Silva Borges F, Zampieri TT, Rebello FP, Ulrich H, De Pasquale R. TREK-1 inhibition promotes synaptic plasticity in the prelimbic cortex. Exp Neurol 2024; 373:114652. [PMID: 38103709 DOI: 10.1016/j.expneurol.2023.114652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 11/28/2023] [Accepted: 12/10/2023] [Indexed: 12/19/2023]
Abstract
Synaptic plasticity is one of the putative mechanisms involved in the maturation of the prefrontal cortex (PFC) during postnatal development. Early life stress (ELS) affects the shaping of cortical circuitries through impairment of synaptic plasticity supporting the onset of mood disorders. Growing evidence suggests that dysfunctional postnatal maturation of the prelimbic division (PL) of the PFC might be related to the emergence of depression. The potassium channel TREK-1 has attracted particular interest among many factors that modulate plasticity, concerning synaptic modifications that could underlie mood disorders. Studies have found that ablation of TREK-1 increases the resilience to depression, while rats exposed to ELS exhibit higher TREK-1 levels in the PL. TREK-1 is regulated by multiple intracellular transduction pathways including the ones activated by metabotropic receptors. In the hippocampal neurons, TREK-1 interacts with the serotonergic system, one of the main factors involved in the action of antidepressants. To investigate possible mechanisms related to the antidepressant role of TREK-1, we used brain slice electrophysiology to evaluate the effects of TREK-1 pharmacological blockade on synaptic plasticity at PL circuitry. We extended this investigation to animals subjected to ELS. Our findings suggest that in non-stressed animals, TREK-1 activity is required for the reduction of synaptic responses mediated by the 5HT1A receptor activation. Furthermore, we demonstrate that TREK-1 blockade promotes activity-dependent long-term depression (LTD) when acting in synergy with 5HT1A receptor stimulation. On the other hand, in ELS animals, TREK-1 blockade reduces synaptic transmission and facilitates LTD expression. These results indicate that TREK-1 inhibition stimulates synaptic plasticity in the PL and this effect is more pronounced in animals subjected to ELS during postnatal development.
Collapse
Affiliation(s)
- José Francis-Oliveira
- Laboratório de Neurofisiologia, Departamento de Fisiologia e Biofísica, Universidade de São Paulo, Butantã, SP 05508-000, Brazil; Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Guilherme Shigueto Vilar Higa
- Laboratório de Neurofisiologia, Departamento de Fisiologia e Biofísica, Universidade de São Paulo, Butantã, SP 05508-000, Brazil; Departamento de Bioquímica, Instituto de Química (USP), Butantã, SP 05508-900, Brazil; Laboratório de Neurogenética, Universidade Federal do ABC, São Bernardo do Campo, SP 09210-580, Brazil
| | - Felipe José Costa Viana
- Laboratório de Neurofisiologia, Departamento de Fisiologia e Biofísica, Universidade de São Paulo, Butantã, SP 05508-000, Brazil
| | - Emily Cruvinel
- Laboratório de Neurofisiologia, Departamento de Fisiologia e Biofísica, Universidade de São Paulo, Butantã, SP 05508-000, Brazil
| | - Estevão Carlos-Lima
- Laboratório de Neurofisiologia, Departamento de Fisiologia e Biofísica, Universidade de São Paulo, Butantã, SP 05508-000, Brazil
| | - Fernando da Silva Borges
- Department of Physiology & Pharmacology, SUNY Downstate Health Sciences University, Brooklyn, NY 11203, USA
| | - Thais Tessari Zampieri
- Laboratório de Neurofisiologia, Departamento de Fisiologia e Biofísica, Universidade de São Paulo, Butantã, SP 05508-000, Brazil
| | - Fernanda Pereira Rebello
- Laboratório de Neurofisiologia, Departamento de Fisiologia e Biofísica, Universidade de São Paulo, Butantã, SP 05508-000, Brazil
| | - Henning Ulrich
- Departamento de Bioquímica, Instituto de Química (USP), Butantã, SP 05508-900, Brazil
| | - Roberto De Pasquale
- Laboratório de Neurofisiologia, Departamento de Fisiologia e Biofísica, Universidade de São Paulo, Butantã, SP 05508-000, Brazil.
| |
Collapse
|
7
|
AlShammari AK, Abd El-Aziz TM, Al-Sabi A. Snake Venom: A Promising Source of Neurotoxins Targeting Voltage-Gated Potassium Channels. Toxins (Basel) 2023; 16:12. [PMID: 38251229 PMCID: PMC10820993 DOI: 10.3390/toxins16010012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 12/11/2023] [Accepted: 12/19/2023] [Indexed: 01/23/2024] Open
Abstract
The venom derived from various sources of snakes represents a vast collection of predominantly protein-based toxins that exhibit a wide range of biological actions, including but not limited to inflammation, pain, cytotoxicity, cardiotoxicity, and neurotoxicity. The venom of a particular snake species is composed of several toxins, while the venoms of around 600 venomous snake species collectively encompass a substantial reservoir of pharmacologically intriguing compounds. Despite extensive research efforts, a significant portion of snake venoms remains uncharacterized. Recent findings have demonstrated the potential application of neurotoxins derived from snake venom in selectively targeting voltage-gated potassium channels (Kv). These neurotoxins include BPTI-Kunitz polypeptides, PLA2 neurotoxins, CRISPs, SVSPs, and various others. This study provides a comprehensive analysis of the existing literature on the significance of Kv channels in various tissues, highlighting their crucial role as proteins susceptible to modulation by diverse snake venoms. These toxins have demonstrated potential as valuable pharmacological resources and research tools for investigating the structural and functional characteristics of Kv channels.
Collapse
Affiliation(s)
- Altaf K. AlShammari
- College of Engineering and Technology, American University of the Middle East, Egaila 54200, Kuwait;
| | - Tarek Mohamed Abd El-Aziz
- Zoology Department, Faculty of Science, Minia University, El-Minia 61519, Egypt
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Ahmed Al-Sabi
- College of Engineering and Technology, American University of the Middle East, Egaila 54200, Kuwait;
| |
Collapse
|
8
|
Esaki H, Deyama S, Izumi S, Katsura A, Nishikawa K, Nishitani N, Kaneda K. Varenicline enhances recognition memory via α7 nicotinic acetylcholine receptors in the medial prefrontal cortex in male mice. Neuropharmacology 2023; 239:109672. [PMID: 37506875 DOI: 10.1016/j.neuropharm.2023.109672] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 07/20/2023] [Accepted: 07/24/2023] [Indexed: 07/30/2023]
Abstract
Previous studies postulated that chronic administration of varenicline, a partial and full agonist at α4β2 and α7 nicotinic acetylcholine receptors (nAChRs), respectively, enhances recognition memory. However, whether its acute administration is effective, on which brain region(s) it acts, and in what signaling it is involved, remain unknown. To address these issues, we conducted a novel object recognition test using male C57BL/6J mice, focusing on the medial prefrontal cortex (mPFC), a brain region associated with nicotine-induced enhancement of recognition memory. Systemic administration of varenicline before the training dose-dependently enhanced recognition memory. Intra-mPFC varenicline infusion also enhanced recognition memory, and this enhancement was blocked by intra-mPFC co-infusion of a selective α7, but not α4β2, nAChR antagonist. Consistent with this, intra-mPFC infusion of a selective α7 nAChR agonist augmented object recognition memory. Furthermore, intra-mPFC co-infusion of U-73122, a phospholipase C (PLC) inhibitor, or 2-aminoethoxydiphenylborane (2-APB), an inositol trisphosphate (IP3) receptor inhibitor, suppressed the varenicline-induced memory enhancement, suggesting that α7 nAChRs may also act as Gq-coupled metabotropic receptors. Additionally, whole-cell recordings from mPFC layer V pyramidal neurons in vitro revealed that varenicline significantly increased the summation of evoked excitatory postsynaptic potentials, and this effect was suppressed by U-73122 or 2-APB. These findings suggest that varenicline might acutely enhance recognition memory via mPFC α7 nAChR stimulation, followed by mPFC neuronal excitation, which is mediated by the activation of PLC and IP3 receptor signaling. Our study provides evidence supporting the potential repositioning of varenicline as a treatment for cognitive impairment.
Collapse
Affiliation(s)
- Hirohito Esaki
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Satoshi Deyama
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Shoma Izumi
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Ayano Katsura
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Keisuke Nishikawa
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Naoya Nishitani
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Katsuyuki Kaneda
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan.
| |
Collapse
|
9
|
Bavan S, Goodkin HP, Papazian DM. Altered Closed State Inactivation Gating in Kv4.2 Channels Results in Developmental and Epileptic Encephalopathies in Human Patients. Hum Mutat 2022; 43:1286-1298. [PMID: 35510384 DOI: 10.1002/humu.24396] [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: 09/21/2021] [Revised: 04/27/2022] [Accepted: 05/01/2022] [Indexed: 11/06/2022]
Abstract
Kv4.2 subunits, encoded by KCND2, serve as the pore-forming components of voltage-gated, inactivating ISA K+ channels expressed in the brain. ISA channels inactivate without opening in response to subthreshold excitatory input, temporarily increasing neuronal excitability, the back propagation of action potentials, and Ca2+ influx into dendrites, thereby regulating mechanisms of spike timing-dependent synaptic plasticity. As previously described, a de novo variant in Kv4.2, p.Val404Met, is associated with an infant-onset developmental and epileptic encephalopathy (DEE) in monozygotic twin boys. The p.Val404Met variant enhances inactivation directly from closed states, but dramatically impairs inactivation after channel opening. We now report the identification of a closely related, novel, de novo variant in Kv4.2, p.Val402Leu, in a boy with an early-onset pharmacoresistant epilepsy that evolved to an epileptic aphasia syndrome (Continuous Spike Wave during Sleep Syndrome). Like p.Val404Met, the p.Val402Leu variant increases the rate of inactivation from closed states, but significantly slows inactivation after the pore opens. Although quantitatively the p.Val402Leu mutation alters channel kinetics less dramatically than p.Val404Met, our results strongly support the conclusion that p.Val402Leu and p.Val404Met cause the clinical features seen in the affected individuals and underscore the importance of closed state inactivation in ISA channels in normal brain development and function. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Selvan Bavan
- Department of Physiology, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095-1571.,Labcorp Drug Development, Huntingdon, PE28 4HS, UK
| | - Howard P Goodkin
- Departments of Neurology and Pediatrics, University of Virginia School of Medicine, Charlottesville, VA, 22903
| | - Diane M Papazian
- Department of Physiology, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095-1571
| |
Collapse
|
10
|
Vallejos MJ, Eadaim A, Hahm ET, Tsunoda S. Age-related changes in Kv4/Shal and Kv1/Shaker expression in Drosophila and a role for reactive oxygen species. PLoS One 2021; 16:e0261087. [PMID: 34932577 PMCID: PMC8691634 DOI: 10.1371/journal.pone.0261087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 11/23/2021] [Indexed: 11/19/2022] Open
Abstract
Age-related changes in ion channel expression are likely to affect neuronal signaling. Here, we examine how age affects Kv4/Shal and Kv1/Shaker K+ channel protein levels in Drosophila. We show that Kv4/Shal protein levels decline sharply from 3 days to 10 days, then more gradually from 10 to 40 days after eclosion. In contrast, Kv1/Shaker protein exhibits a transient increase at 10 days that then stabilizes and eventually declines at 40 days. We present data that begin to show a relationship between reactive oxygen species (ROS), Kv4/Shal, and locomotor performance. We show that Kv4/Shal levels are negatively affected by ROS, and that over-expression of Catalase or RNAi knock-down of the ROS-generating enzyme, Nicotinamide Adenine Dinucleotide Phosphate (NADPH) Oxidase (NOX), can attenuate the loss of Kv4/Shal protein. Finally, we compare levels of Kv4.2 and Kv4.3 in the hippocampus, olfactory bulb, cerebellum, and motor cortex of mice aged 6 weeks and 1 year. While there was no global decline in Kv4.2/4.3 that parallels what we report in Drosophila, we did find that Kv4.2/4.3 are differentially affected in various brain regions; this survey of changes may help inform mammalian studies that examine neuronal function with age.
Collapse
Affiliation(s)
- Maximiliano J. Vallejos
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, United States of America
| | - Abdunaser Eadaim
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, United States of America
| | - Eu-Teum Hahm
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, United States of America
| | - Susan Tsunoda
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, United States of America
- * E-mail:
| |
Collapse
|
11
|
Zhang Y, Tachtsidis G, Schob C, Koko M, Hedrich UBS, Lerche H, Lemke JR, Haeringen A, Ruivenkamp C, Prescott T, Tveten K, Gerstner T, Pruniski B, DiTroia S, VanNoy GE, Rehm HL, McLaughlin H, Bolz HJ, Zechner U, Bryant E, McDonough T, Kindler S, Bähring R. KCND2 variants associated with global developmental delay differentially impair Kv4.2 channel gating. Hum Mol Genet 2021; 30:2300-2314. [PMID: 34245260 DOI: 10.1093/hmg/ddab192] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 07/06/2021] [Accepted: 07/07/2021] [Indexed: 11/13/2022] Open
Abstract
Here, we report on six unrelated individuals, all presenting with early-onset global developmental delay, associated with impaired motor, speech and cognitive development, partly with developmental epileptic encephalopathy and physical dysmorphisms. All individuals carry heterozygous missense variants of KCND2, which encodes the voltage-gated potassium (Kv) channel α-subunit Kv4.2. The amino acid substitutions associated with the variants, p.(Glu323Lys) (E323K), p.(Pro403Ala) (P403A), p.(Val404Leu) (V404L) and p.(Val404Met) (V404M), affect sites known to be critical for channel gating. To unravel their likely pathogenicity, recombinant mutant channels were studied in the absence and presence of auxiliary β-subunits under two-electrode voltage-clamp in Xenopus oocytes. All channel mutants exhibited slowed and incomplete macroscopic inactivation, and the P403A variant in addition slowed activation. Co-expression of KChIP2 or DPP6 augmented the functional expression of both wild-type and mutant channels, however, the auxiliary β-subunit-mediated gating modifications differed from wild-type and among mutants. To simulate the putative setting in the affected individuals, heteromeric Kv4.2 channels (wild-type + mutant) were studied as ternary complexes (containing both KChIP2 and DPP6). In the heteromeric ternary configuration, the E323K variant exhibited only marginal functional alterations compared to homomeric wild-type ternary, compatible with mild loss-of-function. By contrast, the P403A, V404L and V404M variants displayed strong gating impairment in the heteromeric ternary configuration, compatible with loss or gain-of-function. Our results support the etiological involvement of Kv4.2 channel gating impairment in early-onset monogenic global developmental delay. In addition, they suggest that gain-of-function mechanisms associated with a substitution of V404 increase epileptic seizure susceptibility.
Collapse
Affiliation(s)
- Yongqiang Zhang
- Institute for Cellular and Integrative Physiology, Center for Experimental Medicine, University Hospital Hamburg-Eppendorf, Hamburg, Germany.,Southeast University, Nanjing, China
| | - Georgios Tachtsidis
- Institute for Cellular and Integrative Physiology, Center for Experimental Medicine, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Claudia Schob
- Institute for Human Genetics, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Mahmoud Koko
- Department of Neurology and Epileptology, Hertie-Institute for Clinical Brain Research, Tübingen, Germany
| | - Ulrike B S Hedrich
- Department of Neurology and Epileptology, Hertie-Institute for Clinical Brain Research, Tübingen, Germany
| | - Holger Lerche
- Department of Neurology and Epileptology, Hertie-Institute for Clinical Brain Research, Tübingen, Germany
| | - Johannes R Lemke
- University Center for Rare Diseases, Institute for Human Genetics, University Hospital, Leipzig, Germany
| | - Arie Haeringen
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Claudia Ruivenkamp
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Trine Prescott
- Department of Medical Genetics, Telemark Hospital Trust, Skien, Norway
| | - Kristian Tveten
- Department of Medical Genetics, Telemark Hospital Trust, Skien, Norway
| | - Thorsten Gerstner
- Department of Child Neurology and Rehabilitation and Department of Pediatrics, Hospital of Southern Norway, Arendal, Norway
| | - Brianna Pruniski
- Division of Genetics & Metabolism, Phoenix Children's Medical Group, Phoenix, AZ, USA
| | - Stephanie DiTroia
- Center for Mendelian Genomics and Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Grace E VanNoy
- Center for Mendelian Genomics and Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Heidi L Rehm
- Center for Mendelian Genomics and Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Hanno J Bolz
- Senckenberg Centre for Human Genetics, Frankfurt/Main, Germany.,Institute of Human Genetics, University Hospital of Cologne, Cologne, Germany
| | - Ulrich Zechner
- Senckenberg Centre for Human Genetics, Frankfurt/Main, Germany.,Institute of Human Genetics, University Medical Center Mainz, Mainz, Germany
| | - Emily Bryant
- Ann & Robert H Lurie Children's Hospital of Chicago, Department of Pediatrics, Northwestern University Feinberg Scool of Medicine, Chicago, IL, USA
| | - Tiffani McDonough
- Ann & Robert H Lurie Children's Hospital of Chicago, Department of Pediatrics, Northwestern University Feinberg Scool of Medicine, Chicago, IL, USA
| | - Stefan Kindler
- Institute for Human Genetics, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Robert Bähring
- Institute for Cellular and Integrative Physiology, Center for Experimental Medicine, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
12
|
Hegemann RU, Abraham WC. Postsynaptic cell firing triggers bidirectional metaplasticity depending on the LTP induction protocol. J Neurophysiol 2021; 125:1624-1635. [PMID: 33760659 DOI: 10.1152/jn.00514.2020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Cell firing has been reported to variably upregulate or downregulate subsequently induced long-term potentiation (LTP). The aim of this study was to elucidate the parameters critical to driving each direction of the metaplasticity effect. The main focus was on the commonly used θ-burst stimulation (TBS) and high-frequency stimulation (HFS) protocols that are known to trigger distinct intracellular signaling cascades. To study action potential (AP)-induced metaplasticity, we used intracellular recordings from CA1 pyramidal cells of rat hippocampal slices. Somatic current injections were used to induce θ-burst firing (TBF) or high-frequency firing (HFF) for priming purposes, whereas LTP was induced 15 min later via TBS of Schaffer collaterals in stratum radiatum. TBS-LTP was inhibited by both priming protocols. Conversely, HFS-LTP was facilitated by HFF priming but not affected by TBF priming. Interestingly, both priming protocols reduced AP firing during TBS-LTP induction, and this effect correlated with the reduction of TBS-LTP. However, LTP was not rescued by restoring AP firing with somatic current injections during the TBS. Analysis of intrinsic properties revealed few changes, apart from a priming-induced increase in the medium afterhyperpolarization (HFF priming) and a decrease in the EPSP amplitude/slope ratio (TBF priming), which could in principle contribute to the inhibition of TBS-LTP by reducing depolarization and associated Ca2+ influx following synaptic activity or AP backpropagation. Overall, these data indicate that the more physiological TBS protocol for inducing LTP is particularly susceptible to homeostatic feedback inhibition by prior bouts of postsynaptic cell firing.NEW & NOTEWORTHY The induction of LTP in the hippocampus was bidirectionally regulated by prior postsynaptic cell firing, with θ-burst stimulation-induced LTP being consistently impaired by prior spiking, whereas high-frequency stimulation-induced LTP was either not changed or facilitated. Reductions in cell firing during LTP induction did not explain the LTP impairment. Overall, different patterns of postsynaptic firing induce distinct intracellular changes that can increase or decrease LTP depending on the induction protocol.
Collapse
Affiliation(s)
- Regina U Hegemann
- Department of Psychology, Brain Health Research Centre and Brain Research New Zealand, University of Otago, Dunedin, New Zealand
| | - Wickliffe C Abraham
- Department of Psychology, Brain Health Research Centre and Brain Research New Zealand, University of Otago, Dunedin, New Zealand
| |
Collapse
|
13
|
Seifert C, Storch S, Bähring R. Modulation of Kv4.2/KChIP3 interaction by the ceroid lipofuscinosis neuronal 3 protein CLN3. J Biol Chem 2020; 295:12099-12110. [PMID: 32641494 PMCID: PMC7443505 DOI: 10.1074/jbc.ra120.013828] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 06/22/2020] [Indexed: 11/06/2022] Open
Abstract
Voltage-gated potassium (Kv) channels of the Kv4 subfamily associate with Kv channel-interacting proteins (KChIPs), which leads to enhanced surface expression and shapes the inactivation gating of these channels. KChIP3 has been reported to also interact with the late endosomal/lysosomal membrane glycoprotein CLN3 (ceroid lipofuscinosis neuronal 3), which is modified because of gene mutation in juvenile neuronal ceroid lipofuscinosis (JNCL). The present study was undertaken to find out whether and how CLN3, by its interaction with KChIP3, may indirectly modulate Kv4.2 channel expression and function. To this end, we expressed KChIP3 and CLN3, either individually or simultaneously, together with Kv4.2 in HEK 293 cells. We performed co-immunoprecipitation experiments and found a lower amount of KChIP3 bound to Kv4.2 in the presence of CLN3. In whole-cell patch-clamp experiments, we examined the effects of CLN3 co-expression on the KChIP3-mediated modulation of Kv4.2 channels. Simultaneous co-expression of CLN3 and KChIP3 with Kv4.2 resulted in a suppression of the typical KChIP3-mediated modulation; i.e. we observed less increase in current density, less slowing of macroscopic current decay, less acceleration of recovery from inactivation, and a less positively shifted voltage dependence of steady-state inactivation. The suppression of the KChIP3-mediated modulation of Kv4.2 channels was weaker for the JNCL-related missense mutant CLN3R334C and for a JNCL-related C-terminal deletion mutant (CLN3ΔC). Our data support the notion that CLN3 is involved in Kv4.2/KChIP3 somatodendritic A-type channel formation, trafficking, and function, a feature that may be lost in JNCL.
Collapse
Affiliation(s)
- Carolin Seifert
- Institut für Zelluläre und Integrative Physiologie, Zentrum für Experimentelle Medizin, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Stephan Storch
- Klinik und Poliklinik für Kinder- und Jugendmedizin, Pädiatrische Forschung, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Robert Bähring
- Institut für Zelluläre und Integrative Physiologie, Zentrum für Experimentelle Medizin, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
14
|
Aceto G, Re A, Mattera A, Leone L, Colussi C, Rinaudo M, Scala F, Gironi K, Barbati SA, Fusco S, Green T, Laezza F, D'Ascenzo M, Grassi C. GSK3β Modulates Timing-Dependent Long-Term Depression Through Direct Phosphorylation of Kv4.2 Channels. Cereb Cortex 2020; 29:1851-1865. [PMID: 29790931 DOI: 10.1093/cercor/bhy042] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 01/15/2018] [Accepted: 02/07/2018] [Indexed: 12/31/2022] Open
Abstract
Spike timing-dependent plasticity (STDP) is a form of activity-dependent remodeling of synaptic strength that underlies memory formation. Despite its key role in dictating learning rules in the brain circuits, the molecular mechanisms mediating STDP are still poorly understood. Here, we show that spike timing-dependent long-term depression (tLTD) and A-type K+ currents are modulated by pharmacological agents affecting the levels of active glycogen-synthase kinase 3 (GSK3) and by GSK3β knockdown in layer 2/3 of the mouse somatosensory cortex. Moreover, the blockade of A-type K+ currents mimics the effects of GSK3 up-regulation on tLTD and occludes further changes in synaptic strength. Pharmacological, immunohistochemical and biochemical experiments revealed that GSK3β influence over tLTD induction is mediated by direct phosphorylation at Ser-616 of the Kv4.2 subunit, a molecular determinant of A-type K+ currents. Collectively, these results identify the functional interaction between GSK3β and Kv4.2 channel as a novel mechanism for tLTD modulation providing exciting insight into the understanding of GSK3β role in synaptic plasticity.
Collapse
Affiliation(s)
- Giuseppe Aceto
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Agnese Re
- Institute of Cell Biology and Neurobiology, National Research Council, Rome, Italy
| | - Andrea Mattera
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Lucia Leone
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, Rome, Italy.,Fondazione Policlinico Universitario A Gemelli, IRCCS, Rome, Italy
| | - Claudia Colussi
- Institute of Cell Biology and Neurobiology, National Research Council, Rome, Italy
| | - Marco Rinaudo
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Federico Scala
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Katia Gironi
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, Rome, Italy
| | | | - Salvatore Fusco
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, Rome, Italy.,Fondazione Policlinico Universitario A Gemelli, IRCCS, Rome, Italy
| | - Thomas Green
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA
| | - Fernanda Laezza
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA
| | - Marcello D'Ascenzo
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, Rome, Italy.,Fondazione Policlinico Universitario A Gemelli, IRCCS, Rome, Italy
| | - Claudio Grassi
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, Rome, Italy.,Fondazione Policlinico Universitario A Gemelli, IRCCS, Rome, Italy
| |
Collapse
|
15
|
Jang DC, Shim HG, Kim SJ. Intrinsic Plasticity of Cerebellar Purkinje Cells Contributes to Motor Memory Consolidation. J Neurosci 2020; 40:4145-4157. [PMID: 32295816 PMCID: PMC7244189 DOI: 10.1523/jneurosci.1651-19.2020] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 03/26/2020] [Accepted: 03/30/2020] [Indexed: 12/19/2022] Open
Abstract
Intrinsic plasticity of cerebellar Purkinje cells (PCs) has recently been demonstrated in cerebellar local circuits; however, its physiological impact on cerebellar learning and memory remains elusive. Here, we suggest that intrinsic plasticity of PCs is tightly involved in motor memory consolidation based on findings from PC-specific STIM1 knockout male mice, which show severe memory consolidation deficiency in vestibulo-ocular reflex memory. Gain-up training of the vestibulo-ocular reflex produced a decrease in the synaptic weight of PCs in both the WT and KO groups. However, intrinsic plasticity was impaired only in the knockout mice. Furthermore, the observed defects in the intrinsic plasticity of PCs led to the formation of aberrant neural plasticity in the vestibular nucleus neurons. Our results suggest that synergistic modulation of intrinsic and synaptic plasticity in PCs is required for the changes in downstream plasticity in the vestibular nucleus, and thereby contributing to the long-term storage of motor memory.SIGNIFICANCE STATEMENT Synaptic plasticity is a well-known mechanism for learning and memory. Although plasticity of excitability, intrinsic plasticity, of the cerebellar Purkinje cell has been reported in both directions (potentiation and depression), the physiological role of intrinsic plasticity still remains ambiguous. In this study, we suggest that both synaptic and intrinsic plasticity are required for successful memory consolidation in cerebellar eye movement learning. Despite successful induction and maintenance of synaptic plasticity, we found deficits of memory consolidation when there were defects in intrinsic plasticity. Our results suggest that intrinsic plasticity of cerebellar Purkinje cell has a significant role in motor memory consolidation.
Collapse
Affiliation(s)
- Dong Cheol Jang
- Department of Brain and Cognitive Science, College of Natural Science, Seoul National University, Seoul, Republic of Korea, 08826
- Department of Physiology
| | - Hyun Geun Shim
- Department of Physiology
- Department of Biomedical Sciences
| | - Sang Jeong Kim
- Department of Brain and Cognitive Science, College of Natural Science, Seoul National University, Seoul, Republic of Korea, 08826
- Department of Physiology
- Department of Biomedical Sciences
- Neuroscience Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea, 03080
| |
Collapse
|
16
|
Yang X, Bao Y, Xu J, Gong R, Zhang N, Cai L, Xia M, Wang J, Lu W. Long-Lasting Somatic Modifications of Convergent Dendritic Inputs in Hippocampal Neurons. Cereb Cortex 2020; 30:1436-1446. [PMID: 31504279 DOI: 10.1093/cercor/bhz177] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Integrated neural inputs from different dendrites converge at the soma for action potential generation. However, it is unclear how the convergent dendritic inputs interact at the soma and whether they can be further modified there. We report here an entirely new plasticity rule in hippocampal neurons in which repetitive pairing of subthreshold excitatory inputs from proximal apical and basal dendrites at a precise interval induces persistent bidirectional modifications of the two dendritic inputs. Strikingly, the modification of the dendritic inputs specially occurs at soma in the absence of somatic action potential and requires activation of somatic N-methyl-D-aspartate receptors (NMDARs). Once induced, the somatic modification can also be observed in other unpaired dendritic inputs upon their arrival at the soma. We further reveal that the soma can employ an active mechanism to potentiate the dendritic inputs by promoting sustained activation of somatic NMDARs and subsequent down-regulating of the fast inactivating A-type potassium current (IA) at the soma. Thus, the input-timing-dependent somatic plasticity we uncovered here is in sharp contrast to conventional forms of synaptic plasticity that occur at the dendrites and is important to somatic action potential generation.
Collapse
Affiliation(s)
- Xin Yang
- State Key Laboratory of Bioelectronics, Key Laboratory of Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, Jiangsu Province 210096, China
| | - Yifei Bao
- State Key Laboratory of Bioelectronics, Key Laboratory of Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, Jiangsu Province 210096, China
| | - Jindong Xu
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province 226001, People's Republic of China
| | - Ru Gong
- State Key Laboratory of Bioelectronics, Key Laboratory of Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, Jiangsu Province 210096, China
| | - Nan Zhang
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province 226001, People's Republic of China
| | - Lei Cai
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province 226001, People's Republic of China
| | - Mingmei Xia
- State Key Laboratory of Bioelectronics, Key Laboratory of Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, Jiangsu Province 210096, China
| | - Jingjing Wang
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province 226001, People's Republic of China.,Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200011, China
| | - Wei Lu
- State Key Laboratory of Bioelectronics, Key Laboratory of Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, Jiangsu Province 210096, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province 226001, People's Republic of China.,Department of Neurobiology, Nanjing Medical University, Nanjing, Jiangsu Province 210029, China
| |
Collapse
|
17
|
Kv4.1, a Key Ion Channel For Low Frequency Firing of Dentate Granule Cells, Is Crucial for Pattern Separation. J Neurosci 2020; 40:2200-2214. [PMID: 32047055 DOI: 10.1523/jneurosci.1541-19.2020] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 12/30/2019] [Accepted: 01/23/2020] [Indexed: 11/21/2022] Open
Abstract
The dentate gyrus (DG) in the hippocampus may play key roles in remembering distinct episodes through pattern separation, which may be subserved by the sparse firing properties of granule cells (GCs) in the DG. Low intrinsic excitability is characteristic of mature GCs, but ion channel mechanisms are not fully understood. Here, we investigated ionic channel mechanisms for firing frequency regulation in hippocampal GCs using male and female mice, and identified Kv4.1 as a key player. Immunofluorescence analysis showed that Kv4.1 was preferentially expressed in the DG, and its expression level determined by Western blot analysis was higher at 8-week than 3-week-old mice, suggesting a developmental regulation of Kv4.1 expression. With respect to firing frequency, GCs are categorized into two distinctive groups: low-frequency (LF) and high-frequency (HF) firing GCs. Input resistance (R in) of most LF-GCs is lower than 200 MΩ, suggesting that LF-GCs are fully mature GCs. Kv4.1 channel inhibition by intracellular perfusion of Kv4.1 antibody increased firing rates and gain of the input-output relationship selectively in LF-GCs with no significant effect on resting membrane potential and R in, but had no effect in HF-GCs. Importantly, mature GCs from mice depleted of Kv4.1 transcripts in the DG showed increased firing frequency, and these mice showed an impairment in contextual discrimination task. Our findings suggest that Kv4.1 expression occurring at late stage of GC maturation is essential for low excitability of DG networks and thereby contributes to pattern separation.SIGNIFICANCE STATEMENT The sparse activity of dentate granule cells (GCs), which is essential for pattern separation, is supported by high inhibitory inputs and low intrinsic excitability of GCs. Low excitability of GCs is thought to be attributable to a high K+ conductance at resting membrane potentials, but this study identifies Kv4.1, a depolarization-activated K+ channel, as a key ion channel that regulates firing of GCs without affecting resting membrane potentials. Kv4.1 expression is developmentally regulated and Kv4.1 currents are detected only in mature GCs that show low-frequency firing, but not in less mature high-frequency firing GCs. Furthermore, mice depleted of Kv4.1 transcripts in the dentate gyrus show impaired pattern separation, suggesting that Kv4.1 is crucial for sparse coding and pattern separation.
Collapse
|
18
|
Su ZJ, Wang XY, Zhou C, Chai Z. Down-regulation of miR-3068-3p enhances kcnip4-regulated A-type potassium current to protect against glutamate-induced excitotoxicity. J Neurochem 2019; 153:617-630. [PMID: 31792968 DOI: 10.1111/jnc.14932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Revised: 11/26/2019] [Accepted: 11/27/2019] [Indexed: 11/30/2022]
Abstract
The main cause of excitotoxic neuronal death in ischemic stroke is the massive release of glutamate. Recently, microRNAs (miRNAs) have been found to play an essential role in stroke pathology, although the molecular mechanisms remain to be investigated. Here, to identify potential candidate miRNAs involved in excitotoxicity, we treated rat primary cortical neurons with glutamate and found that miR-3068-3p, a novel miRNA, was up-regulated. We hypothesized that restoring miR-3068-3p expression might influence the neuronal injury outcomes. The inhibition of miR-3068-3p, using tough decoy lentiviruses, significantly attenuated the effects of glutamate on neuronal viability and intracellular calcium overload. To unravel the mechanisms, we employed bioinformatics analysis and RNA sequencing to identify downstream target genes. Additional luciferase assays and western blots validated kcnip4, a Kv4-mediated A-type potassium current (IA ) regulator, as a direct target of miR-3068-3p. The inhibition of miR-3068-3p increased kcnip4 expression and vice versa. In addition, the knockdown of kcnip4 by shRNA abolished the protective effect of miR-3068-3p, and over-expressing kcnip4 alone was sufficient to play a neuroprotective role in excitotoxicity. Moreover the inhibition of miR-3068-3p enhanced the IA density, and the pharmacological inhibition of IA abrogated the protective role of miR-3068-3p inhibition and kcnip4 over-expression. Therefore, we conclude that inhibition of miR-3068-3p protects against excitotoxicity via its target gene, kcnip4, and kcnip4-regulated IA . Our data suggest that the miR-3068-3p/kcnip4 axis may serve as a novel target for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Zi-Jun Su
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing, China
| | - Xu-Yi Wang
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing, China
| | - Chen Zhou
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing, China
| | - Zhen Chai
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing, China
| |
Collapse
|
19
|
Translating preclinical findings in clinically relevant new antipsychotic targets: focus on the glutamatergic postsynaptic density. Implications for treatment resistant schizophrenia. Neurosci Biobehav Rev 2019; 107:795-827. [DOI: 10.1016/j.neubiorev.2019.08.019] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Revised: 07/20/2019] [Accepted: 08/22/2019] [Indexed: 02/07/2023]
|
20
|
Newly Discovered Action of HpTx3 from Venom of Heteropoda venatoria on Na v1.7 and Its Pharmacological Implications in Analgesia. Toxins (Basel) 2019; 11:toxins11120680. [PMID: 31757020 PMCID: PMC6950750 DOI: 10.3390/toxins11120680] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 11/17/2019] [Accepted: 11/18/2019] [Indexed: 02/07/2023] Open
Abstract
It has been reported that Heteropodatoxin3 (HpTx3), a peptidic neurotoxin purified from the venom of the spider species Heteropoda venatoria, could inhibit Kv4.2 channels. Our present study newly found that HpTx3 also has potent and selective inhibitory action on Nav1.7, with an IC50 of 135.61 ± 12.98 nM. Without effect on the current–voltage (I-V) relationship of Nav1.7, HpTx3 made minor alternation in the voltage-dependence of activation and steady-state inactivation of Nav1.7 (4.15 mV and 7.29 mV, respectively) by interacting with the extracellular S3–S4 loop (S3b–S4 sequence) in domain II and the domain IV of the Nav channel subtype, showing the characteristics of both pore blocker and gate modifier toxin. During the interaction of HpTx3 with the S3b–S4 sequence of Nav1.7, the amino acid residue D in the sequence played a key role. When administered intraperitoneally or intramuscularly, HpTx3 displayed potent analgesic activity in a dose-dependent manner in different mouse pain models induced by formalin, acetic acid, complete Freund’s adjuvant, hot plate, or spared nerve injury, demonstrating that acute, inflammatory, and neuropathic pains were all effectively inhibited by the toxin. In most cases HpTx3 at doses of ≥ 1mg/kg could produce the analgesic effect comparable to that of 1 mg/kg morphine. These results suggest that HpTx3 not only can be used as a molecular probe to investigate ion channel function and pain mechanism, but also has potential in the development of the drugs that treat the Nav1.7 channel-related pain.
Collapse
|
21
|
Murphy JG, Hoffman DA. A polybasic motif in alternatively spliced KChIP2 isoforms prevents Ca 2+ regulation of Kv4 channels. J Biol Chem 2019; 294:3683-3695. [PMID: 30622142 DOI: 10.1074/jbc.ra118.006549] [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: 11/08/2018] [Revised: 01/04/2019] [Indexed: 11/06/2022] Open
Abstract
The Kv4 family of A-type voltage-gated K+ channels regulates the excitability in hippocampal pyramidal neuron dendrites and are key determinants of dendritic integration, spike timing-dependent plasticity, long-term potentiation, and learning. Kv4.2 channel expression is down-regulated following hippocampal seizures and in epilepsy, suggesting A-type currents as therapeutic targets. In addition to pore-forming Kv4 subunits, modulatory auxiliary subunits called K+ channel-interacting proteins (KChIPs) modulate Kv4 expression and activity and are required to recapitulate native hippocampal A-type currents in heterologous expression systems. KChIP mRNAs contain multiple start sites and alternative exons that generate considerable N-terminal variation and functional diversity in shaping Kv4 currents. As members of the EF-hand domain-containing neuronal Ca2+ sensor protein family, KChIP auxiliary proteins may convey Ca2+ sensitivity upon Kv4 channels; however, to what degree intracellular Ca2+ regulates KChIP-Kv4.2 complexes is unclear. To answer this question, we expressed KChIP2 with Kv4.2 in HEK293T cells, and, with whole-cell patch-clamp electrophysiology, measured an ∼1.5-fold increase in Kv4.2 current density in the presence of elevated intracellular Ca2+ Intriguingly, the Ca2+ regulation of Kv4 current was specific to KChIP2b and KChIP2c splice isoforms that lack a putative polybasic domain that is present in longer KChIP2a1 and KChIP2a isoforms. Site-directed acidification of the basic residues within the polybasic motif of KChIP2a1 rescued Ca2+-mediated regulation of Kv4 current density. These results support divergent Ca2+ regulation of Kv4 channels mediated by alternative splicing of KChIP2 isoforms. They suggest that distinct KChIP-Kv4 interactions may differentially control excitability and function of hippocampal dendrites.
Collapse
Affiliation(s)
- Jonathan G Murphy
- From the NIGMS and .,Section on Molecular Neurophysiology, NICHD, National Institutes of Health, Bethesda, Maryland 20892
| | - Dax A Hoffman
- Section on Molecular Neurophysiology, NICHD, National Institutes of Health, Bethesda, Maryland 20892
| |
Collapse
|
22
|
Ho JK, Nation DA. Cognitive benefits of angiotensin IV and angiotensin-(1-7): A systematic review of experimental studies. Neurosci Biobehav Rev 2018; 92:209-225. [PMID: 29733881 PMCID: PMC8916541 DOI: 10.1016/j.neubiorev.2018.05.005] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 03/23/2018] [Accepted: 05/02/2018] [Indexed: 12/19/2022]
Abstract
OBJECTIVES To explore effects of the brain renin-angiotensin system (RAS) on cognition. DESIGN Systematic review of experimental (non-human) studies assessing cognitive effects of RAS peptides angiotensin-(3-8) [Ang IV] and angiotensin-(1-7) [Ang-(1-7)] and their receptors, the Ang IV receptor (AT4R) and the Mas receptor. RESULTS Of 450 articles identified, 32 met inclusion criteria. Seven of 11 studies of normal animals found Ang IV had beneficial effects on tests of passive or conditioned avoidance and object recognition. In models of cognitive deficit, eight of nine studies found Ang IV and its analogs (Nle1-Ang IV, dihexa, LVV-hemorphin-7) improved performance on spatial working memory and passive avoidance tasks. Two of three studies examining Ang-(1-7) found it benefited memory. Mas receptor removal was associated with reduced fear memory in one study. CONCLUSION Studies of cognitive impairment show salutary effects of acute administration of Ang IV and its analogs, as well as AT4R activation. Brain RAS peptides appear most effective administered intracerebroventricularly, close to the time of learning acquisition or retention testing. Ang-(1-7) shows anti-dementia qualities.
Collapse
Affiliation(s)
- Jean K Ho
- Department of Psychology, University of Southern California, Los Angeles, CA, USA.
| | - Daniel A Nation
- Department of Psychology, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
23
|
Chang JT, Higley MJ. Potassium channels contribute to activity-dependent regulation of dendritic inhibition. Physiol Rep 2018; 6:e13747. [PMID: 29939492 PMCID: PMC6016672 DOI: 10.14814/phy2.13747] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 05/29/2018] [Indexed: 11/24/2022] Open
Abstract
GABAergic inhibition plays a critical role in the regulation of neuronal activity. In the neocortex, inhibitory interneurons that target the dendrites of pyramidal cells influence both electrical and biochemical postsynaptic signaling. Voltage-gated ion channels strongly shape dendritic excitability and the integration of excitatory inputs, but their contribution to GABAergic signaling is less well understood. By combining 2-photon calcium imaging and focal GABA uncaging, we show that voltage-gated potassium channels normally suppress the GABAergic inhibition of calcium signals evoked by back-propagating action potentials in dendritic spines and shafts of cortical pyramidal neurons. Moreover, the voltage-dependent inactivation of these channels leads to enhancement of dendritic calcium inhibition following somatic spiking. Computational modeling reveals that the enhancement of calcium inhibition involves an increase in action potential depolarization coupled with the nonlinear relationship between membrane voltage and calcium channel activation. Overall, our findings highlight the interaction between intrinsic and synaptic properties and reveal a novel mechanism for the activity-dependent regulation of GABAergic inhibition.
Collapse
Affiliation(s)
- Jeremy T. Chang
- Department of NeuroscienceProgram in Cellular Neuroscience, Neurodegeneration and RepairKavli InstituteYale School of MedicineNew HavenConnecticut
| | - Michael J. Higley
- Department of NeuroscienceProgram in Cellular Neuroscience, Neurodegeneration and RepairKavli InstituteYale School of MedicineNew HavenConnecticut
| |
Collapse
|
24
|
Shim HG, Lee YS, Kim SJ. The Emerging Concept of Intrinsic Plasticity: Activity-dependent Modulation of Intrinsic Excitability in Cerebellar Purkinje Cells and Motor Learning. Exp Neurobiol 2018; 27:139-154. [PMID: 30022866 PMCID: PMC6050419 DOI: 10.5607/en.2018.27.3.139] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 06/28/2018] [Accepted: 06/28/2018] [Indexed: 12/15/2022] Open
Abstract
What is memory? How does the brain process the sensory information and modify an organism's behavior? Many neuroscientists have focused on the activity- and experience-dependent modifications of synaptic functions in order to solve these fundamental questions in neuroscience. Recently, the plasticity of intrinsic excitability (called intrinsic plasticity) has emerged as an important element for information processing and storage in the brain. As the cerebellar Purkinje cells are the sole output neurons in the cerebellar cortex and the information is conveyed from a neuron to its relay neurons by forms of action potential firing, the modulation of the intrinsic firing activity may play a critical role in the cerebellar learning. Many voltage-gated and/or Ca2+-activated ion channels are involved in shaping the spiking output as well as integrating synaptic inputs to finely tune the cerebellar output. Recent studies suggested that the modulation of the intrinsic excitability and its plasticity in the cerebellar Purkinje cells might function as an integrator for information processing and memory formation. Moreover, the intrinsic plasticity might also determine the strength of connectivity to the sub-cortical areas such as deep cerebellar nuclei and vestibular nuclei to trigger the consolidation of the cerebellar-dependent memory by transferring the information.
Collapse
Affiliation(s)
- Hyun Geun Shim
- Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Korea
- Department of Biomedical Science, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Yong-Seok Lee
- Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Korea
- Department of Biomedical Science, Seoul National University College of Medicine, Seoul 03080, Korea
- Neuroscience Research Institute, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Sang Jeong Kim
- Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Korea
- Department of Biomedical Science, Seoul National University College of Medicine, Seoul 03080, Korea
- Neuroscience Research Institute, Seoul National University College of Medicine, Seoul 03080, Korea
| |
Collapse
|
25
|
Riehle M, Tsvetkov D, Gohlke BO, Preissner R, Harteneck C, Gollasch M, Nürnberg B. Molecular basis for the sensitivity of TRP channels to polyunsaturated fatty acids. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2018; 391:833-846. [PMID: 29736621 PMCID: PMC6061713 DOI: 10.1007/s00210-018-1507-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 04/17/2018] [Indexed: 11/29/2022]
Abstract
Transient receptor potential (TRP) channels represent a superfamily of unselective cation channels that are subdivided into seven subfamilies based on their sequence homology and differences in gating and functional properties. Little is known about the molecular mechanisms of TRP channel regulation, particularly of the “canonical” TRP (TRPC) subfamily and their activation by polyunsaturated fatty acids (PUFAs). Here, we analyzed the structure-function relationship of Drosophila fruit fly TRPC channels. The primary aim was to uncover the molecular basis of PUFA sensitivity of Drosophila TRP-like (TRPL) and TRPgamma channels. Amino acid (aa) sequence alignment of the three Drosophila TRPC channels revealed 50 aa residues highly conserved in PUFA-sensitive TRPL and TRPgamma channels but not in the PUFA-insensitive TRP channel. Substitution of respective aa in TRPL by corresponding aa of TRP identified 18 residues that are necessary for PUFA-mediated activation of TRPL. Most aa positions are located within a stretch comprising transmembrane domains S2–S4, whereas six aa positions have been assigned to the proximal cytosolic C-terminus. Interestingly, residues I465 and S471 are required for activation by 5,8,11,14-eicosatetraynoic acid (ETYA) but not 5,8,11-eicosatriynoic acid (ETI). As proof of concept, we generated a PUFA-sensitive TRP channel by exchanging the corresponding aa from TRPL to TRP. Our study demonstrates a specific aa pattern in the transmembrane domains S2–S4 and the proximal C-terminus essential for TRP channel activation by PUFAs.
Collapse
Affiliation(s)
- Marc Riehle
- Department of Pharmacology and Experimental Therapy, Institute of Experimental and Clinical Pharmacology and Toxicology, Eberhard Karls University Hospitals and Clinics and Interfaculty Center of Pharmacogenomics and Drug Research (ICePhA), Wilhelmstrasse 56, 72074, Tübingen, Germany
| | - Dmitry Tsvetkov
- Department of Pharmacology and Experimental Therapy, Institute of Experimental and Clinical Pharmacology and Toxicology, Eberhard Karls University Hospitals and Clinics and Interfaculty Center of Pharmacogenomics and Drug Research (ICePhA), Wilhelmstrasse 56, 72074, Tübingen, Germany.,Experimental and Clinical Research Center (ECRC), a joint cooperation of the Charité University Medicine and Max Delbruck Center for Molecular Medicine in the Helmholtz Association, Lindenberger Weg 80, 13125, Berlin, Germany.,Medical Clinic for Nephrology and Internal Intensive Care, Charité Campus Virchow Klinikum, Berlin, Germany
| | - Björn-Oliver Gohlke
- Structural Bioinformatics Group, Institute for Physiology, Charité - University Medicine Berlin, Berlin, Germany
| | - Robert Preissner
- Structural Bioinformatics Group, Institute for Physiology, Charité - University Medicine Berlin, Berlin, Germany
| | - Christian Harteneck
- Department of Pharmacology and Experimental Therapy, Institute of Experimental and Clinical Pharmacology and Toxicology, Eberhard Karls University Hospitals and Clinics and Interfaculty Center of Pharmacogenomics and Drug Research (ICePhA), Wilhelmstrasse 56, 72074, Tübingen, Germany
| | - Maik Gollasch
- Experimental and Clinical Research Center (ECRC), a joint cooperation of the Charité University Medicine and Max Delbruck Center for Molecular Medicine in the Helmholtz Association, Lindenberger Weg 80, 13125, Berlin, Germany. .,Medical Clinic for Nephrology and Internal Intensive Care, Charité Campus Virchow Klinikum, Berlin, Germany.
| | - Bernd Nürnberg
- Department of Pharmacology and Experimental Therapy, Institute of Experimental and Clinical Pharmacology and Toxicology, Eberhard Karls University Hospitals and Clinics and Interfaculty Center of Pharmacogenomics and Drug Research (ICePhA), Wilhelmstrasse 56, 72074, Tübingen, Germany.
| |
Collapse
|
26
|
Kv4.2 autism and epilepsy mutation enhances inactivation of closed channels but impairs access to inactivated state after opening. Proc Natl Acad Sci U S A 2018; 115:E3559-E3568. [PMID: 29581270 DOI: 10.1073/pnas.1717082115] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
A de novo mutation in the KCND2 gene, which encodes the Kv4.2 K+ channel, was identified in twin boys with intractable, infant-onset epilepsy and autism. Kv4.2 channels undergo closed-state inactivation (CSI), a mechanism by which channels inactivate without opening during subthreshold depolarizations. CSI dynamically modulates neuronal excitability and action potential back propagation in response to excitatory synaptic input, controlling Ca2+ influx into dendrites and regulating spike timing-dependent plasticity. Here, we show that the V404M mutation specifically affects the mechanism of CSI, enhancing the inactivation of channels that have not opened while dramatically impairing the inactivation of channels that have opened. The mutation gives rise to these opposing effects by increasing the stability of the inactivated state and in parallel, profoundly slowing the closure of open channels, which according to our data, is required for CSI. The larger volume of methionine compared with valine is a major factor underlying altered inactivation gating. Our results suggest that V404M increases the strength of the physical interaction between the pore gate and the voltage sensor regardless of whether the gate is open or closed. Furthermore, in contrast to previous proposals, our data strongly suggest that physical coupling between the voltage sensor and the pore gate is maintained in the inactivated state. The state-dependent effects of V404M on CSI are expected to disturb the regulation of neuronal excitability and the induction of spike timing-dependent plasticity. Our results strongly support a role for altered CSI gating in the etiology of epilepsy and autism in the affected twins.
Collapse
|
27
|
Prechtel H, Hartmann S, Minge D, Bähring R. Somatodendritic surface expression of epitope-tagged and KChIP binding-deficient Kv4.2 channels in hippocampal neurons. PLoS One 2018; 13:e0191911. [PMID: 29385176 PMCID: PMC5792006 DOI: 10.1371/journal.pone.0191911] [Citation(s) in RCA: 5] [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: 10/10/2017] [Accepted: 01/12/2018] [Indexed: 11/19/2022] Open
Abstract
Kv4.2 channels mediate a subthreshold-activating somatodendritic A-type current (ISA) in hippocampal neurons. We examined the role of accessory Kv channel interacting protein (KChIP) binding in somatodendritic surface expression and activity-dependent decrease in the availability of Kv4.2 channels. For this purpose we transfected cultured hippocampal neurons with cDNA coding for Kv4.2 wild-type (wt) or KChIP binding-deficient Kv4.2 mutants. All channels were equipped with an externally accessible hemagglutinin (HA)-tag and an EGFP-tag, which was attached to the C-terminal end. Combined analyses of EGFP self-fluorescence, surface HA immunostaining and patch-clamp recordings demonstrated similar dendritic trafficking and functional surface expression for Kv4.2[wt]HA,EGFP and the KChIP binding-deficient Kv4.2[A14K]HA,EGFP. Coexpression of exogenous KChIP2 augmented the surface expression of Kv4.2[wt]HA,EGFP but not Kv4.2[A14K]HA,EGFP. Notably, activity-dependent decrease in availability was more pronounced in Kv4.2[wt]HA,EGFP + KChIP2 coexpressing than in Kv4.2[A14K]HA,EGFP + KChIP2 coexpressing neurons. Our results do not support the notion that accessory KChIP binding is a prerequisite for dendritic trafficking and functional surface expression of Kv4.2 channels, however, accessory KChIP binding may play a potential role in Kv4.2 modulation during intrinsic plasticity processes.
Collapse
Affiliation(s)
- Helena Prechtel
- Institut für Zelluläre und Integrative Physiologie, Zentrum für Experimentelle Medizin, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Sven Hartmann
- Institut für Zelluläre und Integrative Physiologie, Zentrum für Experimentelle Medizin, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Daniel Minge
- Institut für Zelluläre und Integrative Physiologie, Zentrum für Experimentelle Medizin, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Robert Bähring
- Institut für Zelluläre und Integrative Physiologie, Zentrum für Experimentelle Medizin, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
- * E-mail:
| |
Collapse
|
28
|
Day SM, Yang W, Ewin S, Zhou X, Ma T. Glucagon-like peptide-1 cleavage product GLP-1 (9-36) amide enhances hippocampal long-term synaptic plasticity in correlation with suppression of Kv4.2 expression and eEF2 phosphorylation. Hippocampus 2017; 27:1264-1274. [PMID: 28833775 DOI: 10.1002/hipo.22795] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 07/31/2017] [Accepted: 08/11/2017] [Indexed: 12/24/2022]
Abstract
Glucagon-like peptide-1 (GLP-1) is an endogenous gut hormone and a key regulator in maintaining glucose homeostasis by stimulating insulin secretion. Its natural cleavage product GLP-1 (9-36), used to be considered a "bio-inactive" metabolite mainly because of its lack of insulinotropic effects and low affinity for GLP-1 receptors, possesses unique properties such as anti-oxidant and cardiovascular protection. Little is known about the role of GLP-1 (9-36) in central nervous system. Here we report that chronic, systemic application of GLP-1 (9-36) in adult mice facilitated both the induction and maintenance phases of hippocampal long-term potentiation (LTP), a major form of synaptic plasticity. In contrast, spatial learning and memory, as assessed by the Morris water maze test, was not altered by GLP-1 (9-36) administration. At the molecular level, GLP-1 (9-36) reduced protein levels of the potassium channel Kv4.2 in hippocampus, which is linked to elevated dendritic membrane excitability. Moreover, GLP-1(9-36) treatment inhibited phosphorylation of mRNA translational factor eEF2, which is associated with increased capacity for de novo protein synthesis. Finally, we showed that the LTP-enhancing effects by GLP-1 (9-36) treatment in vivo were blunted by application of exendin(9-39)amide [EX(9-39)], the GLP-1 receptor (GLP-1R) antagonist, suggesting its role as a GLP-1R agonist. These findings demonstrate that GLP-1 (9-36), which was considered a "bio-inactive" peptide, clearly exerts physiological effects on neuronal plasticity in the hippocampus, a brain region critical for learning and memory.
Collapse
Affiliation(s)
- Stephen M Day
- Department of Internal Medicine-Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157
| | - Wenzhong Yang
- Department of Internal Medicine-Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157
| | - Sarah Ewin
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157
| | - Xueyan Zhou
- Department of Internal Medicine-Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157
| | - Tao Ma
- Department of Internal Medicine-Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157
- Department of Neurobiology and Anatomy, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157
| |
Collapse
|
29
|
Elinder F, Liin SI. Actions and Mechanisms of Polyunsaturated Fatty Acids on Voltage-Gated Ion Channels. Front Physiol 2017; 8:43. [PMID: 28220076 PMCID: PMC5292575 DOI: 10.3389/fphys.2017.00043] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 01/16/2017] [Indexed: 01/29/2023] Open
Abstract
Polyunsaturated fatty acids (PUFAs) act on most ion channels, thereby having significant physiological and pharmacological effects. In this review we summarize data from numerous PUFAs on voltage-gated ion channels containing one or several voltage-sensor domains, such as voltage-gated sodium (NaV), potassium (KV), calcium (CaV), and proton (HV) channels, as well as calcium-activated potassium (KCa), and transient receptor potential (TRP) channels. Some effects of fatty acids appear to be channel specific, whereas others seem to be more general. Common features for the fatty acids to act on the ion channels are at least two double bonds in cis geometry and a charged carboxyl group. In total we identify and label five different sites for the PUFAs. PUFA site 1: The intracellular cavity. Binding of PUFA reduces the current, sometimes as a time-dependent block, inducing an apparent inactivation. PUFA site 2: The extracellular entrance to the pore. Binding leads to a block of the channel. PUFA site 3: The intracellular gate. Binding to this site can bend the gate open and increase the current. PUFA site 4: The interface between the extracellular leaflet of the lipid bilayer and the voltage-sensor domain. Binding to this site leads to an opening of the channel via an electrostatic attraction between the negatively charged PUFA and the positively charged voltage sensor. PUFA site 5: The interface between the extracellular leaflet of the lipid bilayer and the pore domain. Binding to this site affects slow inactivation. This mapping of functional PUFA sites can form the basis for physiological and pharmacological modifications of voltage-gated ion channels.
Collapse
Affiliation(s)
- Fredrik Elinder
- Department of Clinical and Experimental Medicine, Linköping University Linköping, Sweden
| | - Sara I Liin
- Department of Clinical and Experimental Medicine, Linköping University Linköping, Sweden
| |
Collapse
|
30
|
Touzet C. Morvan's syndrome and the sustained absence of all sleep rhythms for months or years: An hypothesis. Med Hypotheses 2016; 94:51-4. [PMID: 27515199 DOI: 10.1016/j.mehy.2016.06.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 06/05/2016] [Accepted: 06/09/2016] [Indexed: 01/03/2023]
Abstract
Despite the predation costs, sleep is ubiquitous in the animal realm. Humans spend a third of their life sleeping, and the quality of sleep has been related to co-morbidity, Alzheimer disease, etc. Excessive wakefulness induces rapid changes in cognitive performances, and it is claimed that one could die of sleep deprivation as quickly as by absence of water. In this context, the fact that a few people are able to go without sleep for months, even years, without displaying any cognitive troubles requires explanations. Theories ascribing sleep to memory consolidation are unable to explain such observations. It is not the case of the theory of sleep as the hebbian reinforcement of the inhibitory synapses (ToS-HRIS). Hebbian learning (Long Term Depression - LTD) guarantees that an efficient inhibitory synapse will lose its efficiency just because it is efficient at avoiding the activation of the post-synaptic neuron. This erosion of the inhibition is replenished by hebbian learning (Long Term Potentiation - LTP) when pre and post-synaptic neurons are active together - which is exactly what happens with the travelling depolarization waves of the slow-wave sleep (SWS). The best documented cases of months-long insomnia are reports of patients with Morvan's syndrome. This syndrome has an autoimmune cause that impedes - among many things - the potassium channels of the post-synaptic neurons, increasing LTP and decreasing LTD. We hypothesize that the absence of inhibitory efficiency erosion during wakefulness (thanks to a decrease of inhibitory LTD) is the cause for an absence of slow-wave sleep (SWS), which results also in the absence of REM sleep.
Collapse
Affiliation(s)
- Claude Touzet
- Aix Marseille Univ, CNRS, Neurosciences Intégratives et Adaptatives UMR 7260, 13331 Marseille, France.
| |
Collapse
|
31
|
Marschollek C, Karimzadeh F, Jafarian M, Ahmadi M, Mohajeri SMR, Rahimi S, Speckmann EJ, Gorji A. Effects of garlic extract on spreading depression: In vitro and in vivo investigations. Nutr Neurosci 2016; 20:127-134. [PMID: 25138625 DOI: 10.1179/1476830514y.0000000148] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
OBJECTIVES The potential use of garlic for prevention and treatment of different types of headaches has been suggested by several medieval literatures. Cortical spreading depression (CSD), a propagating wave of neuroglial depolarization, was established as a target for anti-migraine drugs. This study was designed to investigate the effect of garlic extract on CSD in adult rats. METHODS CSD was induced by KCl microinjection in the somatosensory cortex. The effects of five different concentrations of garlic oil (1-500 μl/l) were tested on different characteristic features of CSD in necocortical slices. In in vivo experiments, the effects of garlic oil on electrophysiological and morphological changes induced by CSD were investigated. RESULTS Garlic oil in a dose-dependent manner decreased the amplitude of CSD but not its duration and velocity in neocortical brain slices. Garlic oil at concentration of 500 μl/l reversibly reduced the amplitude of the field excitatory post-synaptic potentials and inhibited induction of long-term potentiation in the third layer of neocortical slices. In in vivo studies, systemic application of garlic oil (1 ml/l) for three consecutive days reduced the amplitude and repetition rate of CSD. Garlic oil also prevented of CSD-induced reactive astrocytosis in the neocortex. DISCUSSION Garlic oil suppresses CSD, likely via inhibition of synaptic plasticity, and prevents its harmful effects on astrocyte. Further studies are required to identify the exact active ingredient(s) of garlic oil that inhibit CSD and may have the potential to use in treatment of CSD-related disorders.
Collapse
Affiliation(s)
- Claudia Marschollek
- a Institute of Neurophysiology, Westfälische Wilhelms-Universität Münster , Germany
| | | | - Maryam Jafarian
- b Shefa Neuroscience Research Center , Tehran , Iran.,c School of Advanced Technologies in Medicine , Tehran , Iran
| | - Milad Ahmadi
- b Shefa Neuroscience Research Center , Tehran , Iran
| | | | - Sadegh Rahimi
- d Department of Physiology , Mashhad University of Medical Science , Mashhad , Iran
| | | | - Ali Gorji
- a Institute of Neurophysiology, Westfälische Wilhelms-Universität Münster , Germany.,b Shefa Neuroscience Research Center , Tehran , Iran.,e Epilepsy Research Center, Department of Neurosurgery, Department of Neurology , Westfälische Wilhelms-Universität Münster , Germany
| |
Collapse
|
32
|
Gu H, Fang YJ, Liu DD, Chen P, Mei YA. cAMP/PKA Pathways and S56 Phosphorylation Are Involved in AA/PGE2-Induced Increases in rNaV1.4 Current. PLoS One 2015; 10:e0140715. [PMID: 26485043 PMCID: PMC4618696 DOI: 10.1371/journal.pone.0140715] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Accepted: 09/28/2015] [Indexed: 12/19/2022] Open
Abstract
Arachidonic acid (AA) and its metabolites are important second messengers for ion channel modulation. The effects of extracellular application of AA and its non-metabolized analogue on muscle rNaV1.4 Na+ current has been studied, but little is known about the effects of intracellular application of AA on this channel isoform. Here, we report that intracellular application of AA significantly augmented the rNaV1.4 current peak without modulating the steady-state activation and inactivation properties of the rNaV1.4 channel. These results differed from the effects of extracellular application of AA on rNaV1.4 current. The effects of intracellular AA were mimicked by prostaglandin E2 but not eicosatetraynoic acid (ETYA), the non-metabolized analogue of AA, and were eliminated by treatment with cyclooxygenase inhibitors, flufenamic acid, or indomethacin. AA/PGE2-induced activation of rNaV1.4 channels was mimicked by a cAMP analogue (db-cAMP) and eliminated by a PKA inhibitor, PKAi. Furthermore, inhibition of EP2 and EP4 (PGE2 receptors) with AH6809 and AH23848 reduced the intracellular AA/PGE2-induced increase of rNaV1.4 current. Two mutated channels, rNaV1.4S56A and rNaV1.4T21A, were designed to investigate the role of predicted phosphorylation sites in the AA/PGE2–mediated regulation of rNaV1.4 currents. In rNaV1.4S56A, the effects of intracellular db-cAMP, AA, and PGE2 were significantly reduced. The results of the present study suggest that intracellular AA augments rNaV1.4 current by PGE2/EP receptor-mediated activation of the cAMP/PKA pathway, and that the S56 residue on the channel protein is important for this process.
Collapse
Affiliation(s)
- Hua Gu
- School of Life Science and Technology, Tongji University, Shanghai 200092, PR China
- * E-mail: (HG); (YAM)
| | - Yan-Jia Fang
- School of Life Sciences, Institute of Brain Science and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200433, PR China
| | - Dong-Dong Liu
- School of Life Sciences, Institute of Brain Science and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200433, PR China
| | - Ping Chen
- School of Life Science and Technology, Tongji University, Shanghai 200092, PR China
| | - Yan-Ai Mei
- School of Life Sciences, Institute of Brain Science and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200433, PR China
- * E-mail: (HG); (YAM)
| |
Collapse
|
33
|
Gasselin C, Inglebert Y, Debanne D. Homeostatic regulation of h-conductance controls intrinsic excitability and stabilizes the threshold for synaptic modification in CA1 neurons. J Physiol 2015; 593:4855-69. [PMID: 26316265 DOI: 10.1113/jp271369] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 08/15/2015] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS We determined the contribution of the hyperpolarization-activated cationic (h) current (Ih ) to the homeostatic regulation of CA1 pyramidal cells in vitro using chronic treatments (48 h) that either increase (picrotoxin) or decrease (kynurenate) neuronal activity. The h-conductance was found to be up- or down-regulated following chronic activity enhancement or activity deprivation, respectively. This bidirectional plasticity of Ih was found to subsequently alter both apparent input resistance and intrinsic neuronal excitability. Bidirectional homeostatic plasticity of Ih also determined EPSP waveform and EPSP summation tested at 5-30 Hz. Long-term synaptic modification induced by repetitive stimulation of the Schaffer collaterals was found to be constant across treatments in the presence of Ih but not when Ih was blocked pharmacologically. Thus, bidirectional homeostatic regulation of Ih stabilizes induction of long-term synaptic modification in CA1 pyramidal neurons that depends on EPSP summation. ABSTRACT The hyperpolarization-activated cationic (h) current is a voltage-shock absorber, highly expressed in the dendrites of CA1 pyramidal neurons. Up-regulation of Ih has been reported following episodes of intense network activity but the effect of activity deprivation on Ih and the functional consequence of homeostatic regulation of Ih remain unclear. We determined here the contribution of Ih to the homeostatic regulation of CA1 pyramidal cell excitability. Intrinsic neuronal excitability was decreased in neurons treated for 2-3 days with the GABAA channel blocker picrotoxin (PiTx) but increased in neurons treated (2-3 days) with the glutamate receptor antagonist kynurenate (Kyn). Membrane capacitance remained unchanged after treatment but the apparent input resistance was reduced for PiTx-treated neurons and enhanced for Kyn-treated neurons. Maximal Ih conductance was up-regulated after chronic hyperactivity but down-regulated following chronic hypoactivity. Up-regulation of Ih in PiTx-treated cultures was found to accelerate EPSP kinetics and reduce temporal summation of EPSPs whereas opposite effects were observed in Kyn-treated cultures, indicating that homeostatic regulation of Ih may control the induction of synaptic modification depending on EPSP summation. In fact, stimulation of the Schaffer collaterals at 3-10 Hz induced differential levels of plasticity in PiTx-treated and Kyn-treated neurons when Ih was blocked pharmacologically but not in control conditions. These data indicate that homeostatic regulation of Ih normalizes the threshold for long-term synaptic modification that depends on EPSP summation. In conclusion, bidirectional homeostatic regulation of Ih not only controls spiking activity but also stabilizes the threshold for long-term potentiation induced in CA1 pyramidal neurons by repetitive stimulation.
Collapse
Affiliation(s)
- Célia Gasselin
- INSERM, U-1072, Marseille, France.,Aix-Marseille University, Marseille, France.,Neurobiology of ion channels (UNIS), Marseille, France
| | - Yanis Inglebert
- INSERM, U-1072, Marseille, France.,Aix-Marseille University, Marseille, France.,Neurobiology of ion channels (UNIS), Marseille, France
| | - Dominique Debanne
- INSERM, U-1072, Marseille, France.,Aix-Marseille University, Marseille, France.,Neurobiology of ion channels (UNIS), Marseille, France
| |
Collapse
|
34
|
The tetramerization domain potentiates Kv4 channel function by suppressing closed-state inactivation. Biophys J 2015; 107:1090-1104. [PMID: 25185545 DOI: 10.1016/j.bpj.2014.07.038] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Revised: 05/25/2014] [Accepted: 07/01/2014] [Indexed: 01/26/2023] Open
Abstract
A-type Kv4 potassium channels undergo a conformational change toward a nonconductive state at negative membrane potentials, a dynamic process known as pre-open closed states or closed-state inactivation (CSI). CSI causes inhibition of channel activity without the prerequisite of channel opening, thus providing a dynamic regulation of neuronal excitability, dendritic signal integration, and synaptic plasticity at resting. However, the structural determinants underlying Kv4 CSI remain largely unknown. We recently showed that the auxiliary KChIP4a subunit contains an N-terminal Kv4 inhibitory domain (KID) that directly interacts with Kv4.3 channels to enhance CSI. In this study, we utilized the KChIP4a KID to probe key structural elements underlying Kv4 CSI. Using fluorescence resonance energy transfer two-hybrid mapping and bimolecular fluorescence complementation-based screening combined with electrophysiology, we identified the intracellular tetramerization (T1) domain that functions to suppress CSI and serves as a receptor for the binding of KID. Disrupting the Kv4.3 T1-T1 interaction interface by mutating C110A within the C3H1 motif of T1 domain facilitated CSI and ablated the KID-mediated enhancement of CSI. Furthermore, replacing the Kv4.3 T1 domain with the T1 domain from Kv1.4 (without the C3H1 motif) or Kv2.1 (with the C3H1 motif) resulted in channels functioning with enhanced or suppressed CSI, respectively. Taken together, our findings reveal a novel (to our knowledge) role of the T1 domain in suppressing Kv4 CSI, and that KChIP4a KID directly interacts with the T1 domain to facilitate Kv4.3 CSI, thus leading to inhibition of channel function.
Collapse
|
35
|
Tau-dependent Kv4.2 depletion and dendritic hyperexcitability in a mouse model of Alzheimer's disease. J Neurosci 2015; 35:6221-30. [PMID: 25878292 DOI: 10.1523/jneurosci.2552-14.2015] [Citation(s) in RCA: 126] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Neuronal hyperexcitability occurs early in the pathogenesis of Alzheimer's disease (AD) and contributes to network dysfunction in AD patients. In other disorders with neuronal hyperexcitability, dysfunction in the dendrites often contributes, but dendritic excitability has not been directly examined in AD models. We used dendritic patch-clamp recordings to measure dendritic excitability in the CA1 region of the hippocampus. We found that dendrites, more so than somata, of hippocampal neurons were hyperexcitable in mice overexpressing Aβ. This dendritic hyperexcitability was associated with depletion of Kv4.2, a dendritic potassium channel important for regulating dendritic excitability and synaptic plasticity. The antiepileptic drug, levetiracetam, blocked Kv4.2 depletion. Tau was required, as crossing with tau knock-out mice also prevented both Kv4.2 depletion and dendritic hyperexcitability. Dendritic hyperexcitability induced by Kv4.2 deficiency exacerbated behavioral deficits and increased epileptiform activity in hAPP mice. We conclude that increased dendritic excitability, associated with changes in dendritic ion channels including Kv4.2, may contribute to neuronal dysfunction in early stages AD.
Collapse
|
36
|
Lugo JN, Smith GD, Arbuckle EP, White J, Holley AJ, Floruta CM, Ahmed N, Gomez MC, Okonkwo O. Deletion of PTEN produces autism-like behavioral deficits and alterations in synaptic proteins. Front Mol Neurosci 2014; 7:27. [PMID: 24795561 PMCID: PMC3997048 DOI: 10.3389/fnmol.2014.00027] [Citation(s) in RCA: 125] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Accepted: 03/24/2014] [Indexed: 11/13/2022] Open
Abstract
Many genes have been implicated in the underlying cause of autism but each gene accounts for only a small fraction of those diagnosed with autism. There is increasing evidence that activity-dependent changes in neuronal signaling could act as a convergent mechanism for many of the changes in synaptic proteins. One candidate signaling pathway that may have a critical role in autism is the PI3K/AKT/mTOR pathway. A major regulator of this pathway is the negative repressor phosphatase and tensin homolog (PTEN). In the current study we examined the behavioral and molecular consequences in mice with neuron subset-specific deletion of PTEN. The knockout (KO) mice showed deficits in social chamber and social partition test. KO mice demonstrated alterations in repetitive behavior, as measured in the marble burying test and hole-board test. They showed no changes in ultrasonic vocalizations emitted on postnatal day 10 or 12 compared to wildtype (WT) mice. They exhibited less anxiety in the elevated-plus maze test and were more active in the open field test compared to WT mice. In addition to the behavioral alterations, KO mice had elevation of phosphorylated AKT, phosphorylated S6, and an increase in S6K. KO mice had a decrease in mGluR but an increase in total and phosphorylated fragile X mental retardation protein. The disruptions in intracellular signaling may be why the KO mice had a decrease in the dendritic potassium channel Kv4.2 and a decrease in the synaptic scaffolding proteins PSD-95 and SAP102. These findings demonstrate that deletion of PTEN results in long-term alterations in social behavior, repetitive behavior, activity, and anxiety. In addition, deletion of PTEN significantly alters mGluR signaling and many synaptic proteins in the hippocampus. Our data demonstrates that deletion of PTEN can result in many of the behavioral features of autism and may provide insights into the regulation of intracellular signaling on synaptic proteins.
Collapse
Affiliation(s)
- Joaquin N Lugo
- Department of Psychology and Neuroscience, Baylor University Waco, TX, USA ; Institute of Biomedical Studies, Baylor University Waco, TX, USA
| | - Gregory D Smith
- Institute of Biomedical Studies, Baylor University Waco, TX, USA
| | - Erin P Arbuckle
- Institute of Biomedical Studies, Baylor University Waco, TX, USA
| | - Jessika White
- Department of Psychology and Neuroscience, Baylor University Waco, TX, USA
| | - Andrew J Holley
- Department of Psychology and Neuroscience, Baylor University Waco, TX, USA
| | - Crina M Floruta
- Department of Psychology and Neuroscience, Baylor University Waco, TX, USA
| | - Nowrin Ahmed
- Department of Psychology and Neuroscience, Baylor University Waco, TX, USA
| | - Maribel C Gomez
- Department of Psychology and Neuroscience, Baylor University Waco, TX, USA
| | - Obi Okonkwo
- Department of Psychology and Neuroscience, Baylor University Waco, TX, USA
| |
Collapse
|
37
|
Kang MS, Yang YS, Kim SH, Park JM, Eun SY, Jung SC. The Downregulation of Somatic A-Type K(+) Channels Requires the Activation of Synaptic NMDA Receptors in Young Hippocampal Neurons of Rats. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2014; 18:135-41. [PMID: 24757375 PMCID: PMC3994300 DOI: 10.4196/kjpp.2014.18.2.135] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Revised: 02/24/2014] [Accepted: 02/26/2014] [Indexed: 11/24/2022]
Abstract
The downregulation of A-type K+ channels (IA channels) accompanying enhanced somatic excitability can mediate epileptogenic conditions in mammalian central nervous system. As IA channels are dominantly targeted by dendritic and postsynaptic processings during synaptic plasticity, it is presumable that they may act as cellular linkers between synaptic responses and somatic processings under various excitable conditions. In the present study, we electrophysiologically tested if the downregulation of somatic IA channels was sensitive to synaptic activities in young hippocampal neurons. In primarily cultured hippocampal neurons (DIV 6~9), the peak of IA recorded by a whole-cell patch was significantly reduced by high KCl or exogenous glutamate treatment to enhance synaptic activities. However, the pretreatment of MK801 to block synaptic NMDA receptors abolished the glutamate-induced reduction of the IA peak, indicating the necessity of synaptic activation for the reduction of somatic IA. This was again confirmed by glycine treatment, showing a significant reduction of the somatic IA peak. Additionally, the gating property of IA channels was also sensitive to the activation of synaptic NMDA receptors, showing the hyperpolarizing shift in inactivation kinetics. These results suggest that synaptic LTP possibly potentiates somatic excitability via downregulating IA channels in expression and gating kinetics. The consequential changes of somatic excitability following the activity-dependent modulation of synaptic responses may be a series of processings for neuronal functions to determine outputs in memory mechanisms or pathogenic conditions.
Collapse
Affiliation(s)
- Moon-Seok Kang
- Department of Physiology, School of Medicine, Jeju National University, Jeju 690-756, Korea
| | - Yoon-Sil Yang
- Department of Physiology, School of Medicine, Jeju National University, Jeju 690-756, Korea
| | - Seon-Hee Kim
- Department of Physiology, School of Medicine, Jeju National University, Jeju 690-756, Korea
| | - Joo-Min Park
- Department of Physiology, School of Medicine, Jeju National University, Jeju 690-756, Korea
| | - Su-Yong Eun
- Department of Physiology, School of Medicine, Jeju National University, Jeju 690-756, Korea
| | - Sung-Cherl Jung
- Department of Physiology, School of Medicine, Jeju National University, Jeju 690-756, Korea. ; Institute of Medical Science, Jeju National University, Jeju 690-756, Korea
| |
Collapse
|
38
|
Loss of functional A-type potassium channels in the dendrites of CA1 pyramidal neurons from a mouse model of fragile X syndrome. J Neurosci 2014; 33:19442-50. [PMID: 24336711 DOI: 10.1523/jneurosci.3256-13.2013] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Despite the critical importance of voltage-gated ion channels in neurons, very little is known about their functional properties in Fragile X syndrome: the most common form of inherited cognitive impairment. Using three complementary approaches, we investigated the physiological role of A-type K(+) currents (I(KA)) in hippocampal CA1 pyramidal neurons from fmr1-/y mice. Direct measurement of I(KA) using cell-attached patch-clamp recordings revealed that there was significantly less I(KA) in the dendrites of CA1 neurons from fmr1-/y mice. Interestingly, the midpoint of activation for A-type K(+) channels was hyperpolarized for fmr1-/y neurons compared with wild-type, which might partially compensate for the lower current density. Because of the rapid time course for recovery from steady-state inactivation, the dendritic A-type K(+) current in CA1 neurons from both wild-type and fmr1-/y mice is likely mediated by K(V)4 containing channels. The net effect of the differences in I(KA) was that back-propagating action potentials had larger amplitudes producing greater calcium influx in the distal dendrites of fmr1-/y neurons. Furthermore, CA1 pyramidal neurons from fmr1-/y mice had a lower threshold for LTP induction. These data suggest that loss of I(KA) in hippocampal neurons may contribute to dendritic pathophysiology in Fragile X syndrome.
Collapse
|
39
|
Segatto M, Manduca A, Lecis C, Rosso P, Jozwiak A, Swiezewska E, Moreno S, Trezza V, Pallottini V. Simvastatin treatment highlights a new role for the isoprenoid/cholesterol biosynthetic pathway in the modulation of emotional reactivity and cognitive performance in rats. Neuropsychopharmacology 2014; 39:841-54. [PMID: 24108067 PMCID: PMC3924519 DOI: 10.1038/npp.2013.284] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Revised: 10/02/2013] [Accepted: 10/03/2013] [Indexed: 11/09/2022]
Abstract
The aim of the present work was to shed light on the role played by the isoprenoid/cholesterol biosynthetic pathway in the modulation of emotional reactivity and memory consolidation in rodents through the inhibition of the key and rate-limiting enzyme 3-hydroxy 3-methylglutaryl Coenzyme A reductase (HMGR) both in vivo and in vitro with simvastatin. Three-month-old male Wistar rats treated for 21 days with simvastatin or vehicle were tested in the social interaction, elevated plus-maze, and inhibitory avoidance tasks; after behavioral testing, the amygdala, hippocampus, prefrontal cortex, dorsal, and ventral striatum were dissected out for biochemical assays. In order to delve deeper into the molecular mechanisms underlying the observed effects, primary rat hippocampal neurons were used. Our results show that HMGR inhibition by simvastatin induces anxiogenic-like effects in the social interaction but not in the elevated plus-maze test, and improves memory consolidation in the inhibitory avoidance task. These effects are accompanied by imbalances in the activity of specific prenylated proteins, Rab3 and RhoA, involved in neurotransmitter release, and synaptic plasticity, respectively. Taken together, the present findings indicate that the isoprenoid/cholesterol biosynthetic pathway is critically involved in the physiological modulation of both emotional and cognitive processes in rodents.
Collapse
Affiliation(s)
- Marco Segatto
- Department of Science, University Roma Tre, Viale Marconi, Rome, Italy
| | - Antonia Manduca
- Department of Science, University Roma Tre, Viale Marconi, Rome, Italy
| | - Claudio Lecis
- Department of Science, University Roma Tre, Viale Marconi, Rome, Italy
| | - Pamela Rosso
- Department of Science, University Roma Tre, Viale Marconi, Rome, Italy
| | - Adam Jozwiak
- Institute of Biochemistry and Biophysics Polish Academy of Sciences, Warsaw, Poland
| | - Ewa Swiezewska
- Institute of Biochemistry and Biophysics Polish Academy of Sciences, Warsaw, Poland
| | - Sandra Moreno
- Department of Science, University Roma Tre, Viale Marconi, Rome, Italy
| | - Viviana Trezza
- Department of Science, University Roma Tre, Viale Marconi, Rome, Italy
| | | |
Collapse
|
40
|
Yang YS, Kim KD, Eun SY, Jung SC. Roles of somatic A-type K(+) channels in the synaptic plasticity of hippocampal neurons. Neurosci Bull 2014; 30:505-14. [PMID: 24526657 DOI: 10.1007/s12264-013-1399-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2013] [Accepted: 07/19/2013] [Indexed: 01/11/2023] Open
Abstract
In the mammalian brain, information encoding and storage have been explained by revealing the cellular and molecular mechanisms of synaptic plasticity at various levels in the central nervous system, including the hippocampus and the cerebral cortices. The modulatory mechanisms of synaptic excitability that are correlated with neuronal tasks are fundamental factors for synaptic plasticity, and they are dependent on intracellular Ca(2+)-mediated signaling. In the present review, the A-type K(+) (IA) channel, one of the voltage-dependent cation channels, is considered as a key player in the modulation of Ca(2+) influx through synaptic NMDA receptors and their correlated signaling pathways. The cellular functions of IA channels indicate that they possibly play as integral parts of synaptic and somatic complexes, completing the initiation and stabilization of memory.
Collapse
Affiliation(s)
- Yoon-Sil Yang
- Department of Physiology, School of Medicine, Jeju National University, Jeju, 690756, Republic of Korea
| | | | | | | |
Collapse
|
41
|
Brager DH, Johnston D. Channelopathies and dendritic dysfunction in fragile X syndrome. Brain Res Bull 2014; 103:11-7. [PMID: 24462643 DOI: 10.1016/j.brainresbull.2014.01.002] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2013] [Revised: 01/06/2014] [Accepted: 01/13/2014] [Indexed: 11/17/2022]
Abstract
Dendritic spine abnormalities and the metabotropic glutamate receptor theory put the focus squarely on synapses and protein synthesis as the cellular locus of fragile X syndrome. Synapses however, are only partly responsible for information processing in neuronal networks. Neurotransmitter triggered excitatory postsynaptic potentials (EPSPs) are shaped and integrated by dendritic voltage-gated ion channels. These EPSPs, and in some cases the resultant dendritic spikes, are further modified by dendritic voltage-gated ion channels as they propagate to the soma. If the resultant somatic depolarization is large enough, action potential(s) will be triggered and propagate both orthodromically down the axon, where it may trigger neurotransmitter release, and antidromically back into the dendritic tree, where it can activate and modify dendritic voltage-gated and receptor activated ion channels. Several channelopathies, both soma-dendritic (L-type calcium channels, Slack potassium channels, h-channels, A-type potassium channels) and axo-somatic (BK channels and delayed rectifier potassium channels) were identified in the fmr1-/y mouse model of fragile X syndrome. Pathological function of these channels will strongly influence the excitability of individual neurons as well as overall network function. In this chapter we discuss the role of voltage-gated ion channels in neuronal processing and describe how identified channelopathies in models of fragile X syndrome may play a role in dendritic pathophysiology.
Collapse
Affiliation(s)
- Darrin H Brager
- Center for Learning and Memory, University of Texas at Austin, Austin, TX 78712, United States.
| | - Daniel Johnston
- Center for Learning and Memory, University of Texas at Austin, Austin, TX 78712, United States
| |
Collapse
|
42
|
Zlomuzica A, Dere D, Dere E. The histamine H1 receptor and recollection-based discrimination in a temporal order memory task in the mouse. Pharmacol Biochem Behav 2013; 111:58-63. [DOI: 10.1016/j.pbb.2013.08.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2013] [Revised: 07/23/2013] [Accepted: 08/16/2013] [Indexed: 11/26/2022]
|
43
|
Gomes GM, Dalmolin GD, Cordeiro MDN, Gomez MV, Ferreira J, Rubin MA. The selective A-type K+ current blocker Tx3-1 isolated from the Phoneutria nigriventer venom enhances memory of naïve and Aβ(25-35)-treated mice. Toxicon 2013; 76:23-7. [PMID: 23994427 DOI: 10.1016/j.toxicon.2013.08.059] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Revised: 08/07/2013] [Accepted: 08/13/2013] [Indexed: 11/15/2022]
Abstract
Potassium channels regulate many neuronal functions, including neuronal excitability and synaptic plasticity, contributing, by these means, to mnemonic processes. In particular, A-type K(+) currents (IA) play a key role in hippocampal synaptic plasticity. Therefore, we evaluated the effect of the peptidic toxin Tx3-1, a selective blocker of IA currents, extracted from the venom of the spider Phoneutria nigriventer, on memory of mice. Administration of Tx3-1 (i.c.v., 300 pmol/site) enhanced both short- and long-term memory consolidation of mice tested in the novel object recognition task. In comparison, 4-aminopyridine (4-AP; i.c.v., 30-300 pmol/site), a non-selective K(+) channel blocker did not alter long-term memory and caused toxic side effects such as circling, freezing and tonic-clonic seizures. Moreover, Tx3-1 (i.c.v., 10-100 pmol/site) restored memory of Aβ25-35-injected mice, and exhibited a higher potency to improve memory of Aβ25-35-injected mice when compared to control group. These results show the effect of the selective blocker of IA currents Tx3-1 in both short- and long-term memory retention and in memory impairment caused by Aβ25-35, reinforcing the role of IA in physiological and pathological memory processes.
Collapse
Affiliation(s)
- Guilherme M Gomes
- Graduation Program in Biological Sciences, Toxicological Biochemistry, Building 18, Room 2203, Center of Exact and Natural Sciences, Federal University of Santa Maria, Santa Maria, RS 97105-900, Brazil
| | | | | | | | | | | |
Collapse
|
44
|
Saikosaponin a Enhances Transient Inactivating Potassium Current in Rat Hippocampal CA1 Neurons. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2013; 2013:413092. [PMID: 23554830 PMCID: PMC3608310 DOI: 10.1155/2013/413092] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/16/2012] [Accepted: 01/01/2013] [Indexed: 11/18/2022]
Abstract
Saikosaponin a (SSa), a main constituent of the Chinese herb Bupleurum chinense DC., has been demonstrated to have antiepileptic activity. Recent studies have shown that SSa could inhibit NMDA receptor current and persistent sodium current. However, the effects of SSa on potassium (K+) currents remain unclear. In this study, we tested the effect of SSa on 4AP-induced epileptiform discharges and K+ currents in CA1 neurons of rat hippocampal slices. We found that SSa significantly inhibited epileptiform discharges frequency and duration in hippocampal CA1 neurons in the 4AP seizure model in a dose-dependent manner with an IC50
of 0.7 μM. SSa effectively increased the amplitude of ITotal
and IA, significantly negative-shifted the activation curve, and positive-shifted steady-state curve of IA. However, SSa induced no significant changes in the amplitude and activation curve of IK. In addition, SSa significantly increased the amplitude of 4AP-sensitive K+ current, while there was no significant change in the amplitude of TEA-sensitive K+ current. Together, our data indicate that SSa inhibits epileptiform discharges induced by 4AP in a dose-dependent manner and that SSa exerts selectively enhancing effects on IA. These increases in IA may contribute to the anticonvulsant mechanisms of SSa.
Collapse
|
45
|
Heler R, Bell JK, Boland LM. Homology model and targeted mutagenesis identify critical residues for arachidonic acid inhibition of Kv4 channels. Channels (Austin) 2013; 7:74-84. [PMID: 23334377 PMCID: PMC3667888 DOI: 10.4161/chan.23453] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Polyunsaturated fatty acids such as arachidonic acid (AA) exhibit inhibitory modulation of Kv4 potassium channels. Molecular docking approaches using a Kv4.2 homology model predicted a membrane-embedded binding pocket for AA comprised of the S4-S5 linker on one subunit and several hydrophobic residues within S3, S5 and S6 from an adjacent subunit. The pocket is conserved among Kv4 channels. We tested the hypothesis that modulatory effects of AA on Kv4.2/KChIP channels require access to this site. Targeted mutation of a polar residue (K318) and a nonpolar residue (G314) within the S4-S5 linker as well as a nonpolar residue in S3 (V261) significantly impaired the effects of AA on K (+) currents in Xenopus oocytes. These residues may be important in stabilizing (K318) or regulating access to (V261, G314) the negatively charged carboxylate moiety on the fatty acid. Structural specificity was supported by the lack of disruption of AA effects observed with mutations at residues located near, but not within the predicted binding pocket. Furthermore, we found that the crystal structure of the related Kv1.2/2.1 chimera lacks the structural features present in the proposed AA docking site of Kv4.2 and the Kv1.2/2.1 K (+) currents were unaffected by AA. We simulated the mutagenic substitutions in our Kv4.2 model to demonstrate how specific mutations may disrupt the putative AA binding pocket. We conclude that AA inhibits Kv4 channel currents and facilitates current decay by binding within a hydrophobic pocket in the channel in which K318 within the S4-S5 linker is a critical residue for AA interaction.
Collapse
Affiliation(s)
- Robert Heler
- Department of Biology, University of Richmond, Richmond, VA, USA
| | | | | |
Collapse
|
46
|
Carter BC, Giessel AJ, Sabatini BL, Bean BP. Transient sodium current at subthreshold voltages: activation by EPSP waveforms. Neuron 2012; 75:1081-93. [PMID: 22998875 DOI: 10.1016/j.neuron.2012.08.033] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/21/2012] [Indexed: 10/27/2022]
Abstract
Tetrodotoxin (TTX)-sensitive sodium channels carry large transient currents during action potentials and also "persistent" sodium current, a noninactivating TTX-sensitive current present at subthreshold voltages. We examined gating of subthreshold sodium current in dissociated cerebellar Purkinje neurons and hippocampal CA1 neurons, studied at 37°C with near-physiological ionic conditions. Unexpectedly, in both cell types small voltage steps at subthreshold voltages activated a substantial component of transient sodium current as well as persistent current. Subthreshold EPSP-like waveforms also activated a large component of transient sodium current, but IPSP-like waveforms engaged primarily persistent sodium current with only a small additional transient component. Activation of transient as well as persistent sodium current at subthreshold voltages produces amplification of EPSPs that is sensitive to the rate of depolarization and can help account for the dependence of spike threshold on depolarization rate, as previously observed in vivo.
Collapse
Affiliation(s)
- Brett C Carter
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | |
Collapse
|
47
|
Tigerholm J, Migliore M, Fransén E. Integration of synchronous synaptic input in CA1 pyramidal neuron depends on spatial and temporal distributions of the input. Hippocampus 2012; 23:87-99. [PMID: 22996230 DOI: 10.1002/hipo.22061] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/16/2012] [Indexed: 11/09/2022]
Abstract
Highly synchronized neural firing has been discussed in relation to learning and memory, for instance sharp-wave activity in hippocampus. We were interested to study how a postsynaptic CA1 pyramidal neuron would integrate input of different levels of synchronicity. In previous work using computational modeling we studied how the integration depends on dendritic conductances. We found that the transient A-type potassium channel K(A) was able to selectively suppress input of high synchronicity. In recent years, compartmentalization of dendritic integration has been shown. We were therefore interested to study the influence of localization and pattern of synaptic input over the dendritic tree of the CA1 pyramidal neuron. We find that the selective suppression increases when synaptic inputs are placed on oblique dendrites further out from the soma. The suppression also increases along the radial axis from the apical trunk out to the end of oblique dendrites. We also find that the K(A) channel suppresses the occurrence of dendritic spikes. Moreover, recent studies have shown interaction between synaptic inputs. We therefore studied the influence of apical tuft input on the integration studied above. We find that excitatory input provides a modulatory influence reducing the capacity of K(A) to suppress synchronized activity, thus facilitating the excitatory drive of oblique dendritic input. Conversely, inhibitory tuft input increases the suppression by K(A) providing a larger control of oblique depolarizing factors on the CA1 pyramidal neuron in terms of what constitutes the most effective level of synchronicity. Furthermore, we show that the selective suppression studied above depends on the conductance of the K(A) channel. K(A) , as several other potassium channels, is modulated by several neuromodulators, for instance acetylcholine and dopamine, both of which have been discussed in relation to learning and memory. We suggest that dendritic conductances and their modulatory systems may be part of the regulation of processing of information, in particular for how network synchronicity affects learning and memory.
Collapse
Affiliation(s)
- Jenny Tigerholm
- Department of Computational Biology, School of Computer Science and Communication, KTH Royal Institute of Technology, SE-106 91 Stockholm, Sweden.
| | | | | |
Collapse
|
48
|
An L, Liu S, Yang Z, Zhang T. Cognitive impairment in rats induced by nano-CuO and its possible mechanisms. Toxicol Lett 2012; 213:220-7. [PMID: 22820425 DOI: 10.1016/j.toxlet.2012.07.007] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2012] [Revised: 06/11/2012] [Accepted: 07/10/2012] [Indexed: 11/18/2022]
Affiliation(s)
- Lei An
- College of Life Sciences, Nankai University, Tianjin 300071, PR China
| | | | | | | |
Collapse
|
49
|
Witzel K, Fischer P, Bähring R. Hippocampal A-type current and Kv4.2 channel modulation by the sulfonylurea compound NS5806. Neuropharmacology 2012; 63:1389-403. [PMID: 22964468 DOI: 10.1016/j.neuropharm.2012.08.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Revised: 08/16/2012] [Accepted: 08/18/2012] [Indexed: 12/24/2022]
Abstract
We examined the effects of the sulfonylurea compound NS5806 on neuronal A-type channel function. Using whole-cell patch-clamp we studied the effects of NS5806 on the somatodendritic A-type current (I(SA)) in cultured hippocampal neurons and the currents mediated by Kv4.2 channels coexpressed with different auxiliary β-subunits, including both Kv channel interacting proteins (KChIPs) and dipeptidyl aminopeptidase-related proteins (DPPs), in HEK 293 cells. The amplitude of the I(SA) component in hippocampal neurons was reduced in the presence of 20 μM NS5806. I(SA) decay kinetics were slowed and the recovery kinetics accelerated, but the voltage dependence of steady-state inactivation was shifted to more negative potentials by NS5806. The peak amplitudes of currents mediated by ternary Kv4.2 channel complexes, associated with DPP6-S (short splice-variant) and either KChIP2, KChIP3 or KChIP4, were potentiated and their macroscopic inactivation slowed by NS5806, whereas the currents mediated by binary Kv4.2 channels, associated only with DPP6-S, were suppressed, and the NS5806-mediated slowing of macroscopic inactivation was less pronounced. Neither potentiation nor suppression and no effect on current decay kinetics in the presence of NS5806 were observed for Kv4.2 channels associated with KChIP3 and the N-type inactivation-conferring DPP6a splice-variant. For all recombinant channel complexes, NS5806 slowed the recovery from inactivation and shifted the voltage dependence of steady-state inactivation to more negative potentials. Our results demonstrate the activity of NS5806 on native I(SA) and possible molecular correlates in the form of recombinant Kv4.2 channels complexed with different KChIPs and DPPs, and they shed some light on the mechanism of NS5806 action.
Collapse
Affiliation(s)
- Katrin Witzel
- Institut für Zelluläre und Integrative Physiologie, Zentrum für Experimentelle Medizin, Universitätsklinikum Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | | | | |
Collapse
|
50
|
Ohtsuki G, Piochon C, Adelman JP, Hansel C. SK2 channel modulation contributes to compartment-specific dendritic plasticity in cerebellar Purkinje cells. Neuron 2012; 75:108-20. [PMID: 22794265 PMCID: PMC3398406 DOI: 10.1016/j.neuron.2012.05.025] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/14/2012] [Indexed: 02/06/2023]
Abstract
Small-conductance Ca(2+)-activated K(+) channels (SK channels) modulate excitability and curtail excitatory postsynaptic potentials (EPSPs) in neuronal dendrites. Here, we demonstrate long-lasting plasticity of intrinsic excitability (IE) in dendrites that results from changes in the gain of this regulatory mechanism. Using dendritic patch-clamp recordings from rat cerebellar Purkinje cells, we find that somatic depolarization or parallel fiber (PF) burst stimulation induce long-term amplification of synaptic responses to climbing fiber (CF) or PF stimulation and enhance the amplitude of passively propagated sodium spikes. Dendritic plasticity is mimicked and occluded by the SK channel blocker apamin and is absent in Purkinje cells from SK2 null mice. Triple-patch recordings from two dendritic sites and the soma and confocal calcium imaging studies show that local stimulation limits dendritic plasticity to the activated compartment of the dendrite. This plasticity mechanism allows Purkinje cells to adjust the SK2-mediated control of dendritic excitability in an activity-dependent manner.
Collapse
Affiliation(s)
- Gen Ohtsuki
- Department of Neurobiology, University of Chicago, Chicago, Illinois 60637, USA
- Department of Neuroscience, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands
| | - Claire Piochon
- Department of Neurobiology, University of Chicago, Chicago, Illinois 60637, USA
| | - John P. Adelman
- Vollum Institute, Oregon Health & Science University, Portland, Oregon 97239, USA
| | - Christian Hansel
- Department of Neurobiology, University of Chicago, Chicago, Illinois 60637, USA
- Department of Neuroscience, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands
| |
Collapse
|