1
|
Jenkins PM, Bender KJ. Axon initial segment structure and function in health and disease. Physiol Rev 2025; 105:765-801. [PMID: 39480263 DOI: 10.1152/physrev.00030.2024] [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: 08/01/2024] [Revised: 10/18/2024] [Accepted: 10/23/2024] [Indexed: 11/06/2024] Open
Abstract
At the simplest level, neurons are structured to integrate synaptic input and perform computational transforms on that input, converting it into an action potential (AP) code. This process, converting synaptic input into AP output, typically occurs in a specialized region of the axon termed the axon initial segment (AIS). The AIS, as its name implies, is often contained to the first section of axon abutted to the soma and is home to a dizzying array of ion channels, attendant scaffolding proteins, intracellular organelles, extracellular proteins, and, in some cases, synapses. The AIS serves multiple roles as the final arbiter for determining if inputs are sufficient to evoke APs, as a gatekeeper that physically separates the somatodendritic domain from the axon proper, and as a regulator of overall neuronal excitability, dynamically tuning its size to best suit the needs of parent neurons. These complex roles have received considerable attention from experimentalists and theoreticians alike. Here, we review recent advances in our understanding of the AIS and its role in neuronal integration and polarity in health and disease.
Collapse
Affiliation(s)
- Paul M Jenkins
- Departments of Pharmacology and Psychiatry, University of Michigan Medical School, Ann Arbor, Michigan, United States
| | - Kevin J Bender
- Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, California, United States
| |
Collapse
|
2
|
Chen X, Feng Y, Quinn RJ, Pountney DL, Richardson DR, Mellick GD, Ma L. Potassium Channels in Parkinson's Disease: Potential Roles in Its Pathogenesis and Innovative Molecular Targets for Treatment. Pharmacol Rev 2023; 75:758-788. [PMID: 36918260 DOI: 10.1124/pharmrev.122.000743] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 01/05/2023] [Accepted: 03/07/2023] [Indexed: 03/16/2023] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized by selective loss of dopaminergic neurons in the substantia nigra pars compacta (SNpc) region of the midbrain. The loss of neurons results in a subsequent reduction of dopamine in the striatum, which underlies the core motor symptoms of PD. To date, there are no effective treatments to stop, slow, or reverse the pathologic progression of dopaminergic neurodegeneration. This unfortunate predicament is because of the current early stages in understanding the biologic targets and pathways involved in PD pathogenesis. Ion channels have become emerging targets for new therapeutic development for PD due to their essential roles in neuronal function and neuroinflammation. Potassium channels are the most prominent ion channel family and have been shown to be critically important in PD pathology because of their roles in modulating neuronal excitability, neurotransmitter release, synaptic transmission, and neuroinflammation. In this review, members of the subfamilies of voltage-gated K+ channels, inward rectifying K+ channels, and Ca2+-activated K+ channels are described. Evidence of the role of these channels in PD etiology is discussed together with the latest views on related pathologic mechanisms and their potential as biologic targets for developing neuroprotective drugs for PD. SIGNIFICANCE STATEMENT: Parkinson's disease (PD) is the second most common neurodegenerative disorder, featuring progressive degeneration of dopaminergic neurons in the midbrain. It is a multifactorial disease involving multiple risk factors and complex pathobiological mechanisms. Mounting evidence suggests that ion channels play vital roles in the pathogenesis and progression of PD by regulating neuronal excitability and immune cell function. Therefore, they have become "hot" biological targets for PD, as demonstrated by multiple clinical trials of drug candidates targeting ion channels for PD therapy.
Collapse
Affiliation(s)
- Xiaoyi Chen
- School of Environment and Science (Y.F., D.R.R., G.D.M., L.M.) and Centre for Cancer Cell Biology and Drug Discovery (D.R.R.), Griffith Institute for Drug Discovery (X.C., Y.F., R.J.Q., D.R.R., G.D.M., L.M.), Griffith University, Nathan, Brisbane, Queensland, Australia; and School of Pharmacy and Medical Science, Griffith University, Gold Coast, Queenslandstate, Australia (D.L.P.)
| | - Yunjiang Feng
- School of Environment and Science (Y.F., D.R.R., G.D.M., L.M.) and Centre for Cancer Cell Biology and Drug Discovery (D.R.R.), Griffith Institute for Drug Discovery (X.C., Y.F., R.J.Q., D.R.R., G.D.M., L.M.), Griffith University, Nathan, Brisbane, Queensland, Australia; and School of Pharmacy and Medical Science, Griffith University, Gold Coast, Queenslandstate, Australia (D.L.P.)
| | - Ronald J Quinn
- School of Environment and Science (Y.F., D.R.R., G.D.M., L.M.) and Centre for Cancer Cell Biology and Drug Discovery (D.R.R.), Griffith Institute for Drug Discovery (X.C., Y.F., R.J.Q., D.R.R., G.D.M., L.M.), Griffith University, Nathan, Brisbane, Queensland, Australia; and School of Pharmacy and Medical Science, Griffith University, Gold Coast, Queenslandstate, Australia (D.L.P.)
| | - Dean L Pountney
- School of Environment and Science (Y.F., D.R.R., G.D.M., L.M.) and Centre for Cancer Cell Biology and Drug Discovery (D.R.R.), Griffith Institute for Drug Discovery (X.C., Y.F., R.J.Q., D.R.R., G.D.M., L.M.), Griffith University, Nathan, Brisbane, Queensland, Australia; and School of Pharmacy and Medical Science, Griffith University, Gold Coast, Queenslandstate, Australia (D.L.P.)
| | - Des R Richardson
- School of Environment and Science (Y.F., D.R.R., G.D.M., L.M.) and Centre for Cancer Cell Biology and Drug Discovery (D.R.R.), Griffith Institute for Drug Discovery (X.C., Y.F., R.J.Q., D.R.R., G.D.M., L.M.), Griffith University, Nathan, Brisbane, Queensland, Australia; and School of Pharmacy and Medical Science, Griffith University, Gold Coast, Queenslandstate, Australia (D.L.P.)
| | - George D Mellick
- School of Environment and Science (Y.F., D.R.R., G.D.M., L.M.) and Centre for Cancer Cell Biology and Drug Discovery (D.R.R.), Griffith Institute for Drug Discovery (X.C., Y.F., R.J.Q., D.R.R., G.D.M., L.M.), Griffith University, Nathan, Brisbane, Queensland, Australia; and School of Pharmacy and Medical Science, Griffith University, Gold Coast, Queenslandstate, Australia (D.L.P.)
| | - Linlin Ma
- School of Environment and Science (Y.F., D.R.R., G.D.M., L.M.) and Centre for Cancer Cell Biology and Drug Discovery (D.R.R.), Griffith Institute for Drug Discovery (X.C., Y.F., R.J.Q., D.R.R., G.D.M., L.M.), Griffith University, Nathan, Brisbane, Queensland, Australia; and School of Pharmacy and Medical Science, Griffith University, Gold Coast, Queenslandstate, Australia (D.L.P.)
| |
Collapse
|
3
|
Wang ZW, Trussell LO, Vedantham K. Regulation of Neurotransmitter Release by K + Channels. ADVANCES IN NEUROBIOLOGY 2023; 33:305-331. [PMID: 37615872 DOI: 10.1007/978-3-031-34229-5_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/25/2023]
Abstract
K+ channels play potent roles in the process of neurotransmitter release by influencing the action potential waveform and modulating neuronal excitability and release probability. These diverse effects of K+ channel activation are ensured by the wide variety of K+ channel genes and their differential expression in different cell types. Accordingly, a variety of K+ channels have been implicated in regulating neurotransmitter release, including the Ca2+- and voltage-gated K+ channel Slo1 (also known as BK channel), voltage-gated K+ channels of the Kv3 (Shaw-type), Kv1 (Shaker-type), and Kv7 (KCNQ) families, G-protein-gated inwardly rectifying K+ (GIRK) channels, and SLO-2 (a Ca2+-. Cl-, and voltage-gated K+ channel in C. elegans). These channels vary in their expression patterns, subcellular localization, and biophysical properties. Their roles in neurotransmitter release may also vary depending on the synapse and physiological or experimental conditions. This chapter summarizes key findings about the roles of K+ channels in regulating neurotransmitter release.
Collapse
Affiliation(s)
- Zhao-Wen Wang
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, CT, USA.
| | - Laurence O Trussell
- Oregon Hearing Research Center & Vollum Institute, Oregon Health and Science University, Portland, OR, USA
| | - Kiranmayi Vedantham
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, CT, USA
| |
Collapse
|
4
|
Zhang Y, Li D, Darwish Y, Fu X, Trussell LO, Huang H. KCNQ Channels Enable Reliable Presynaptic Spiking and Synaptic Transmission at High Frequency. J Neurosci 2022; 42:3305-3315. [PMID: 35256530 PMCID: PMC9034779 DOI: 10.1523/jneurosci.0363-20.2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Revised: 02/06/2022] [Accepted: 02/26/2022] [Indexed: 11/21/2022] Open
Abstract
The presynaptic action potential (AP) is required to drive calcium influx into nerve terminals, resulting in neurotransmitter release. Accordingly, the AP waveform is crucial in determining the timing and strength of synaptic transmission. The calyx of Held nerve terminals of rats of either sex showed minimum changes in AP waveform during high-frequency AP firing. We found that the stability of the calyceal AP waveform requires KCNQ (KV7) K+ channel activation during high-frequency spiking activity. High-frequency presynaptic spikes gradually led to accumulation of KCNQ channels in open states which kept interspike membrane potential sufficiently negative to maintain Na+ channel availability. Blocking KCNQ channels during stimulus trains led to inactivation of presynaptic Na+, and to a lesser extent KV1 channels, thereby reducing the AP amplitude and broadening AP duration. Moreover, blocking KCNQ channels disrupted the stable calcium influx and glutamate release required for reliable synaptic transmission at high frequency. Thus, while KCNQ channels are generally thought to prevent hyperactivity of neurons, we find that in axon terminals these channels function to facilitate reliable high-frequency synaptic signaling needed for sensory information processing.SIGNIFICANCE STATEMENT The presynaptic spike results in calcium influx required for neurotransmitter release. For this reason, the spike waveform is crucial in determining the timing and strength of synaptic transmission. Auditory information is encoded by spikes phase locked to sound frequency at high rates. The calyx of Held nerve terminals in the auditory brainstem show minimum changes in spike waveform during high-frequency spike firing. We found that activation of KCNQ K+ channel builds up during high-frequency firing and its activation helps to maintain a stable spike waveform and reliable synaptic transmission. While KCNQ channels are generally thought to prevent hyperexcitability of neurons, we find that in axon terminals these channels function to facilitate high-frequency synaptic signaling during auditory information processing.
Collapse
Affiliation(s)
- Yihui Zhang
- Department of Cell and Molecular Biology, Tulane University, New Orleans, Louisiana 70118
| | - Dainan Li
- Department of Cell and Molecular Biology, Tulane University, New Orleans, Louisiana 70118
| | - Youad Darwish
- Department of Cell and Molecular Biology, Tulane University, New Orleans, Louisiana 70118
| | - Xin Fu
- Brain Institute, Tulane University, New Orleans, LA 70118
| | - Laurence O Trussell
- Oregon Hearing Research Center and Vollum Institute, Oregon Health and Science University, Portland, OR 97239
| | - Hai Huang
- Department of Cell and Molecular Biology, Tulane University, New Orleans, Louisiana 70118
- Brain Institute, Tulane University, New Orleans, LA 70118
| |
Collapse
|
5
|
Biba N, Becq H, Pallesi-Pocachard E, Sarno S, Granjeaud S, Montheil A, Kurz M, Villard L, Milh M, Santini PPL, Aniksztejn L. Time-limited alterations in cortical activity of a knock-in mice model of KCNQ2-related developmental and epileptic encephalopathy. J Physiol 2022; 600:2429-2460. [PMID: 35389519 DOI: 10.1113/jp282536] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 03/10/2022] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS The electrophysiological impact of the pathogenic c.821C>T mutation of the KCNQ2 gene (p.T274M variant in Kv7.2 subunit) related to Developmental and Epileptic Encephalopathy has been analyzed both in vivo and ex-vivo in layers II/III and V of motor cortical slice from a knock-in mice model during development at neonatal, post-weaning and juvenile stages. M current density and conductance are decreased and excitability of layers II/III pyramidal cells is increased in slices from neonatal and post-weaning KI mice but not from juvenile KI mice. M current and excitability of layer V pyramidal cells are impacted in KI mice only at post-weaning stage. Spontaneous GABAergic network-driven events are recorded until post-weaning stage and their frequency are increased in layers II/III of the KI mice. KI mice displayed spontaneous seizures preferentially at post-weaning rather than at juvenile stages. ABSTRACT De novo missense variants in the KCNQ2 gene encoding the Kv7.2 subunit of the voltage-gated potassium Kv7/M channels are the main cause of Developmental and Epileptic Encephalopathy (DEE) with neonatal onset. While seizures usually resolve during development, cognitive/motor deficits persist. To better understand the cellular mechanisms underlying network dysfunction and their progression over time, we investigated in vivo, using local field potential recordings of freely moving animals, and ex-vivo in layers II/III and V of motor cortical slices, using patch-clamp recordings, the electrophysiological properties of pyramidal cells from a heterozygous knock-in (KI) mouse model carrying the Kv7.2 p.T274M pathogenic variant during neonatal, post-weaning and juvenile developmental stages. We found that KI mice displayed spontaneous seizures preferentially at post-weaning rather than at juvenile stages. At the cellular level, the variant led to a reduction in M current density/conductance and to neuronal hyperexcitability. These alterations were observed during the neonatal period in pyramidal cells of layers II /III and during post-weaning stage in pyramidal cells of layer V. Moreover, there was an increase in the frequency of spontaneous network driven events mediated by GABA receptors suggesting that the excitability of interneurons was also increased. However, all these alterations were no more observed in layers II/III and V of juvenile mice. Thus, our data indicate that the action of the variant is developmentally regulated. This raises the possibility that the age related seizure remission observed in KCNQ2-related DEE patient results from a time limited alteration of Kv7 channels activity and neuronal excitability. Abstract figure legend Knock-in mice harboring the heterozygous pathogenic p.T274M variant in the Kv7.2 subunit (c.821C>T mutation of the KCNQ2 gene) related to Developmental and Epileptic Encephalopathy displayed epileptic seizures preferentially at post-weaning rather than at juvenile developmental stages. At cellular level, in motor cortical slices the variant led to a reduction in M current density, to a hyperexcitability of pyramidal cells and to an increase in the frequency of spontaneous network driven events mediated by GABA receptors. All these alterations are time limited and are observed in pyramidal cells of neonatal mice until post-weaning but not of juvenile mice in which the pyramidal cells have electrophysiological properties similar to those of wild-type mice. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Najoua Biba
- INSERM, INMED (U1249), Aix-Marseille University, Turing centre for living system, Marseille, France
| | - Hélène Becq
- INSERM, INMED (U1249), Aix-Marseille University, Turing centre for living system, Marseille, France
| | - Emilie Pallesi-Pocachard
- INSERM, INMED (U1249), Aix-Marseille University, Turing centre for living system, Marseille, France
| | - Stefania Sarno
- INSERM, INMED (U1249), Aix-Marseille University, Turing centre for living system, Marseille, France
| | - Samuel Granjeaud
- Centre de Recherche en Cancérologie de Marseille, INSERM, U1068, Institut Paoli Calmettes, CNRS, UMR7258, Aix-Marseille University UM 105, Marseille, France
| | - Aurélie Montheil
- INSERM, INMED (U1249), Aix-Marseille University, Turing centre for living system, Marseille, France
| | - Marie Kurz
- INSERM, INMED (U1249), Aix-Marseille University, Turing centre for living system, Marseille, France
| | - Laurent Villard
- Aix-Marseille University, INSERM, MMG, Marseille, France.,Department of Medical Genetics, La Timone Childrens's Hospital, Marseille, France
| | - Mathieu Milh
- Aix-Marseille University, INSERM, MMG, Marseille, France.,Department of Pediatric Neurology, La Timone Children's Hospital, Marseille, France
| | | | - Laurent Aniksztejn
- INSERM, INMED (U1249), Aix-Marseille University, Turing centre for living system, Marseille, France
| |
Collapse
|
6
|
Ye H, Liu ZX, He YJ, Wang X. Effects of M currents on the persistent activity of pyramidal neurons in mouse primary auditory cortex. J Neurophysiol 2022; 127:1269-1278. [PMID: 35294269 DOI: 10.1152/jn.00332.2021] [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
Neuronal persistent activity (PA) is a common phenomenon observed in many types of neurons. PA can be induced in neurons in the mouse auditory nucleus by activating cholinergic receptors with carbachol (CCh), a dual muscarinic and nicotinic receptor agonist. PA is presumed to be associated with learning-related auditory plasticity at the cellular level. However, the mechanism is not clearly understood. Many studies have reported that muscarinic cholinergic receptor agonists inhibit muscarinic-sensitive potassium channels (M channels). Potassium influx through M channels produces potassium currents, called M currents, which play an essential role in regulating neural excitability and synaptic plasticity. Further study is needed to determine whether M currents affect the PA of auditory central neurons and provide additional analysis of the variations in electrophysiological properties. We used in vitro whole-cell patch-clamp recordings in isolated mouse brain slices to investigate the effects of M currents on the PA in pyramidal neurons in layer V of the primary auditory cortex (AI-L5). We found that blocking M currents with XE991 depolarized the AI-L5 pyramidal neurons, which significantly increased the input resistance. The active threshold and threshold intensity were significantly reduced, indicating that the intrinsic excitability was enhanced. Our results also showed that blocking M currents with XE991 switched the neuronal firing patterns in the AI-L5 pyramidal neurons from regular-spiking to intrinsic-bursting. Blocking M currents facilitated PA by increasing the plateau potential and enhancing intrinsic excitability. Our results suggested that blocking M currents might facilitate the PA in AI-L5 pyramidal neurons, which underlies auditory plasticity.
Collapse
Affiliation(s)
- Huan Ye
- Hubei Key Lab of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, China
| | - Zhen-Xu Liu
- Hubei Key Lab of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, China
| | - Ya-Jie He
- Hubei Key Lab of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, China
| | - Xin Wang
- Hubei Key Lab of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, China
| |
Collapse
|
7
|
Yuan JH, Estacion M, Mis MA, Tanaka BS, Schulman BR, Chen L, Liu S, Dib-Hajj FB, Dib-Hajj SD, Waxman SG. KCNQ variants and pain modulation: a missense variant in Kv7.3 contributes to pain resilience. Brain Commun 2021; 3:fcab212. [PMID: 34557669 PMCID: PMC8454204 DOI: 10.1093/braincomms/fcab212] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 07/13/2021] [Accepted: 07/29/2021] [Indexed: 12/02/2022] Open
Abstract
There is a pressing need for understanding of factors that confer resilience to pain. Gain-of-function mutations in sodium channel Nav1.7 produce hyperexcitability of dorsal root ganglion neurons underlying inherited erythromelalgia, a human genetic model of neuropathic pain. While most individuals with erythromelalgia experience excruciating pain, occasional outliers report more moderate pain. These differences in pain profiles in blood-related erythromelalgia subjects carrying the same pain-causative Nav1.7 mutation and markedly different pain experience provide a unique opportunity to investigate potential genetic factors that contribute to inter-individual variability in pain. We studied a patient with inherited erythromelalgia and a Nav1.7 mutation (c.4345T>G, p. F1449V) with severe pain as is characteristic of most inherited erythromelalgia patients, and her mother who carries the same Nav1.7 mutation with a milder pain phenotype. Detailed six-week daily pain diaries of pain episodes confirmed their distinct pain profiles. Electrophysiological studies on subject-specific induced pluripotent stem cell-derived sensory neurons from each of these patients showed that the excitability of these cells paralleled their pain phenotype. Whole-exome sequencing identified a missense variant (c.2263C>T, p. D755N) in KCNQ3 (Kv7.3) in the pain resilient mother. Voltage-clamp recordings showed that co-expression of Kv7.2-wild type (WT)/Kv7.3-D755N channels produced larger M-currents than that of Kv7.2-WT/Kv7.3-WT. The difference in excitability of the patient-specific induced pluripotent stem cell-derived sensory neurons was mimicked by modulating M-current levels using the dynamic clamp and a model of the mutant Kv7.2-WT/Kv7.3-D755N channels. These results show that a 'pain-in-a-dish' model can be used to explicate genetic contributors to pain, and confirm that KCNQ variants can confer pain resilience via an effect on peripheral sensory neurons.
Collapse
Affiliation(s)
- Jun-Hui Yuan
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06520, USA
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06520, USA
- Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Mark Estacion
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06520, USA
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06520, USA
- Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Malgorzata A Mis
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06520, USA
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06520, USA
- Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Brian S Tanaka
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06520, USA
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06520, USA
- Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Betsy R Schulman
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06520, USA
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06520, USA
- Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Lubin Chen
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06520, USA
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06520, USA
- Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Shujun Liu
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06520, USA
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06520, USA
- Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Fadia B Dib-Hajj
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06520, USA
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06520, USA
- Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Sulayman D Dib-Hajj
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06520, USA
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06520, USA
- Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Stephen G Waxman
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06520, USA
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06520, USA
- Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT 06516, USA
| |
Collapse
|
8
|
Esaki H, Izumi S, Fukao A, Nishitani N, Deyama S, Kaneda K. Nicotine enhances object recognition memory through inhibition of voltage-dependent potassium 7 channels in the medial prefrontal cortex of mice. J Pharmacol Sci 2021; 147:58-61. [PMID: 34294373 DOI: 10.1016/j.jphs.2021.05.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 04/23/2021] [Accepted: 05/06/2021] [Indexed: 11/19/2022] Open
Abstract
Nicotine administration enhances object recognition memory. However, target brain regions and cellular mechanisms underlying the nicotine effects remain unclear. In mice, the novel object recognition test revealed that systemic nicotine administration before training enhanced object recognition memory. Moreover, this effect was inhibited by infusion of retigabine, a selective voltage-dependent potassium 7 (Kv7) channel opener, into the medial prefrontal cortex (mPFC) before nicotine administration. Additionally, infusion of XE-991, a selective Kv7 channel blocker, into the mPFC before training enhanced object recognition memory. Therefore, Kv7 channels in the mPFC may be at least partly involved in nicotine-induced 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
| | - Akari Fukao
- 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
| | - 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
|
9
|
Naffaa MM, Al-Ewaidat OA. Ligand modulation of KCNQ-encoded (K V7) potassium channels in the heart and nervous system. Eur J Pharmacol 2021; 906:174278. [PMID: 34174270 DOI: 10.1016/j.ejphar.2021.174278] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 06/06/2021] [Accepted: 06/18/2021] [Indexed: 10/21/2022]
Abstract
KCNQ-encoded (KV7) potassium channels are diversely distributed in the human tissues, associated with many physiological processes and pathophysiological conditions. These channels are increasingly used as drug targets for treating diseases. More selective and potent molecules on various types of the KV7 channels are desirable for appropriate therapies. The recent knowledge of the structure and function of human KCNQ-encoded channels makes it more feasible to achieve these goals. This review discusses the role and mechanism of action of many molecules in modulating the function of the KCNQ-encoded potassium channels in the heart and nervous system. The effects of these compounds on KV7 channels help to understand their involvement in many diseases, and to search for more selective and potent ligands to be used in the treatment of many disorders such as various types of cardiac arrhythmias, epilepsy, and pain.
Collapse
Affiliation(s)
- Moawiah M Naffaa
- Department of Cell Biology, Duke University School of Medicine, Durham, NC, 27710, USA; Department of Psychology and Neuroscience, Duke University, Durham, NC 27708, USA.
| | - Ola A Al-Ewaidat
- Faculty of Medicine, The University of Jordan, Amman, 11942, Jordan
| |
Collapse
|
10
|
The M-current works in tandem with the persistent sodium current to set the speed of locomotion. PLoS Biol 2020; 18:e3000738. [PMID: 33186352 PMCID: PMC7688130 DOI: 10.1371/journal.pbio.3000738] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 11/25/2020] [Accepted: 10/13/2020] [Indexed: 01/20/2023] Open
Abstract
The central pattern generator (CPG) for locomotion is a set of pacemaker neurons endowed with inherent bursting driven by the persistent sodium current (INaP). How they proceed to regulate the locomotor rhythm remained unknown. Here, in neonatal rodents, we identified a persistent potassium current critical in regulating pacemakers and locomotion speed. This current recapitulates features of the M-current (IM): a subthreshold noninactivating outward current blocked by 10,10-bis(4-pyridinylmethyl)-9(10H)-anthracenone dihydrochloride (XE991) and enhanced by N-(2-chloro-5-pyrimidinyl)-3,4-difluorobenzamide (ICA73). Immunostaining and mutant mice highlight an important role of Kv7.2-containing channels in mediating IM. Pharmacological modulation of IM regulates the emergence and the frequency regime of both pacemaker and CPG activities and controls the speed of locomotion. Computational models captured these results and showed how an interplay between IM and INaP endows the locomotor CPG with rhythmogenic properties. Overall, this study provides fundamental insights into how IM and INaP work in tandem to set the speed of locomotion.
Collapse
|
11
|
Osuntokun OS, Akinsomisoye SO, Olayiwola G, Adedokun KI, Oladokun OO. Carbamazepine adversely altered the pituitary-testicular axis with resultant reproductive dysfunctions than levetiracetam or carbamazepine-levetiracetam adjuvant treatment in male Wistar rat. Andrologia 2020; 52:e13871. [PMID: 33126292 DOI: 10.1111/and.13871] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 09/06/2020] [Accepted: 09/16/2020] [Indexed: 12/12/2022] Open
Abstract
This study investigated the on-toward reactions of individual or adjunctive treatment with carbamazepine (CBZ) and levetiracetam (LEV) on the pituitary-testicular axis in male rats. Twenty-four male Wistar rats were randomised into 4 groups (n = 6) and received daily intraperitoneal (i.p) treatment of normal saline (0.1 ml/day); CBZ (25 mg/kg i.p); LEV (50 mg/kg i.p); or combination of CBZ (12.5 mg/kg) and LEV (25 mg/kg) for 4 weeks. The serum concentration of luteinising hormone (LH), follicle-stimulating hormone (FSH), and testosterone was determined. Also, the seminal profile and histomorphological status of the testis were determined. Data were analysed using descriptive and inferential statistics. The control and test groups were compared using Student's t test, analysis of variance (ANOVA), and Student-Newman-Keuls post hoc analysis where appropriate, while the results presented as mean ± SEM in graphs or tables. The level of significance was taken at p < .05. The percentage motility, viability, and concentration of FSH decreased significantly in all the treatment groups, while the testis was presented with various forms of histomorphological aberrations. This study concludes that CBZ, and CBZ + LEV adjunctive treatments alter the pituitary-testicular axis with evidence of hormonal deregulation and alteration in the reproductive functions' indices, while LEV treatment remains the safest.
Collapse
Affiliation(s)
- Opeyemi Samson Osuntokun
- Department of Physiology, Faculty of Basic Medical Sciences, College of Health Sciences, Osun State University, Osogbo, Nigeria
| | - Stephen Olumide Akinsomisoye
- Department of Physiological Sciences, Faculty of Basic Medical Sciences, College of Health Sciences, Obafemi Awolowo University, Ile-Ife, Nigeria
| | - Gbola Olayiwola
- Department of Clinical Pharmacy and Pharmacy Administration, Faculty of Pharmacy Obafemi, Awolowo University, Ile-Ife, Nigeria
| | - Kabiru Isola Adedokun
- Department of Physiology, Faculty of Basic Medical Sciences, College of Health Sciences, Osun State University, Osogbo, Nigeria
| | - Olayemi Olutobi Oladokun
- Department of Physiology, Faculty of Basic Medical Sciences, College of Health Sciences, Osun State University, Osogbo, Nigeria
| |
Collapse
|
12
|
Baculis BC, Zhang J, Chung HJ. The Role of K v7 Channels in Neural Plasticity and Behavior. Front Physiol 2020; 11:568667. [PMID: 33071824 PMCID: PMC7530275 DOI: 10.3389/fphys.2020.568667] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 08/28/2020] [Indexed: 12/12/2022] Open
Abstract
Activity-dependent persistent changes in neuronal intrinsic excitability and synaptic strength are widely thought to underlie learning and memory. Voltage-gated KCNQ/Kv7 potassium channels have been of great interest as the potential targets for memory disorders due to the beneficial effects of their antagonists in cognition. Importantly, de novo dominant mutations in their neuronal subunits KCNQ2/Kv7.2 and KCNQ3/Kv7.3 are associated with epilepsy and neurodevelopmental disorder characterized by developmental delay and intellectual disability. The role of Kv7 channels in neuronal excitability and epilepsy has been extensively studied. However, their functional significance in neural plasticity, learning, and memory remains largely unknown. Here, we review recent studies that support the emerging roles of Kv7 channels in intrinsic and synaptic plasticity, and their contributions to cognition and behavior.
Collapse
Affiliation(s)
- Brian C Baculis
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Jiaren Zhang
- Department of Molecular Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Hee Jung Chung
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, United States.,Department of Molecular Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| |
Collapse
|
13
|
Djouhri L, Zeidan A, Abd El-Aleem SA, Smith T. Cutaneous Aβ-Non-nociceptive, but Not C-Nociceptive, Dorsal Root Ganglion Neurons Exhibit Spontaneous Activity in the Streptozotocin Rat Model of Painful Diabetic Neuropathy in vivo. Front Neurosci 2020; 14:530. [PMID: 32528247 PMCID: PMC7263321 DOI: 10.3389/fnins.2020.00530] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 04/29/2020] [Indexed: 12/30/2022] Open
Abstract
Diabetic peripheral neuropathic pain (DPNP) is the most devastating complication of diabetes mellitus. Unfortunately, successful therapy for DPNP remains a challenge because its pathogenesis is still elusive. However, DPNP is believed to be due partly to abnormal hyperexcitability of dorsal root ganglion (DRG) neurons, but the relative contributions of specific functional subtypes remain largely unknown. Here, using the strepotozotocin (STZ) rat model of DPNP induced by a STZ injection (60 mg/kg, i.p), and intracellular recordings of action potentials (APs) from DRG neurons in anesthetized rats, we examined electrophysiological changes in C-and Aβ-nociceptive and Aβ-low threshold mechanoreceptive (LTM) neurons that may contribute to DPNP. Compared with control, we found in STZ-rats with established pain hypersensitivity (5 weeks post-STZ) several significant changes including: (a) A 23% increase in the incidence of spontaneous activity (SA) in Aβ-LTMs (but not C-mechanosensitive nociceptors) that may cause dysesthesias/paresthesia suffered by DPNP patients, (b) membrane hyperpolarization and a ∼85% reduction in SA rate in Aβ-LTMs by Kv7 channel activation with retigabine (6 mg/kg, i.v.) suggesting that Kv7/M channels may be involved in mechanisms of SA generation in Aβ-LTMs, (c) decreases in AP duration and in duration and amplitude of afterhyperpolarization (AHP) in C-and/or Aβ-nociceptors. These faster AP and AHP kinetics may lead to repetitive firing and an increase in afferent input to the CNS and thereby contribute to DPNP development, and (d) a decrease in the electrical thresholds of Aβ-nociceptors that may contribute to their sensitization, and thus to the resulting hypersensitivity associated with DPNP.
Collapse
Affiliation(s)
- Laiche Djouhri
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Asad Zeidan
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Seham A. Abd El-Aleem
- Department of Histology and Cell Biology, University of Manchester, Manchester, United Kingdom
- Department of Pathology, Faculty of Medicine, Minia University, Minya, Egypt
| | - Trevor Smith
- Medical Physics and Biomedical Engineering, University College London, London, United Kingdom
| |
Collapse
|
14
|
Zhu C, Lin R, Liu C, Huang M, Lin F, Zhang G, Zhang Y, Miao J, Lin W, Huang H. The Antagonism of 5-HT6 Receptor Attenuates Current-Induced Spikes and Improves Long-Term Potentiation via the Regulation of M-Currents in a Pilocarpine-Induced Epilepsy Model. Front Pharmacol 2020; 11:475. [PMID: 32425770 PMCID: PMC7212420 DOI: 10.3389/fphar.2020.00475] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 03/26/2020] [Indexed: 12/02/2022] Open
Abstract
Recent studies have documented that reduced M-current promotes epileptogenesis and attenuates synaptic remodeling. Neurite growth is closely related to the level of 5-HT6 receptor (5-HT6R) in the central nervous system. However, little research is available regarding the relation between 5-HT6R and M-current and the role of 5-HT6R in M-current regulation. Herein, we found that the expression of 5-HT6R was notably increased and the expression of KNCQ2/3, the main components of the M channel, was decreased in a time-dependent manner in pilocarpine-induced chronic epileptic hippocampus. Interestingly, antagonism of 5-HT6R by SB271046 upregulated the expression of KCNQ2 but not KCNQ3. SB271046 greatly alleviated excitatory/inhibitory imbalance and improved the impaired LTP in the chronic epileptic hippocampus. Further mechanism exploration revealed that the above effects of SB271046 can be reversed by the M-channel inhibitor XE991, which also confirmed that SB271046 can indeed improve abnormal M current. These data indicate that the antagonism of 5-HT6R may decrease the excitability of hippocampal pyramidal neurons in chronic epileptic rats and improve the impaired long-term potentiation by upregulating the expression of KCNQ2 in the M-channel.
Collapse
Affiliation(s)
- Chaofeng Zhu
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Rong Lin
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Changyun Liu
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Mingzhu Huang
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Feng Lin
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Gan Zhang
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Yuying Zhang
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Junjie Miao
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Wanhui Lin
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Huapin Huang
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China.,Department of Electrophysiology, Fujian Key Laboratory of Molecular Neurology, Fuzhou, China.,Department of Geriatrics, Fujian Medical University Union Hospital, Fuzhou, China
| |
Collapse
|
15
|
So EC, Liu PY, Wu SN. Effectiveness in the inhibition of dapagliflozin and canagliflozin on M-type K + current and α-methylglucoside-induced current in pituitary tumor (GH 3) and pheochromocytoma PC12 cells. Eur J Pharmacol 2020; 879:173141. [PMID: 32353360 DOI: 10.1016/j.ejphar.2020.173141] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Revised: 04/21/2020] [Accepted: 04/21/2020] [Indexed: 12/22/2022]
Abstract
Dapagliflozin (DAPA) or canagliflozin (CANA), Na+-dependent glucose co-transporter type 2 (SGLT2) inhibitors, were used for treatment of type II diabetes mellitus. Addition of DAPA or CANA suppressed M-type K+ current (IK(M)) in pituitary tumor (GH3) and pheochromocytoma PC12 cells. The IC50 value for DAPA- or CANA-mediated inhibition of IK(M) in GH3 cells was 0.11 or 0.42 μM, respectively. The presence of DAPA (0.1 μM) shifted the steady-state activation of IK(M) to less depolarized potential without changing the gating charge of the current. During high-frequency depolarizing pulses, IK(M) magnitude was reduced by DAPA; however, DAPA-induced block of IK(M) remained effective. The amplitude of neither erg-mediated K+ current nor hyperpolarization-activated cation current in GH3 cells was modified in the presence of 1 μM DAPA. Alternatively, addition of DAPA, CANA, phlorizin or chlorotoxin effectively suppressed α-methylglucoside-(αMG-) induced current (IαMG) in GH3 cells, albeit inability of tefluthrin (activator of INa) to suppress this current. DAPA shifted the charge-voltage relation of presteady-state IαMG in a rightward and downward direction with no change in the gating charge of the IαMG. Under current-clamp recordings, subsequent additions of DAPA, but still in the continued presence of αMG, increased the firing rate of spontaneous action potentials stimulated by αMG. Our results suggested that activity of SGLT was expressed functionally in GH3 and PC12 cells. Therefore, inhibitory actions of DAPA or CANA on the amplitude and gating of IK(M) might provide a yet unidentified mechanism through which the SGLT1 or SGLT2 activity were attenuated in unclamped cells occurring in vivo.
Collapse
Affiliation(s)
- Edmund Cheung So
- Department of Anesthesia and Medical Research, An Nan Hospital, China Medical University, Tainan City, Taiwan; Graduate Institute of Medical Sciences, Chang Jung Christian University, Tainan City, Taiwan
| | - Ping-Yen Liu
- Division of Cardiovascular Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan City, Taiwan
| | - Sheng-Nan Wu
- Institute of Basic Medical Sciences, National Cheng Kung University Medical College, Tainan City, Taiwan; Department of Physiology, National Cheng Kung University Medical College, Tainan City, Taiwan.
| |
Collapse
|
16
|
Yousuf H, Nye AN, Moyer JR. Heterogeneity of neuronal firing type and morphology in retrosplenial cortex of male F344 rats. J Neurophysiol 2020; 123:1849-1863. [PMID: 32267193 DOI: 10.1152/jn.00577.2019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The rodent granular retrosplenial cortex (gRSC) has reciprocal connections to the hippocampus to support fear memories. Although activity-dependent plasticity occurs within the RSC during memory formation, the intrinsic and morphological properties of RSC neurons are poorly understood. The present study used whole-cell recordings to examine intrinsic neuronal firing and morphology of neurons in layer 2/3 (L2/3) and layer 5 (L5) of the gRSC in adult male rats. Five different classifications were observed: regular-spiking (RS), regular-spiking afterdepolarization (RSADP), late-spiking (LS), burst-spiking (BS), and fast-spiking (FS) neurons. RSADP neurons were the most commonly observed neuronal class, identified by their robust spike frequency adaptation and pronounced afterdepolarization (ADP) following an action potential (AP). They also had the most extensive dendritic branching compared with other cell types. LS neurons were predominantly found in L2/3 and exhibited a long delay before onset of their initial AP. They also had reduced dendritic branching compared with other cell types. BS neurons were limited to L5 and generated an initial burst of two or more APs. FS neurons demonstrated sustained firing and little frequency adaptation and were the only nonpyramidal firing type. Relative to adults, RS neurons from juvenile rats (PND 14-30) lacked an ADP and were less excitable. Bath application of group 1 mGluR blockers attenuated the ADP in adult neurons. In other fear-related brain structures, the ADP has been shown to enhance excitability and synaptic plasticity. Thus, understanding cellular mechanisms of the gRSC will provide insight regarding its precise role in memory-related processes across the lifespan.NEW & NOTEWORTHY This is the first study to demonstrate that granular retrosplenial cortical (gRSC) neurons exhibit five distinctive firing types: regular spiking (RS), regular spiking with an afterdepolarization (RSADP), late spiking (LS), burst spiking (BS), and fast spiking (FS). RSADP neurons were the most frequently observed cell type in adult gRSC neurons. Interestingly, RS neurons without an ADP were most common in gRSC neurons of juvenile rats (PND 14-30). Thus, the ADP property, which was previously shown to enhance neuronal excitability, emerges during development.
Collapse
Affiliation(s)
- Hanna Yousuf
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin
| | - Andrew N Nye
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin
| | - James R Moyer
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin.,Department of Biological Sciences University of Wisconsin-Milwaukee, Milwaukee, Wisconsin
| |
Collapse
|
17
|
Fernández-Fernández D, Lamas JA. Metabotropic Modulation of Potassium Channels During Synaptic Plasticity. Neuroscience 2020; 456:4-16. [PMID: 32114098 DOI: 10.1016/j.neuroscience.2020.02.025] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 02/14/2020] [Accepted: 02/18/2020] [Indexed: 01/06/2023]
Abstract
Besides their primary function mediating the repolarization phase of action potentials, potassium channels exquisitely and ubiquitously regulate the resting membrane potential of neurons and therefore have a key role establishing their intrinsic excitability. This group of proteins is composed of a very diverse collection of voltage-dependent and -independent ion channels, whose specific distribution is finely tuned at the level of the synapse. Both at the presynaptic and postsynaptic membranes, different types of potassium channels are subjected to modulation by second messenger signaling cascades triggered by metabotropic receptors, which in this way serve as a link between neurotransmitter actions and changes in the neuron membrane excitability. On the one hand, by regulating the resting membrane potential of the postsynaptic membrane, potassium channels appear to be critical towards setting the threshold for the induction of long-term potentiation and depression. On the other hand, these channels maintain the presynaptic membrane potential under control, therefore influencing the probability of neurotransmitter release underlying different forms of short-term plasticity. In the present review, we examine in detail the role of metabotropic receptors translating their activation by different neurotransmitters into a final effect modulating several types of potassium channels. Furthermore, we evaluate the consequences that this interplay has on the induction and maintenance of different forms of synaptic plasticity.
Collapse
Affiliation(s)
- D Fernández-Fernández
- Laboratory of Neuroscience, Biomedical Research Center (CINBIO), University of Vigo, Vigo, Galicia, Spain.
| | - J A Lamas
- Laboratory of Neuroscience, Biomedical Research Center (CINBIO), University of Vigo, Vigo, Galicia, Spain
| |
Collapse
|
18
|
Wei AD, Ramirez JM. Presynaptic Mechanisms and KCNQ Potassium Channels Modulate Opioid Depression of Respiratory Drive. Front Physiol 2019; 10:1407. [PMID: 31824331 PMCID: PMC6882777 DOI: 10.3389/fphys.2019.01407] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Accepted: 10/31/2019] [Indexed: 01/02/2023] Open
Abstract
Opioid-induced respiratory depression (OIRD) is the major cause of death associated with opioid analgesics and drugs of abuse, but the underlying cellular and molecular mechanisms remain poorly understood. We investigated opioid action in vivo in unanesthetized mice and in in vitro medullary slices containing the preBötzinger Complex (preBötC), a locus critical for breathing and inspiratory rhythm generation. Although hypothesized as a primary mechanism, we found that mu-opioid receptor (MOR1)-mediated GIRK activation contributed only modestly to OIRD. Instead, mEPSC recordings from genetically identified Dbx1-derived interneurons, essential for rhythmogenesis, revealed a prevalent presynaptic mode of action for OIRD. Consistent with MOR1-mediated suppression of presynaptic release as a major component of OIRD, Cacna1a KO slices lacking P/Q-type Ca2+ channels enhanced OIRD. Furthermore, OIRD was mimicked and reversed by KCNQ potassium channel activators and blockers, respectively. In vivo whole-body plethysmography combined with systemic delivery of GIRK- and KCNQ-specific potassium channel drugs largely recapitulated these in vitro results, and revealed state-dependent modulation of OIRD. We propose that respiratory failure from OIRD results from a general reduction of synaptic efficacy, leading to a state-dependent collapse of rhythmic network activity.
Collapse
Affiliation(s)
- Aguan D. Wei
- Seattle Children’s Research Institute, Center for Integrative Brain Research, Seattle, WA, United States
- Department of Neurological Surgery, University of Washington School of Medicine, Seattle, WA, United States
| | - Jan-Marino Ramirez
- Seattle Children’s Research Institute, Center for Integrative Brain Research, Seattle, WA, United States
- Department of Neurological Surgery, University of Washington School of Medicine, Seattle, WA, United States
| |
Collapse
|
19
|
Carver CM, Hastings SD, Cook ME, Shapiro MS. Functional responses of the hippocampus to hyperexcitability depend on directed, neuron-specific KCNQ2 K + channel plasticity. Hippocampus 2019; 30:435-455. [PMID: 31621989 DOI: 10.1002/hipo.23163] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 07/24/2019] [Accepted: 08/30/2019] [Indexed: 12/14/2022]
Abstract
M-type (KCNQ2/3) K+ channels play dominant roles in regulation of active and passive neuronal discharge properties such as resting membrane potential, spike-frequency adaptation, and hyper-excitatory states. However, plasticity of M-channel expression and function in nongenetic forms of epileptogenesis are still not well understood. Using transgenic mice with an EGFP reporter to detect expression maps of KCNQ2 mRNA, we assayed hyperexcitability-induced alterations in KCNQ2 transcription across subregions of the hippocampus. Pilocarpine and pentylenetetrazol chemoconvulsant models of seizure induction were used, and brain tissue examined 48 hr later. We observed increases in KCNQ2 mRNA in CA1 and CA3 pyramidal neurons after chemoconvulsant-induced hyperexcitability at 48 hr, but no significant change was observed in dentate gyrus (DG) granule cells. Using chromogenic in situ hybridization assays, changes to KCNQ3 transcription were not detected after hyper-excitation challenge, but the results for KCNQ2 paralleled those using the KCNQ2-mRNA reporter mice. In mice 7 days after pilocarpine challenge, levels of KCNQ2 mRNA were similar in all regions to those from control mice. In brain-slice electrophysiology recordings, CA1 pyramidal neurons demonstrated increased M-current amplitudes 48 hr after hyperexcitability; however, there were no significant changes to DG granule cell M-current amplitude. Traumatic brain injury induced significantly greater KCNQ2 expression in the hippocampal hemisphere that was ipsilateral to the trauma. In vivo, after a secondary challenge with subconvulsant dose of pentylenetetrazole, control mice were susceptible to tonic-clonic seizures, whereas mice administered the M-channel opener retigabine were protected from such seizures. This study demonstrates that increased excitatory activity promotes KCNQ2 upregulation in the hippocampus in a cell-type specific manner. Such novel ion channel expressional plasticity may serve as a compensatory mechanism after a hyperexcitable event, at least in the short term. The upregulation described could be potentially leveraged in anticonvulsant enhancement of KCNQ2 channels as therapeutic target for preventing onset of epileptogenic seizures.
Collapse
Affiliation(s)
- Chase M Carver
- Department of Cellular and Integrative Physiology, University of Texas Health San Antonio, San Antonio, Texas
| | - Shayne D Hastings
- Department of Cellular and Integrative Physiology, University of Texas Health San Antonio, San Antonio, Texas
| | - Mileah E Cook
- Department of Cellular and Integrative Physiology, University of Texas Health San Antonio, San Antonio, Texas
| | - Mark S Shapiro
- Department of Cellular and Integrative Physiology, University of Texas Health San Antonio, San Antonio, Texas
| |
Collapse
|
20
|
Martinello K, Giacalone E, Migliore M, Brown DA, Shah MM. The subthreshold-active K V7 current regulates neurotransmission by limiting spike-induced Ca 2+ influx in hippocampal mossy fiber synaptic terminals. Commun Biol 2019; 2:145. [PMID: 31044170 PMCID: PMC6486593 DOI: 10.1038/s42003-019-0408-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 03/29/2019] [Indexed: 12/23/2022] Open
Abstract
Little is known about the properties and function of ion channels that affect synaptic terminal-resting properties. One particular subthreshold-active ion channel, the Kv7 potassium channel, is highly localized to axons, but its role in regulating synaptic terminal intrinsic excitability and release is largely unexplored. Using electrophysiological recordings together with computational modeling, we found that the KV7 current was active at rest in adult hippocampal mossy fiber synaptic terminals and enhanced their membrane conductance. The current also restrained action potential-induced Ca2+ influx via N- and P/Q-type Ca2+ channels in boutons. This was associated with a substantial reduction in the spike half-width and afterdepolarization following presynaptic spikes. Further, by constraining spike-induced Ca2+ influx, the presynaptic KV7 current decreased neurotransmission onto CA3 pyramidal neurons and short-term synaptic plasticity at the mossy fiber-CA3 synapse. This is a distinctive mechanism by which KV7 channels influence hippocampal neuronal excitability and synaptic plasticity.
Collapse
Affiliation(s)
| | | | - Michele Migliore
- Institute of Biophysics, National Research Council, 90146 Palermo, Italy
| | - David A. Brown
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, WC1E 6BT UK
| | - Mala M. Shah
- UCL School of Pharmacy University College London, London, WC1N 1AX UK
| |
Collapse
|
21
|
Gq-Coupled Muscarinic Receptor Enhancement of KCNQ2/3 Channels and Activation of TRPC Channels in Multimodal Control of Excitability in Dentate Gyrus Granule Cells. J Neurosci 2018; 39:1566-1587. [PMID: 30593498 DOI: 10.1523/jneurosci.1781-18.2018] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 12/17/2018] [Accepted: 12/20/2018] [Indexed: 12/21/2022] Open
Abstract
KCNQ (Kv7, "M-type") K+ channels and TRPC (transient receptor potential, "canonical") cation channels are coupled to neuronal discharge properties and are regulated via Gq/11-protein-mediated signals. Stimulation of Gq/11-coupled receptors both consumes phosphatidylinositol 4,5-bisphosphate (PIP2) via phosphalipase Cβ hydrolysis and stimulates PIP2 synthesis via rises in Ca2+ i and other signals. Using brain-slice electrophysiology and Ca2+ imaging from male and female mice, we characterized threshold K+ currents in dentate gyrus granule cells (DGGCs) and CA1 pyramidal cells, the effects of Gq/11-coupled muscarinic M1 acetylcholine (M1R) stimulation on M current and on neuronal discharge properties, and elucidated the intracellular signaling mechanisms involved. We observed disparate signaling cascades between DGGCs and CA1 neurons. DGGCs displayed M1R enhancement of M-current, rather than suppression, due to stimulation of PIP2 synthesis, which was paralleled by increased PIP2-gated G-protein coupled inwardly rectifying K+ currents as well. Deficiency of KCNQ2-containing M-channels ablated the M1R-induced enhancement of M-current in DGGCs. Simultaneously, M1R stimulation in DGGCs induced robust increases in [Ca2+]i, mostly due to TRPC currents, consistent with, and contributing to, neuronal depolarization and hyperexcitability. CA1 neurons did not display such multimodal signaling, but rather M current was suppressed by M1R stimulation in these cells, similar to the previously described actions of M1R stimulation on M-current in peripheral ganglia that mostly involves PIP2 depletion. Therefore, these results point to a pleiotropic network of cholinergic signals that direct cell-type-specific, precise control of hippocampal function with strong implications for hyperexcitability and epilepsy.SIGNIFICANCE STATEMENT At the neuronal membrane, protein signaling cascades consisting of ion channels and metabotropic receptors govern the electrical properties and neurotransmission of neuronal networks. Muscarinic acetylcholine receptors are G-protein-coupled metabotropic receptors that control the excitability of neurons through regulating ion channels, intracellular Ca2+ signals, and other second-messenger cascades. We have illuminated previously unknown actions of muscarinic stimulation on the excitability of hippocampal principal neurons that include M channels, TRPC (transient receptor potential, "canonical") cation channels, and powerful regulation of lipid metabolism. Our results show that these signaling pathways, and mechanisms of excitability, are starkly distinct between peripheral ganglia and brain, and even between different principal neurons in the hippocampus.
Collapse
|
22
|
Diao Y, Yan W, Sun W, Luo Y, Li J, Yin Y. The dual role of KCNQ/M channels upon OGD or OGD/R insults in cultured cortical neurons of mice: Timing is crucial in targeting M-channels against ischemic injur ies. J Cell Physiol 2018; 234:12714-12726. [PMID: 30523632 DOI: 10.1002/jcp.27889] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 11/13/2018] [Indexed: 01/09/2023]
Abstract
KCNQ/M potassium channels play a vital role in neuronal excitability; however, it is required to explore their pharmacological modulation on N-Methyl- d-aspartic acid receptors (NMDARs)-mediated glutamatergic transmission of neurons upon ischemic insults. In the current study, both presynaptic glutamatergic release and activities of NMDARs were measured by NMDAR-induced miniature excitatory postsynaptic currents (mEPSCs) in cultured cortical neurons of C57 mice undergoing oxygen and glucose deprivation (OGD) or OGD/reperfusion (OGD/R). The KCNQ/M-channel opener, retigabine (RTG), suppressed the overactivation of postsynaptic NMDARs induced by OGD and then NO transient; RTG also decreased OGD-induced neuronal death measured with MTT assay, suggesting the beneficial role of KCNQ/M-channels for the neurons exposed to ischemic insults. However, when the neurons exposed to the subsequent reperfusion, KCNQ/M-channels played a differential role from its protective effect. OGD/R increased presynaptic glutamatergic release, which was further augmented by RTG or decreased by KCNQ/M-channel blocker, XE991. Reactive oxygen species (ROS) were produced partly in a NO-dependent manner. In addition, XE991 decreased neuronal injuries upon reperfusion measured with DCF and PI staining. Meanwhile, the addition of RTG upon OGD or XE991 upon reperfusion can reverse OGD or OGD/R-reduced mitochondrial membrane potential. Our present study indicates the dual role of KCNQ/M-channels in OGD and OGD/R, which will decide the fate of neurons. Provided that activation of KCNQ/M-channels has differential effects on neuronal injuries during OGD or OGD/R, we propose that therapy targeting KCNQ/M-channels may be effective for ischemic injuries but the proper timing is so crucial for the corresponding treatment.
Collapse
Affiliation(s)
- Yu Diao
- Department of Neurobiology, Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Weijie Yan
- Department of Neurobiology, Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Wei Sun
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yanlin Luo
- Department of Neurobiology, Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Junfa Li
- Department of Neurobiology, Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Yanling Yin
- Department of Neurobiology, Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| |
Collapse
|
23
|
Dewell RB, Gabbiani F. M current regulates firing mode and spike reliability in a collision-detecting neuron. J Neurophysiol 2018; 120:1753-1764. [PMID: 30044671 DOI: 10.1152/jn.00363.2018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
All animals must detect impending collisions to escape and reliably discriminate them from nonthreatening stimuli, thus preventing false alarms. Therefore, it is no surprise that animals have evolved highly selective and sensitive neurons dedicated to such tasks. We examined a well-studied collision-detection neuron in the grasshopper ( Schistocerca americana) using in vivo electrophysiology, pharmacology, and computational modeling. This lobula giant movement detector (LGMD) neuron is excitable by inputs originating from each ommatidia of the compound eye. It possesses many intrinsic properties that increase its selectivity to objects approaching on a collision course, including switching between burst and nonburst firing. In this study, we demonstrate that the LGMD neuron exhibits a large M current, generated by noninactivating K+ channels, that shortens the temporal window of dendritic integration, regulates a firing mode switch between burst and isolated spiking, increases the precision of spike timing, and increases the reliability of spike propagation to downstream motor centers. By revealing how the M current increases the LGMD's ability to detect impending collisions, our results suggest that similar channels may play an analogous role in other collision detection circuits. NEW & NOTEWORTHY The ability to reliably detect impending collisions is a critical survival skill. The nervous systems of many animals have developed dedicated neurons for accomplishing this task. We used a mix of in vivo electrophysiology and computational modeling to investigate the role of M potassium channels within one such collision-detecting neuron and show that through regulation of burst firing and enhancement of spiking reliability, the M current increases the ability to detect impending collisions.
Collapse
Affiliation(s)
- Richard B Dewell
- Department of Neuroscience, Baylor College of Medicine , Houston, Texas
| | - Fabrizio Gabbiani
- Department of Neuroscience, Baylor College of Medicine , Houston, Texas.,Department of Electrical and Computer Engineering, Rice University , Houston, Texas
| |
Collapse
|
24
|
Ghezzi F, Monni L, Nistri A. Functional up-regulation of the M-current by retigabine contrasts hyperexcitability and excitotoxicity on rat hypoglossal motoneurons. J Physiol 2018; 596:2611-2629. [PMID: 29736957 DOI: 10.1113/jp275906] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 04/23/2018] [Indexed: 12/14/2022] Open
Abstract
KEY POINTS Excessive neuronal excitability characterizes several neuropathological conditions, including neurodegenerative diseases such as amyotrophic lateral sclerosis. Hypoglossal motoneurons (HMs), which control tongue muscles, are extremely vulnerable to this disease and undergo damage and death when exposed to an excessive glutamate extracellular concentration that causes excitotoxicity. Our laboratory devised an in vitro model of excitotoxicity obtained by pharmacological blockade of glutamate transporters. In this paradigm, HMs display hyperexcitability, collective bursting and eventually cell death. The results of the present study show that pharmacological up-regulation of a K+ current (M-current), via application of the anti-convulsant retigabine, prevented all hallmarks of HM excitotoxicity, comprising bursting, generation of reactive oxygen species, expression of toxic markers and cell death. ○Our data may have translational value to develop new treatments against neurological diseases by using positive pharmacological modulators of the M-current. ABSTRACT Neuronal hyperexcitability is a symptom characterizing several neurodegenerative disorders, including amyotrophic lateral sclerosis (ALS). In the ALS bulbar form, hypoglossal motoneurons (HMs) are an early target for neurodegeneration because of their high vulnerability to metabolic insults. In recent years, our laboratory has developed an in vitro model of a brainstem slice comprising the hypoglossal nucleus in which HM neurodegeneration is achieved by blocking glutamate clearance with dl-threo-β-benzyloxyaspartate (TBOA), thus leading to delayed excitotoxicity. During this process, HMs display a set of hallmarks such as hyperexcitability (and network bursting), reactive oxygen species (ROS) generation and, finally, cell death. The present study aimed to investigate whether blocking early hyperexcitability and bursting with the anti-convulsant drug retigabine was sufficient to achieve neuroprotection against excitotoxicity. Retigabine is a selective positive allosteric modulator of the M-current (IM ), an endogenous mechanism that neurons (comprising HMs) express to dampen excitability. Retigabine (10 μm; co-applied with TBOA) contrasted ROS generation, release of endogenous toxic factors into the HM cytoplasm and excitotoxicity-induced HM death. Electrophysiological experiments showed that retigabine readily contrasted and arrested bursting evoked by TBOA administration. Because neuronal IM subunits (Kv7.2, Kv7.3 and Kv7.5) were expressed in the hypoglossal nucleus and in functionally connected medullary nuclei, we suggest that they were responsible for the strong reduction in network excitability, a potent phenomenon for achieving neuroprotection against TBOA-induced excitotoxicity. The results of the present study may have translational value for testing novel positive pharmacological modulators of the IM under pathological conditions (including neurodegenerative disorders) characterized by excessive neuronal excitability.
Collapse
Affiliation(s)
- Filippo Ghezzi
- Department of Neuroscience, International School for Advanced Studies (SISSA), Trieste, Italy
| | - Laura Monni
- Department of Neuroscience, International School for Advanced Studies (SISSA), Trieste, Italy
| | - Andrea Nistri
- Department of Neuroscience, International School for Advanced Studies (SISSA), Trieste, Italy
| |
Collapse
|
25
|
Pharmacological modulation of the voltage-gated neuronal Kv7/KCNQ/M-channel alters the intrinsic excitability and synaptic responses of pyramidal neurons in rat prefrontal cortex slices. Acta Pharmacol Sin 2017; 38:1248-1256. [PMID: 28603289 DOI: 10.1038/aps.2017.72] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Accepted: 02/09/2017] [Indexed: 12/20/2022]
Abstract
The prefrontal cortex (PFC) critical for higher cognition is implicated in neuropsychiatric diseases, such as Alzheimer's disease, depression and schizophrenia. The voltage-activated Kv7/KCNQ/M-channel or M-current modulates the neuronal excitability that defines the fundamental mechanism of brain function. However, whether M-current functions to regulate the excitability of PFC neurons remains elusive. In this study, we recorded the native M-current from PFC layer V pyramidal neurons in rat brain slices and showed that it modulated the intrinsic excitability and synaptic responses of PFC pyramidal neurons. Application of a specific M-channel blocker XE991 (40 μmol/L) or opener retigabine (10 μmol/L) resulted in inhibition or activation of M-current, respectively. In the current-clamp recordings, inhibition of M-current was evidenced by the increased average spike frequency and the reduced first inter-spike interval (ISI), spike onset latency and fast afterhyperpolarization (fAHP), whereas activation of M-current caused opposite responses. Furthermore, inhibition of M-current significantly increased the amplitude of excitatory postsynaptic potentials (EPSPs) and depolarized the resting membrane potential (RMP) without affecting the miniature EPSC (mEPSC) frequency. These data demonstrate that voltage-gated neuronal Kv7/KCNQ/M-current modulates the excitability and synaptic transmission of PFC neurons, suggesting that pharmacological modulation of M-current in the PFC may exert beneficial effects on cognitive deficits implicated in the pathophysiology of neuropsychiatric disorders.
Collapse
|
26
|
Kang S, Li J, Zuo W, Fu R, Gregor D, Krnjevic K, Bekker A, Ye JH. Ethanol Withdrawal Drives Anxiety-Related Behaviors by Reducing M-type Potassium Channel Activity in the Lateral Habenula. Neuropsychopharmacology 2017; 42:1813-1824. [PMID: 28387223 PMCID: PMC5520788 DOI: 10.1038/npp.2017.68] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 03/17/2017] [Accepted: 03/26/2017] [Indexed: 02/04/2023]
Abstract
Alcohol use disorders (AUDs) and anxiety disorders (ADs) are often seen concurrently, but their underlying cellular basis is unclear. For unclear reasons, the lateral habenula (LHb), a key brain region involved in the pathophysiology of ADs, becomes hyperactive after ethanol withdrawal. M-type K+ channels (M-channels), important regulators of neuronal activity, are abundant in the LHb, yet little is known about their role in AUDs and associated ADs. We report here that in rats at 24 h withdrawal from systemic ethanol administration (either by intraperitoneal injection, 2 g/kg, twice/day, for 7 days; or intermittent drinking 20% ethanol in a two-bottle free choice protocol for 8 weeks), the basal firing rate and the excitability of LHb neurons in brain slices was higher, whereas the amplitude of medium afterhyperpolarization and M-type K+ currents were smaller, when compared to ethanol naive rats. Concordantly, M-channel blocker (XE991)-induced increase in the spontaneous firing rate in LHb neurons was smaller. The protein expression of M-channel subunits, KCNQ2/3 in the LHb was also smaller. Moreover, anxiety levels (tested in open field, marble burying, and elevated plus maze) were higher, which were alleviated by LHb inhibition either chemogenetically or by local infusion of the M-channel opener, retigabine. Intra-LHb infusion of retigabine also reduced ethanol consumption and preference. These findings reveal an important role of LHb M-channels in the expression of AUDs and ADs, and suggest that the M-channels could be a potential therapeutic target for alcoholics.
Collapse
Affiliation(s)
- Seungwoo Kang
- Department of Anesthesiology, Rutgers, The State University of New Jersey, New Jersey Medical School, Newark, NJ, USA,Department of Pharmacology, Physiology, and Neuroscience, Rutgers, The State University of New Jersey, New Jersey Medical School, Newark, NJ, USA
| | - Jing Li
- Department of Anesthesiology, Rutgers, The State University of New Jersey, New Jersey Medical School, Newark, NJ, USA,Department of Pharmacology, Physiology, and Neuroscience, Rutgers, The State University of New Jersey, New Jersey Medical School, Newark, NJ, USA
| | - Wanhong Zuo
- Department of Anesthesiology, Rutgers, The State University of New Jersey, New Jersey Medical School, Newark, NJ, USA,Department of Pharmacology, Physiology, and Neuroscience, Rutgers, The State University of New Jersey, New Jersey Medical School, Newark, NJ, USA
| | - Rao Fu
- Department of Anesthesiology, Rutgers, The State University of New Jersey, New Jersey Medical School, Newark, NJ, USA,Department of Pharmacology, Physiology, and Neuroscience, Rutgers, The State University of New Jersey, New Jersey Medical School, Newark, NJ, USA
| | - Danielle Gregor
- Department of Anesthesiology, Rutgers, The State University of New Jersey, New Jersey Medical School, Newark, NJ, USA,Department of Pharmacology, Physiology, and Neuroscience, Rutgers, The State University of New Jersey, New Jersey Medical School, Newark, NJ, USA
| | | | - Alex Bekker
- Department of Anesthesiology, Rutgers, The State University of New Jersey, New Jersey Medical School, Newark, NJ, USA
| | - Jiang-Hong Ye
- Department of Anesthesiology, Rutgers, The State University of New Jersey, New Jersey Medical School, Newark, NJ, USA,Department of Pharmacology, Physiology, and Neuroscience, Rutgers, The State University of New Jersey, New Jersey Medical School, Newark, NJ, USA,Department of Anesthesiology, Rutgers, The State University of New Jersey, New Jersey Medical School, 185 South Orange Avenue, Newark, NJ, 07103, USA, Tel: 973 972 1866, Fax: 973 972 0582, E-mail:
| |
Collapse
|
27
|
Owen B, Reddy R, Grover LM. Nonspecific block of voltage-gated potassium channels has greater effect on distal schaffer collaterals than proximal schaffer collaterals during periods of high activity. Physiol Rep 2017; 5:5/14/e13354. [PMID: 28747510 PMCID: PMC5532488 DOI: 10.14814/phy2.13354] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 06/21/2017] [Indexed: 02/05/2023] Open
Abstract
Previous studies established different responses between proximal and distal portions of Schaffer collateral axons during high‐frequency and burst stimulation, with distal axons demonstrating biphasic changes in excitability (hyperexcitability followed by depression), but proximal axons showing only monophasic depression. Voltage‐dependent potassium (KV) channels are important determinants of axonal excitability, and block of KV channels can promote axon hyperexcitability. We therefore hypothesized that block of KV channels should lead to biphasic response changes in proximal Schaffer collaterals, like those seen in distal Schaffer collaterals. To test this hypothesis, we made extracellular recordings of distal Schaffer collateral responses in stratum radiatum of hippocampal area CA1 and proximal Schaffer collateral responses in stratum pyramidale of area CA3 during high‐frequency stimulation (HFS) at 100 Hz and burst stimulation at 200 msec intervals (5 Hz or theta frequency). We then applied a nonselective KV channel blocker, tetraethlylammonium (TEA, 10 mmol/L) or 4‐aminopyridine (4‐AP, 100 μmol/L), and assessed effects on Schaffer collateral responses. Surprisingly, block of KV channels had little or no effect on proximal Schaffer collateral responses during high‐frequency or burst stimulation. In contrast, KV channel blockade caused more rapid depression of distal Schaffer collateral responses during both high‐frequency and burst stimulation. These findings indicate that KV channels are important for maintaining distal, but not proximal, Schaffer collateral excitability during period of sustained high activity. Differential sensitivity of distal versus proximal Schaffer collaterals to KV channel block may reflect differences in channel density, diversity, or subcellular localization.
Collapse
Affiliation(s)
- Benjamin Owen
- Department of Biomedical Sciences, Marshall University School of Medicine, Huntington, West Virginia, 25755
| | - Rishi Reddy
- Department of Biomedical Sciences, Marshall University School of Medicine, Huntington, West Virginia, 25755
| | - Lawrence M Grover
- Department of Biomedical Sciences, Marshall University School of Medicine, Huntington, West Virginia, 25755
| |
Collapse
|
28
|
Zhang Y, Bucher D, Nadim F. Ionic mechanisms underlying history-dependence of conduction delay in an unmyelinated axon. eLife 2017; 6. [PMID: 28691900 PMCID: PMC5519330 DOI: 10.7554/elife.25382] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 07/06/2017] [Indexed: 11/13/2022] Open
Abstract
Axonal conduction velocity can change substantially during ongoing activity, thus modifying spike interval structures and, potentially, temporal coding. We used a biophysical model to unmask mechanisms underlying the history-dependence of conduction. The model replicates activity in the unmyelinated axon of the crustacean stomatogastric pyloric dilator neuron. At the timescale of a single burst, conduction delay has a non-monotonic relationship with instantaneous frequency, which depends on the gating rates of the fast voltage-gated Na+ current. At the slower timescale of minutes, the mean value and variability of conduction delay increase. These effects are because of hyperpolarization of the baseline membrane potential by the Na+/K+ pump, balanced by an h-current, both of which affect the gating of the Na+ current. We explore the mechanisms of history-dependence of conduction delay in axons and develop an empirical equation that accurately predicts this history-dependence, both in the model and in experimental measurements.
Collapse
Affiliation(s)
- Yang Zhang
- Department of Mathematical Sciences, New Jersey Institute of Technology, Newark, United States
| | - Dirk Bucher
- Federated Department of Biological Sciences, NJIT and Rutgers University, Newark, United States
| | - Farzan Nadim
- Department of Mathematical Sciences, New Jersey Institute of Technology, Newark, United States.,Federated Department of Biological Sciences, NJIT and Rutgers University, Newark, United States
| |
Collapse
|
29
|
Ghezzi F, Corsini S, Nistri A. Electrophysiological characterization of the M-current in rat hypoglossal motoneurons. Neuroscience 2017; 340:62-75. [DOI: 10.1016/j.neuroscience.2016.10.048] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 10/14/2016] [Accepted: 10/19/2016] [Indexed: 10/20/2022]
|
30
|
Greene DL, Hoshi N. Modulation of Kv7 channels and excitability in the brain. Cell Mol Life Sci 2016; 74:495-508. [PMID: 27645822 DOI: 10.1007/s00018-016-2359-y] [Citation(s) in RCA: 108] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 08/25/2016] [Accepted: 09/06/2016] [Indexed: 11/26/2022]
Abstract
Neuronal Kv7 channels underlie a voltage-gated non-inactivating potassium current known as the M-current. Due to its particular characteristics, Kv7 channels show pronounced control over the excitability of neurons. We will discuss various factors that have been shown to drastically alter the activity of this channel such as protein and phospholipid interactions, phosphorylation, calcium, and numerous neurotransmitters. Kv7 channels locate to key areas for the control of action potential initiation and propagation. Moreover, we will explore the dynamic surface expression of the channel modulated by neurotransmitters and neural activity. We will also focus on known principle functions of neural Kv7 channels: control of resting membrane potential and spiking threshold, setting the firing frequency, afterhyperpolarization after burst firing, theta resonance, and transient hyperexcitability from neurotransmitter-induced suppression of the M-current. Finally, we will discuss the contribution of altered Kv7 activity to pathologies such as epilepsy and cognitive deficits.
Collapse
Affiliation(s)
- Derek L Greene
- Department of Pharmacology, University of California, 360 Med Surge II, Irvine, CA, 92697, USA
| | - Naoto Hoshi
- Department of Pharmacology, University of California, 360 Med Surge II, Irvine, CA, 92697, USA.
- Department of Physiology and Biophysics, University of California, Irvine, USA.
| |
Collapse
|
31
|
Atherton LA, Prince LY, Tsaneva-Atanasova K. Bifurcation analysis of a two-compartment hippocampal pyramidal cell model. J Comput Neurosci 2016; 41:91-106. [PMID: 27221619 PMCID: PMC4927618 DOI: 10.1007/s10827-016-0606-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Revised: 04/19/2016] [Accepted: 04/25/2016] [Indexed: 01/27/2023]
Abstract
The Pinsky-Rinzel model is a non-smooth 2-compartmental CA3 pyramidal cell model that has been used widely within the field of neuroscience. Here we propose a modified (smooth) system that captures the qualitative behaviour of the original model, while allowing the use of available, numerical continuation methods to perform full-system bifurcation and fast-slow analysis. We study the bifurcation structure of the full system as a function of the applied current and the maximal calcium conductance. We identify the bifurcations that shape the transitions between resting, bursting and spiking behaviours, and which lead to the disappearance of bursting when the calcium conductance is reduced. Insights gained from this analysis, are then used to firstly illustrate how the irregular spiking activity found between bursting and stable spiking states, can be influenced by phase differences in the calcium and dendritic voltage, which lead to corresponding changes in the calcium-sensitive potassium current. Furthermore, we use fast-slow analysis to investigate the mechanisms of bursting and show that bursting in the model is dependent on the intermediately slow variable, calcium, while the other slow variable, the activation gate of the afterhyperpolarisation current, does not contribute to setting the intraburst dynamics but participates in setting the interburst interval. Finally, we discuss how some of the described bifurcations affect spiking behaviour, during sharp-wave ripples, in a larger network of Pinsky-Rinzel cells.
Collapse
Affiliation(s)
- Laura A Atherton
- Engineering Mathematics, and Physiology, Pharmacology & Neuroscience, University of Bristol, Bristol, England, UK
| | - Luke Y Prince
- Physiology, Pharmacology & Neuroscience, University of Bristol, Bristol, England, UK
| | - Krasimira Tsaneva-Atanasova
- Department of Mathematics, College of Engineering, Mathematics and Physical Sciences, & EPSRC Centre for Predictive Modelling in Healthcare, University of Exeter, Exeter, Devon, EX4 4QF, UK.
| |
Collapse
|
32
|
Rogawski MA, Löscher W, Rho JM. Mechanisms of Action of Antiseizure Drugs and the Ketogenic Diet. Cold Spring Harb Perspect Med 2016; 6:a022780. [PMID: 26801895 PMCID: PMC4852797 DOI: 10.1101/cshperspect.a022780] [Citation(s) in RCA: 186] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Antiseizure drugs (ASDs), also termed antiepileptic drugs, are the main form of symptomatic treatment for people with epilepsy, but not all patients become free of seizures. The ketogenic diet is one treatment option for drug-resistant patients. Both types of therapy exert their clinical effects through interactions with one or more of a diverse set of molecular targets in the brain. ASDs act by modulation of voltage-gated ion channels, including sodium, calcium, and potassium channels; by enhancement of γ-aminobutyric acid (GABA)-mediated inhibition through effects on GABAA receptors, the GABA transporter 1 (GAT1) GABA uptake transporter, or GABA transaminase; through interactions with elements of the synaptic release machinery, including synaptic vesicle 2A (SV2A) and α2δ; or by blockade of ionotropic glutamate receptors, including α-amino-3-hydroxy-5-methyl-4-isoxazole-propionate (AMPA) receptors. The ketogenic diet leads to increases in circulating ketones, which may contribute to the efficacy in treating pharmacoresistant seizures. Production in the brain of inhibitory mediators, such as adenosine, or ion channel modulators, such as polyunsaturated fatty acids, may also play a role. Metabolic effects, including diversion from glycolysis, are a further postulated mechanism. For some ASDs and the ketogenic diet, effects on multiple targets may contribute to activity. Better understanding of the ketogenic diet will inform the development of improved drug therapies to treat refractory seizures.
Collapse
Affiliation(s)
- Michael A Rogawski
- Department of Neurology, University of California, Davis, Sacramento, California 95817
| | - Wolfgang Löscher
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine, Hannover, Germany
| | - Jong M Rho
- Department of Pediatrics, University of Calgary, Calgary, Alberta, Canada Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada Department of Physiology and Pharmacology, University of Calgary, Alberta, Canada
| |
Collapse
|
33
|
Ohura S, Kamiya H. Excitability tuning of axons in the central nervous system. J Physiol Sci 2016; 66:189-96. [PMID: 26493201 PMCID: PMC10717993 DOI: 10.1007/s12576-015-0415-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Accepted: 10/01/2015] [Indexed: 12/15/2022]
Abstract
The axon is a long neuronal process that originates from the soma and extends towards the presynaptic terminals. The pioneering studies on the squid giant axon or the spinal cord motoneuron established that the axon conducts action potentials faithfully to the presynaptic terminals with self-regenerative processes of membrane excitation. Recent studies challenged the notion that the fundamental understandings obtained from the study of squid giant axons are readily applicable to the axons in the mammalian central nervous system (CNS). These studies revealed that the functional and structural properties of the CNS axons are much more variable than previously thought. In this review article, we summarize the recent understandings of axon physiology in the mammalian CNS due to progress in the subcellular recording techniques which allow direct recordings from the axonal membranes, with emphasis on the hippocampal mossy fibers as a representative en passant axons typical for cortical axons.
Collapse
Affiliation(s)
- Shunsuke Ohura
- Department of Neurobiology, Hokkaido University Graduate School of Medicine, N15 W7 Kita-ku, Sapporo, 060-8638, Japan
| | - Haruyuki Kamiya
- Department of Neurobiology, Hokkaido University Graduate School of Medicine, N15 W7 Kita-ku, Sapporo, 060-8638, Japan.
| |
Collapse
|
34
|
Knudstrup S, Zochowski M, Booth V. Network burst dynamics under heterogeneous cholinergic modulation of neural firing properties and heterogeneous synaptic connectivity. Eur J Neurosci 2016; 43:1321-39. [PMID: 26869313 DOI: 10.1111/ejn.13210] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Revised: 01/19/2016] [Accepted: 02/08/2016] [Indexed: 01/16/2023]
Abstract
The characteristics of neural network activity depend on intrinsic neural properties and synaptic connectivity in the network. In brain networks, both of these properties are critically affected by the type and levels of neuromodulators present. The expression of many of the most powerful neuromodulators, including acetylcholine (ACh), varies tonically and phasically with behavioural state, leading to dynamic, heterogeneous changes in intrinsic neural properties and synaptic connectivity properties. Namely, ACh significantly alters neural firing properties as measured by the phase response curve in a manner that has been shown to alter the propensity for network synchronization. The aim of this simulation study was to build an understanding of how heterogeneity in cholinergic modulation of neural firing properties and heterogeneity in synaptic connectivity affect the initiation and maintenance of synchronous network bursting in excitatory networks. We show that cells that display different levels of ACh modulation have differential roles in generating network activity: weakly modulated cells are necessary for burst initiation and provide synchronizing drive to the rest of the network, whereas strongly modulated cells provide the overall activity level necessary to sustain burst firing. By applying several quantitative measures of network activity, we further show that the existence of network bursting and its characteristics, such as burst duration and intraburst synchrony, are dependent on the fraction of cell types providing the synaptic connections in the network. These results suggest mechanisms underlying ACh modulation of brain oscillations and the modulation of seizure activity during sleep states.
Collapse
Affiliation(s)
- Scott Knudstrup
- Department of Mathematics, University of Michigan, 530 Church St, Ann Arbor, MI, 48109, USA
| | - Michal Zochowski
- Department of Physics and Biophysics Program, University of Michigan, 450 Church St, Ann Arbor, MI, 48109, USA
| | - Victoria Booth
- Department of Mathematics, University of Michigan, 530 Church St, Ann Arbor, MI, 48109, USA.,Department of Anesthesiology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
35
|
King B, Rizwan AP, Asmara H, Heath NC, Engbers JDT, Dykstra S, Bartoletti TM, Hameed S, Zamponi GW, Turner RW. IKCa channels are a critical determinant of the slow AHP in CA1 pyramidal neurons. Cell Rep 2016; 11:175-82. [PMID: 25865881 DOI: 10.1016/j.celrep.2015.03.026] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2014] [Revised: 01/30/2015] [Accepted: 03/10/2015] [Indexed: 12/23/2022] Open
Abstract
Control over the frequency and pattern of neuronal spike discharge depends on Ca2+-gated K+ channels that reduce cell excitability by hyperpolarizing the membrane potential. The Ca2+-dependent slow afterhyperpolarization (sAHP) is one of the most prominent inhibitory responses in the brain, with sAHP amplitude linked to a host of circuit and behavioral functions, yet the channel that underlies the sAHP has defied identification for decades. Here, we show that intermediate-conductance Ca2+-dependent K+ (IKCa) channels underlie the sAHP generated by trains of synaptic input or postsynaptic stimuli in CA1 hippocampal pyramidal cells. These findings are significant in providing a molecular identity for the sAHP of central neurons that will identify pharmacological tools capable of potentially modifying the several behavioral or disease states associated with the sAHP.
Collapse
|
36
|
Friedman LK, Slomko AM, Wongvravit JP, Naseer Z, Hu S, Wan WY, Ali SS. Efficacy of Retigabine on Acute Limbic Seizures in Adult Rats. J Epilepsy Res 2015; 5:46-59. [PMID: 26819936 PMCID: PMC4724852 DOI: 10.14581/jer.15010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 09/01/2015] [Indexed: 12/17/2022] Open
Abstract
Background and Purpose: The efficacy of retigabine (RGB), a positive allosteric modulator of K+ channels indicated for adjunct treatment of partial seizures, was studied in two adult models of kainic acid (KA)-induced status epilepticus to determine it’s toleratbility. Methods: Retigabine was administered systemiclly at high (5 mg/kg) and low (1–2 mg/kg) doses either 30 min prior to or 2 hr after KA-induced status epilepticus. High (1 µg/µL) and low (0.25 µg/µL) concentrations of RGB were also delivered by intrahippocampal microinjection in the presence of KA. Results: Dose-dependent effects of RGB were observed with both models. Lower doses increased seizure behavior latency and reduced the number of single spikes and synchronized burst events in the electroencephalogram (EEG). Higher doses worsened seizure behavior, produced severe ataxia, and increased spiking activity. Animals treated with RGB that were resistant to seizures did not exhibit significant injury or loss in GluR1 expression; however if stage 5–6 seizures were reached, typical hippocampal injury and depletion of GluR1 subunit protein in vulernable pyramidal fields occurred. Conclusions: RGB was neuroprotective only if seizures were significantly attenuated. GluR1 was simultaneously suppressed in the resistant granule cell layer in presence of RGB which may weaken excitatory transmission. Biphasic effects observed herein suggest that the human dosage must be carefully scrutinized to produce the optimal clinical response.
Collapse
Affiliation(s)
- L K Friedman
- Department of Cell Biology & Anatomy, New York Medical College, Valhalla, NY, USA
| | - A M Slomko
- Department of Cell Biology & Anatomy, New York Medical College, Valhalla, NY, USA
| | - J P Wongvravit
- Department of Cell Biology & Anatomy, New York Medical College, Valhalla, NY, USA
| | - Z Naseer
- Department of Cell Biology & Anatomy, New York Medical College, Valhalla, NY, USA
| | - S Hu
- Department of Cell Biology & Anatomy, New York Medical College, Valhalla, NY, USA
| | - W Y Wan
- Department of Cell Biology & Anatomy, New York Medical College, Valhalla, NY, USA
| | - S S Ali
- Department of Cell Biology & Anatomy, New York Medical College, Valhalla, NY, USA
| |
Collapse
|
37
|
Loss of Local Astrocyte Support Disrupts Action Potential Propagation and Glutamate Release Synchrony from Unmyelinated Hippocampal Axon Terminals In Vitro. J Neurosci 2015; 35:11105-17. [PMID: 26245971 DOI: 10.1523/jneurosci.1289-15.2015] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
UNLABELLED Neuron-astrocyte interactions are critical for proper CNS development and function. Astrocytes secrete factors that are pivotal for synaptic development and function, neuronal metabolism, and neuronal survival. Our understanding of this relationship, however, remains incomplete due to technical hurdles that have prevented the removal of astrocytes from neuronal circuits without changing other important conditions. Here we overcame this obstacle by growing solitary rat hippocampal neurons on microcultures that were comprised of either an astrocyte bed (+astrocyte) or a collagen bed (-astrocyte) within the same culture dish. -Astrocyte autaptic evoked EPSCs, but not IPSCs, displayed an altered temporal profile, which included increased synaptic delay, increased time to peak, and severe glutamate release asynchrony, distinct from previously described quantal asynchrony. Although we observed minimal alteration of the somatically recorded action potential waveform, action potential propagation was altered. We observed a longer latency between somatic initiation and arrival at distal locations, which likely explains asynchronous EPSC peaks, and we observed broadening of the axonal spike, which likely underlies changes to evoked EPSC onset. No apparent changes in axon structure were observed, suggesting altered axonal excitability. In conclusion, we propose that local astrocyte support has an unappreciated role in maintaining glutamate release synchrony by disturbing axonal signal propagation. SIGNIFICANCE STATEMENT Certain glial cell types (oligodendrocytes, Schwann cells) facilitate the propagation of neuronal electrical signals, but a role for astrocytes has not been identified despite many other functions of astrocytes in supporting and modulating neuronal signaling. Under identical global conditions, we cultured neurons with or without local astrocyte support. Without local astrocytes, glutamate transmission was desynchronized by an alteration of the waveform and arrival time of axonal action potentials to synaptic terminals. GABA transmission was not disrupted. The disruption did not involve detectable morphological changes to axons of glutamate neurons. Our work identifies a developmental role for astrocytes in the temporal precision of excitatory signals.
Collapse
|
38
|
Wójtowicz T, Brzdąk P, Mozrzymas JW. Diverse impact of acute and long-term extracellular proteolytic activity on plasticity of neuronal excitability. Front Cell Neurosci 2015; 9:313. [PMID: 26321914 PMCID: PMC4530619 DOI: 10.3389/fncel.2015.00313] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 07/28/2015] [Indexed: 12/13/2022] Open
Abstract
Learning and memory require alteration in number and strength of existing synaptic connections. Extracellular proteolysis within the synapses has been shown to play a pivotal role in synaptic plasticity by determining synapse structure, function, and number. Although synaptic plasticity of excitatory synapses is generally acknowledged to play a crucial role in formation of memory traces, some components of neural plasticity are reflected by nonsynaptic changes. Since information in neural networks is ultimately conveyed with action potentials, scaling of neuronal excitability could significantly enhance or dampen the outcome of dendritic integration, boost neuronal information storage capacity and ultimately learning. However, the underlying mechanism is poorly understood. With this regard, several lines of evidence and our most recent study support a view that activity of extracellular proteases might affect information processing in neuronal networks by affecting targets beyond synapses. Here, we review the most recent studies addressing the impact of extracellular proteolysis on plasticity of neuronal excitability and discuss how enzymatic activity may alter input-output/transfer function of neurons, supporting cognitive processes. Interestingly, extracellular proteolysis may alter intrinsic neuronal excitability and excitation/inhibition balance both rapidly (time of minutes to hours) and in long-term window. Moreover, it appears that by cleavage of extracellular matrix (ECM) constituents, proteases may modulate function of ion channels or alter inhibitory drive and hence facilitate active participation of dendrites and axon initial segments (AISs) in adjusting neuronal input/output function. Altogether, a picture emerges whereby both rapid and long-term extracellular proteolysis may influence some aspects of information processing in neurons, such as initiation of action potential, spike frequency adaptation, properties of action potential and dendritic backpropagation.
Collapse
Affiliation(s)
- Tomasz Wójtowicz
- Laboratory of Neuroscience, Department of Biophysics, Wroclaw Medical University Wroclaw, Poland
| | - Patrycja Brzdąk
- Department of Animal Physiology, Institute of Experimental Biology, Wroclaw University Wroclaw, Poland
| | - Jerzy W Mozrzymas
- Laboratory of Neuroscience, Department of Biophysics, Wroclaw Medical University Wroclaw, Poland ; Department of Animal Physiology, Institute of Experimental Biology, Wroclaw University Wroclaw, Poland
| |
Collapse
|
39
|
β-Secretase BACE1 regulates hippocampal and reconstituted M-currents in a β-subunit-like fashion. J Neurosci 2015; 35:3298-311. [PMID: 25716831 DOI: 10.1523/jneurosci.3127-14.2015] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The β-secretase BACE1 is widely known for its pivotal role in the amyloidogenic pathway leading to Alzheimer's disease, but how its action on transmembrane proteins other than the amyloid precursor protein affects the nervous system is only beginning to be understood. We report here that BACE1 regulates neuronal excitability through an unorthodox, nonenzymatic interaction with members of the KCNQ (Kv7) family that give rise to the M-current, a noninactivating potassium current with slow kinetics. In hippocampal neurons from BACE1(-/-) mice, loss of M-current enhanced neuronal excitability. We relate the diminished M-current to the previously reported epileptic phenotype of BACE1-deficient mice. In HEK293T cells, BACE1 amplified reconstituted M-currents, altered their voltage dependence, accelerated activation, and slowed deactivation. Biochemical evidence strongly suggested that BACE1 physically associates with channel proteins in a β-subunit-like fashion. Our results establish BACE1 as a physiologically essential constituent of regular M-current function and elucidate a striking new feature of how BACE1 impacts on neuronal activity in the intact and diseased brain.
Collapse
|
40
|
Early-onset epileptic encephalopathy caused by gain-of-function mutations in the voltage sensor of Kv7.2 and Kv7.3 potassium channel subunits. J Neurosci 2015; 35:3782-93. [PMID: 25740509 DOI: 10.1523/jneurosci.4423-14.2015] [Citation(s) in RCA: 144] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Mutations in Kv7.2 (KCNQ2) and Kv7.3 (KCNQ3) genes, encoding for voltage-gated K(+) channel subunits underlying the neuronal M-current, have been associated with a wide spectrum of early-onset epileptic disorders ranging from benign familial neonatal seizures to severe epileptic encephalopathies. The aim of the present work has been to investigate the molecular mechanisms of channel dysfunction caused by voltage-sensing domain mutations in Kv7.2 (R144Q, R201C, and R201H) or Kv7.3 (R230C) recently found in patients with epileptic encephalopathies and/or intellectual disability. Electrophysiological studies in mammalian cells transfected with human Kv7.2 and/or Kv7.3 cDNAs revealed that each of these four mutations stabilized the activated state of the channel, thereby producing gain-of-function effects, which are opposite to the loss-of-function effects produced by previously found mutations. Multistate structural modeling revealed that the R201 residue in Kv7.2, corresponding to R230 in Kv7.3, stabilized the resting and nearby voltage-sensing domain states by forming an intricate network of electrostatic interactions with neighboring negatively charged residues, a result also confirmed by disulfide trapping experiments. Using a realistic model of a feedforward inhibitory microcircuit in the hippocampal CA1 region, an increased excitability of pyramidal neurons was found upon incorporation of the experimentally defined parameters for mutant M-current, suggesting that changes in network interactions rather than in intrinsic cell properties may be responsible for the neuronal hyperexcitability by these gain-of-function mutations. Together, the present results suggest that gain-of-function mutations in Kv7.2/3 currents may cause human epilepsy with a severe clinical course, thus revealing a previously unexplored level of complexity in disease pathogenetic mechanisms.
Collapse
|
41
|
Hönigsperger C, Marosi M, Murphy R, Storm JF. Dorsoventral differences in Kv7/M-current and its impact on resonance, temporal summation and excitability in rat hippocampal pyramidal cells. J Physiol 2015; 593:1551-80. [PMID: 25656084 PMCID: PMC4386960 DOI: 10.1113/jphysiol.2014.280826] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Accepted: 12/08/2014] [Indexed: 12/15/2022] Open
Abstract
In rodent hippocampi, the connections, gene expression and functions differ along the dorsoventral (D-V) axis. CA1 pyramidal cells show increasing excitability along the D-V axis, although the underlying mechanism is not known. In the present study, we investigated how the M-current (IM ), caused by Kv7/M (KCNQ) potassium channels, and known to often control neuronal excitability, contributes to D-V differences in intrinsic properties of CA1 pyramidal cells. Using whole-cell patch clamp recordings and the selective Kv7/M blocker 10,10-bis(4-pyridinylmethyl)-9(10H)-anthracenone dihydrochloride (XE991) in hippocampal slices from 3- to 4-week-old rats, we found that: (i) IM had a stronger impact on subthreshold electrical properties in dorsal than ventral CA1 pyramidal cells, including input resistance, temporal summation of artificial synaptic potentials, and M-resonance; (ii) IM activated at more negative potentials (left-shifted) and had larger peak amplitude in the dorsal than ventral CA1; and (iii) the initial spike threshold (during ramp depolarizations) was elevated, and the medium after-hyperpolarization and spike frequency adaptation were increased (i.e. excitability was lower) in the dorsal rather than ventral CA1. These differences were abolished or reduced by application of XE991, indicating that they were caused by IM . Thus, it appears that IM has stronger effects in dorsal than in ventral rat CA1 pyramidal cells because of a larger maximal M-conductance and left-shifted activation curve in the dorsal cells. These mechanisms may contribute to D-V differences in the rate and phase coding of position by CA1 place cells, and may also enhance epileptiform activity in ventral CA1.
Collapse
Affiliation(s)
- Christoph Hönigsperger
- Department of Physiology, Institute of Basic Medical Sciences, University of OsloOslo, Norway
| | - Máté Marosi
- Department of Physiology, Institute of Basic Medical Sciences, University of OsloOslo, Norway
| | - Ricardo Murphy
- Department of Physiology, Institute of Basic Medical Sciences, University of OsloOslo, Norway
| | - Johan F Storm
- Department of Physiology, Institute of Basic Medical Sciences, University of OsloOslo, Norway
| |
Collapse
|
42
|
Caminos E, Vaquero CF, Martinez-Galan JR. Relationship between rat retinal degeneration and potassium channel KCNQ5 expression. Exp Eye Res 2014; 131:1-11. [PMID: 25499209 DOI: 10.1016/j.exer.2014.12.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Revised: 12/02/2014] [Accepted: 12/09/2014] [Indexed: 11/29/2022]
Abstract
KCNQ5/Kv7.5 is a low-threshold non-inactivating voltage-gated potassium channel preferentially targeted to excitatory endings in brain neurons. The M-type current is mediated by KCNQ5 channel subunits in monkey retinal pigment epithelium cells and in brain neurons. This study was undertaken to analyze KCNQ5 expression and the interaction signals of KCNQ5 with other proteins in normal rat retina and during photoreceptor degeneration. The KCNQ5 expression pattern was studied by immunocytochemistry and Western blot in normal rat retinas (Sprague-Dawley, SD) and P23H-1 rats as a retinitis pigmentosa model. The physical interactions of KCNQ5 with calmodulin (CaM), vesicular glutamate transporter 1 (VGluT1) and glial fibrillary acidic protein (GFAP) were analyzed by in situ proximity ligation assays and were supported by calcium recording. KCNQ5 expression was found in the plexiform layers, ganglion cell layer and basal membrane of the retinal pigment epithelium. The physical interactions among KCNQ5 and CaM, VGluT1 and GFAP changed with age and during retinal degeneration. The maximal level of KCNQ5/CaM interaction was found when photoreceptors had almost completely disappeared; the KCNQ5/VGluT1 interaction signal decreased and the KCNQ5/GFAP interaction increased in the inner retina, while degeneration progressed. The basal calcium levels in the astrocytes and neurons of P23H-1 were higher than in the control SD retinas. This study demonstrates that KCNQ5 is present in the rat retina where its activity may be moderated by CaM. Retinal degeneration progression in P23H-1 rats can be followed by an interaction between KCNQ5 with CaM in an in situ system. The relationship between KCNQ5 and VGluT1 or GFAP needs to be more cautiously interpreted.
Collapse
Affiliation(s)
- Elena Caminos
- School of Medicine and Institute for Research in Neurological Disabilities (IDINE), University of Castilla-La Mancha, Albacete, Spain.
| | - Cecilia F Vaquero
- School of Medicine and Regional Center for Biomedical Research (CRIB), University of Castilla-La Mancha, Albacete, Spain.
| | - Juan R Martinez-Galan
- School of Medicine and Institute for Research in Neurological Disabilities (IDINE), University of Castilla-La Mancha, Albacete, Spain.
| |
Collapse
|
43
|
Li C, Huang P, Lu Q, Zhou M, Guo L, Xu X. KCNQ/Kv7 channel activator flupirtine protects against acute stress-induced impairments of spatial memory retrieval and hippocampal LTP in rats. Neuroscience 2014; 280:19-30. [PMID: 25234320 DOI: 10.1016/j.neuroscience.2014.09.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Revised: 09/03/2014] [Accepted: 09/05/2014] [Indexed: 12/17/2022]
Abstract
Spatial memory retrieval and hippocampal long-term potentiation (LTP) are impaired by stress. KCNQ/Kv7 channels are closely associated with memory and the KCNQ/Kv7 channel activator flupirtine represents neuroprotective effects. This study aims to test whether KCNQ/Kv7 channel activation prevents acute stress-induced impairments of spatial memory retrieval and hippocampal LTP. Rats were placed on an elevated platform in the middle of a bright room for 30 min to evoke acute stress. The expression of KCNQ/Kv7 subunits was analyzed at 1, 3 and 12 h after stress by Western blotting. Spatial memory was examined by the Morris water maze (MWM) and the field excitatory postsynaptic potential (fEPSP) in the hippocampal CA1 area was recorded in vivo. Acute stress transiently decreased the expression of KCNQ2 and KCNQ3 in the hippocampus. Acute stress impaired the spatial memory retrieval and hippocampal LTP, the KCNQ/Kv7 channel activator flupirtine prevented the impairments, and the protective effects of flupirtine were blocked by XE-991 (10,10-bis(4-Pyridinylmethyl)-9(10H)-anthracenone), a selective KCNQ channel blocker. Furthermore, acute stress decreased the phosphorylation of glycogen synthase kinase-3β (GSK-3β) at Ser9 in the hippocampus, and flupirtine inhibited the reduction. These results suggest that the KCNQ/Kv7 channels may be a potential target for protecting both hippocampal synaptic plasticity and spatial memory retrieval from acute stress influences.
Collapse
Affiliation(s)
- C Li
- Department of Pharmacology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - P Huang
- Department of Pharmacology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Q Lu
- Department of Pharmacology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan 430030, China
| | - M Zhou
- Department of Pharmacology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - L Guo
- Department of Pharmacology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan 430030, China
| | - X Xu
- Department of Pharmacology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan 430030, China.
| |
Collapse
|
44
|
Anderson UA, Carson C, Johnston L, Joshi S, Gurney AM, McCloskey KD. Functional expression of KCNQ (Kv7) channels in guinea pig bladder smooth muscle and their contribution to spontaneous activity. Br J Pharmacol 2014; 169:1290-304. [PMID: 23586426 PMCID: PMC3746117 DOI: 10.1111/bph.12210] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2012] [Revised: 03/15/2013] [Accepted: 03/26/2013] [Indexed: 12/30/2022] Open
Abstract
Background and Purpose The aim of the study was to determine whether KCNQ channels are functionally expressed in bladder smooth muscle cells (SMC) and to investigate their physiological significance in bladder contractility. Experimental Approach KCNQ channels were examined at the genetic, protein, cellular and tissue level in guinea pig bladder smooth muscle using RT-PCR, immunofluorescence, patch-clamp electrophysiology, calcium imaging, detrusor strip myography, and a panel of KCNQ activators and inhibitors. Key Results KCNQ subtypes 1–5 are expressed in bladder detrusor smooth muscle. Detrusor strips typically displayed TTX-insensitive myogenic spontaneous contractions that were increased in amplitude by the KCNQ channel inhibitors XE991, linopirdine or chromanol 293B. Contractility was inhibited by the KCNQ channel activators flupirtine or meclofenamic acid (MFA). The frequency of Ca2+-oscillations in SMC contained within bladder tissue sheets was increased by XE991. Outward currents in dispersed bladder SMC, recorded under conditions where BK and KATP currents were minimal, were significantly reduced by XE991, linopirdine, or chromanol, and enhanced by flupirtine or MFA. XE991 depolarized the cell membrane and could evoke transient depolarizations in quiescent cells. Flupirtine (20 μM) hyperpolarized the cell membrane with a simultaneous cessation of any spontaneous electrical activity. Conclusions and Implications These novel findings reveal the role of KCNQ currents in the regulation of the resting membrane potential of detrusor SMC and their important physiological function in the control of spontaneous contractility in the guinea pig bladder.
Collapse
Affiliation(s)
- U A Anderson
- Centre for Cancer Research and Cell Biology, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, UK
| | | | | | | | | | | |
Collapse
|
45
|
Conditional deletions of epilepsy-associated KCNQ2 and KCNQ3 channels from cerebral cortex cause differential effects on neuronal excitability. J Neurosci 2014; 34:5311-21. [PMID: 24719109 DOI: 10.1523/jneurosci.3919-13.2014] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
KCNQ2 and KCNQ3 potassium channels have emerged as central regulators of pyramidal neuron excitability and spiking behavior. However, despite an abundance of evidence demonstrating that KCNQ2/3 heteromers underlie critical potassium conductances, it is unknown whether KCNQ2, KCNQ3, or both are obligatory for maintaining normal pyramidal neuron excitability. Here, we demonstrate that conditional deletion of Kcnq2 from cerebral cortical pyramidal neurons in mice results in abnormal electrocorticogram activity and early death, whereas similar deletion of Kcnq3 does not. At the cellular level, Kcnq2-null, but not Kcnq3-null, CA1 pyramidal neurons show increased excitability manifested as a decreased medium afterhyperpolarization and a longer-lasting afterdepolarization. As a result, these Kcnq2-deficient neurons are hyperexcitable, responding to current injections with an increased number and frequency of action potentials. Biochemically, the Kcnq2 deficiency secondarily results in a substantial loss of KCNQ3 and KCNQ5 protein levels, whereas loss of Kcnq3 only leads to a modest reduction of other KCNQ channels. Consistent with this finding, KCNQ allosteric activators can still markedly dampen neuronal excitability in Kcnq3-null pyramidal neurons, but have only weak effects in Kcnq2-null pyramidal neurons. Together, our data reveal the indispensable function of KCNQ2 channels at both the cellular and systems levels, and demonstrate that pyramidal neurons have near normal excitability in the absence of KCNQ3 channels.
Collapse
|
46
|
Battefeld A, Tran BT, Gavrilis J, Cooper EC, Kole MHP. Heteromeric Kv7.2/7.3 channels differentially regulate action potential initiation and conduction in neocortical myelinated axons. J Neurosci 2014; 34:3719-32. [PMID: 24599470 PMCID: PMC3942587 DOI: 10.1523/jneurosci.4206-13.2014] [Citation(s) in RCA: 130] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Revised: 01/27/2014] [Accepted: 01/30/2014] [Indexed: 12/14/2022] Open
Abstract
Rapid energy-efficient signaling along vertebrate axons is achieved through intricate subcellular arrangements of voltage-gated ion channels and myelination. One recently appreciated example is the tight colocalization of K(v)7 potassium channels and voltage-gated sodium (Na(v)) channels in the axonal initial segment and nodes of Ranvier. The local biophysical properties of these K(v)7 channels and the functional impact of colocalization with Na(v) channels remain poorly understood. Here, we quantitatively examined K(v)7 channels in myelinated axons of rat neocortical pyramidal neurons using high-resolution confocal imaging and patch-clamp recording. K(v)7.2 and 7.3 immunoreactivity steeply increased within the distal two-thirds of the axon initial segment and was mirrored by the conductance density estimates, which increased from ~12 (proximal) to 150 pS μm(-2) (distal). The axonal initial segment and nodal M-currents were similar in voltage dependence and kinetics, carried by K(v)7.2/7.3 heterotetramers, 4% activated at the resting membrane potential and rapidly activated with single-exponential time constants (~15 ms at 28 mV). Experiments and computational modeling showed that while somatodendritic K(v)7 channels are strongly activated by the backpropagating action potential to attenuate the afterdepolarization and repetitive firing, axonal K(v)7 channels are minimally recruited by the forward-propagating action potential. Instead, in nodal domains K(v)7.2/7.3 channels were found to increase Na(v) channel availability and action potential amplitude by stabilizing the resting membrane potential. Thus, K(v)7 clustering near axonal Na(v) channels serves specific and context-dependent roles, both restraining initiation and enhancing conduction of the action potential.
Collapse
Affiliation(s)
- Arne Battefeld
- Netherlands Institute for Neuroscience, Royal Academy of Arts and Sciences, 1105 BA, Amsterdam, The Netherlands
| | - Baouyen T. Tran
- Baylor College of Medicine, Baylor Plaza, Houston, Texas 77030
| | - Jason Gavrilis
- Eccles Institute for Neuroscience, The Australian National University, Canberra 0200, Australian Capital Territory, Australia, and
- Department of Audiology and Speech Pathology, University of Melbourne, Melbourne, Victoria 3010, Australia
| | | | - Maarten H. P. Kole
- Netherlands Institute for Neuroscience, Royal Academy of Arts and Sciences, 1105 BA, Amsterdam, The Netherlands
- Eccles Institute for Neuroscience, The Australian National University, Canberra 0200, Australian Capital Territory, Australia, and
| |
Collapse
|
47
|
A unified approach to linking experimental, statistical and computational analysis of spike train data. PLoS One 2014; 9:e85269. [PMID: 24465520 PMCID: PMC3894976 DOI: 10.1371/journal.pone.0085269] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Accepted: 11/24/2013] [Indexed: 11/24/2022] Open
Abstract
A fundamental issue in neuroscience is how to identify the multiple biophysical mechanisms through which neurons generate observed patterns of spiking activity. In previous work, we proposed a method for linking observed patterns of spiking activity to specific biophysical mechanisms based on a state space modeling framework and a sequential Monte Carlo, or particle filter, estimation algorithm. We have shown, in simulation, that this approach is able to identify a space of simple biophysical models that were consistent with observed spiking data (and included the model that generated the data), but have yet to demonstrate the application of the method to identify realistic currents from real spike train data. Here, we apply the particle filter to spiking data recorded from rat layer V cortical neurons, and correctly identify the dynamics of an slow, intrinsic current. The underlying intrinsic current is successfully identified in four distinct neurons, even though the cells exhibit two distinct classes of spiking activity: regular spiking and bursting. This approach – linking statistical, computational, and experimental neuroscience – provides an effective technique to constrain detailed biophysical models to specific mechanisms consistent with observed spike train data.
Collapse
|
48
|
Le Duigou C, Simonnet J, Teleñczuk MT, Fricker D, Miles R. Recurrent synapses and circuits in the CA3 region of the hippocampus: an associative network. Front Cell Neurosci 2014; 7:262. [PMID: 24409118 PMCID: PMC3884140 DOI: 10.3389/fncel.2013.00262] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2013] [Accepted: 12/02/2013] [Indexed: 11/29/2022] Open
Abstract
In the CA3 region of the hippocampus, pyramidal cells excite other pyramidal cells and interneurons. The axons of CA3 pyramidal cells spread throughout most of the region to form an associative network. These connections were first drawn by Cajal and Lorente de No. Their physiological properties were explored to understand epileptiform discharges generated in the region. Synapses between pairs of pyramidal cells involve one or few release sites and are weaker than connections made by mossy fibers on CA3 pyramidal cells. Synapses with interneurons are rather effective, as needed to control unchecked excitation. We examine contributions of recurrent synapses to epileptiform synchrony, to the genesis of sharp waves in the CA3 region and to population oscillations at theta and gamma frequencies. Recurrent connections in CA3, as other associative cortices, have a lower connectivity spread over a larger area than in primary sensory cortices. This sparse, but wide-ranging connectivity serves the functions of an associative network, including acquisition of neuronal representations as activity in groups of CA3 cells and completion involving the recall from partial cues of these ensemble firing patterns.
Collapse
Affiliation(s)
- Caroline Le Duigou
- Centre de Recherche de l'Institut du Cerveau et de la Moelle, INSERM U975, CHU Pitié-Salpêtrière, Université Pierre et Marie Curie Paris, France
| | - Jean Simonnet
- Centre de Recherche de l'Institut du Cerveau et de la Moelle, INSERM U975, CHU Pitié-Salpêtrière, Université Pierre et Marie Curie Paris, France
| | - Maria T Teleñczuk
- Centre de Recherche de l'Institut du Cerveau et de la Moelle, INSERM U975, CHU Pitié-Salpêtrière, Université Pierre et Marie Curie Paris, France
| | - Desdemona Fricker
- Centre de Recherche de l'Institut du Cerveau et de la Moelle, INSERM U975, CHU Pitié-Salpêtrière, Université Pierre et Marie Curie Paris, France
| | - Richard Miles
- Centre de Recherche de l'Institut du Cerveau et de la Moelle, INSERM U975, CHU Pitié-Salpêtrière, Université Pierre et Marie Curie Paris, France
| |
Collapse
|
49
|
Mateos-Aparicio P, Murphy R, Storm JF. Complementary functions of SK and Kv7/M potassium channels in excitability control and synaptic integration in rat hippocampal dentate granule cells. J Physiol 2013; 592:669-93. [PMID: 24366266 DOI: 10.1113/jphysiol.2013.267872] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The dentate granule cells (DGCs) form the most numerous neuron population of the hippocampal memory system, and its gateway for cortical input. Yet, we have only limited knowledge of the intrinsic membrane properties that shape their responses. Since SK and Kv7/M potassium channels are key mechanisms of neuronal spiking and excitability control, afterhyperpolarizations (AHPs) and synaptic integration, we studied their functions in DGCs. The specific SK channel blockers apamin or scyllatoxin increased spike frequency (excitability), reduced early spike frequency adaptation, fully blocked the medium-duration AHP (mAHP) after a single spike or spike train, and increased postsynaptic EPSP summation after spiking, but had no effect on input resistance (Rinput) or spike threshold. In contrast, blockade of Kv7/M channels by XE991 increased Rinput, lowered the spike threshold, and increased excitability, postsynaptic EPSP summation, and EPSP-spike coupling, but only slightly reduced mAHP after spike trains (and not after single spikes). The SK and Kv7/M channel openers 1-EBIO and retigabine, respectively, had effects opposite to the blockers. Computational modelling reproduced many of these effects. We conclude that SK and Kv7/M channels have complementary roles in DGCs. These mechanisms may be important for the dentate network function, as CA3 neurons can be activated or inhibition recruited depending on DGC firing rate.
Collapse
Affiliation(s)
- Pedro Mateos-Aparicio
- Department of Physiology, IMB, University of Oslo, PB 1104 Blindern, 0317 Oslo, Norway.
| | | | | |
Collapse
|
50
|
Shah NH, Aizenman E. Voltage-gated potassium channels at the crossroads of neuronal function, ischemic tolerance, and neurodegeneration. Transl Stroke Res 2013; 5:38-58. [PMID: 24323720 DOI: 10.1007/s12975-013-0297-7] [Citation(s) in RCA: 116] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Revised: 09/14/2013] [Accepted: 10/14/2013] [Indexed: 11/29/2022]
Abstract
Voltage-gated potassium (Kv) channels are widely expressed in the central and peripheral nervous system and are crucial mediators of neuronal excitability. Importantly, these channels also actively participate in cellular and molecular signaling pathways that regulate the life and death of neurons. Injury-mediated increased K(+) efflux through Kv2.1 channels promotes neuronal apoptosis, contributing to widespread neuronal loss in neurodegenerative disorders such as Alzheimer's disease and stroke. In contrast, some forms of neuronal activity can dramatically alter Kv2.1 channel phosphorylation levels and influence their localization. These changes are normally accompanied by modifications in channel voltage dependence, which may be neuroprotective within the context of ischemic injury. Kv1 and Kv7 channel dysfunction leads to neuronal hyperexcitability that critically contributes to the pathophysiology of human clinical disorders such as episodic ataxia and epilepsy. This review summarizes the neurotoxic, neuroprotective, and neuroregulatory roles of Kv channels and highlights the consequences of Kv channel dysfunction on neuronal physiology. The studies described in this review thus underscore the importance of normal Kv channel function in neurons and emphasize the therapeutic potential of targeting Kv channels in the treatment of a wide range of neurological diseases.
Collapse
Affiliation(s)
- Niyathi Hegde Shah
- Department of Neurobiology, University of Pittsburgh School of Medicine, 3500 Terrace Street, E1456 BST, Pittsburgh, PA, 15261, USA,
| | | |
Collapse
|