1
|
Incontro S, Musella ML, Sammari M, Di Scala C, Fantini J, Debanne D. Lipids shape brain function through ion channel and receptor modulations: physiological mechanisms and clinical perspectives. Physiol Rev 2025; 105:137-207. [PMID: 38990068 DOI: 10.1152/physrev.00004.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 05/28/2024] [Accepted: 07/01/2024] [Indexed: 07/12/2024] Open
Abstract
Lipids represent the most abundant molecular type in the brain, with a fat content of ∼60% of the dry brain weight in humans. Despite this fact, little attention has been paid to circumscribe the dynamic role of lipids in brain function and disease. Membrane lipids such as cholesterol, phosphoinositide, sphingolipids, arachidonic acid, and endocannabinoids finely regulate both synaptic receptors and ion channels that ensure critical neural functions. After a brief introduction on brain lipids and their respective properties, we review here their role in regulating synaptic function and ion channel activity, action potential propagation, neuronal development, and functional plasticity and their contribution in the development of neurological and neuropsychiatric diseases. We also provide possible directions for future research on lipid function in brain plasticity and diseases.
Collapse
Affiliation(s)
| | | | - Malika Sammari
- UNIS, INSERM, Aix-Marseille Université, Marseille, France
| | | | | | | |
Collapse
|
2
|
Debanne D, Mylonaki K, Musella ML, Russier M. Voltage-gated ion channels in epilepsies: circuit dysfunctions and treatments. Trends Pharmacol Sci 2024; 45:1018-1032. [PMID: 39406591 DOI: 10.1016/j.tips.2024.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 09/16/2024] [Accepted: 09/19/2024] [Indexed: 11/10/2024]
Abstract
Epileptic encephalopathies are generally considered to be functional disruptions in the balance between neural excitation and inhibition. Excitatory and inhibitory voltage-gated ion channels are key targets of antiepileptic drugs, playing a critical role in regulating neuronal excitation and synaptic transmission. Recent research has highlighted the significance of ion channels in various aspects of epilepsy, including presynaptic neurotransmitter release, intrinsic neuronal excitability, and neural synchrony. Genetic alterations in excitatory and inhibitory ion channels within principal neurons and in inhibitory interneurons have also been identified as key contributors to the development of epilepsies. We review these recent studies and shed light on the bidirectional relationship between epilepsy and neuronal excitability and the latest advancements in pharmacological therapeutics targeting ion channels for epilepsy treatment.
Collapse
|
3
|
Xie C, Kessi M, Yin F, Peng J. Roles of KCNA2 in Neurological Diseases: from Physiology to Pathology. Mol Neurobiol 2024; 61:8491-8517. [PMID: 38517617 DOI: 10.1007/s12035-024-04120-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 03/10/2024] [Indexed: 03/24/2024]
Abstract
Potassium voltage-gated channel subfamily a member 2 (Kv1.2, encoded by KCNA2) is highly expressed in the central and peripheral nervous systems. Based on the patch clamp studies, gain-of function (GOF), loss-of-function (LOF), and a mixed type (GOF/LOF) variants can cause different conditions/disorders. KCNA2-related neurological diseases include epilepsy, intellectual disability (ID), attention deficit/hyperactive disorder (ADHD), autism spectrum disorder (ASD), pain as well as autoimmune and movement disorders. Currently, the molecular mechanisms for the reported variants in causing diverse disorders are unknown. Consequently, this review brings up to date the related information regarding the structure and function of Kv1.2 channel, expression patterns, neuronal localizations, and tetramerization as well as important cell and animal models. In addition, it provides updates on human genetic variants, genotype-phenotype correlations especially highlighting the deep insight into clinical prognosis of KCNA2-related developmental and epileptic encephalopathy, mechanisms, and the potential treatment targets for all KCNA2-related neurological disorders.
Collapse
Affiliation(s)
- Changning Xie
- Department of Pediatrics, Xiangya Hospital, Central South University, Xiangya Road 87, Hunan, Changsha, 410008, China
| | - Miriam Kessi
- Department of Pediatrics, Xiangya Hospital, Central South University, Xiangya Road 87, Hunan, Changsha, 410008, China
| | - Fei Yin
- Department of Pediatrics, Xiangya Hospital, Central South University, Xiangya Road 87, Hunan, Changsha, 410008, China
| | - Jing Peng
- Department of Pediatrics, Xiangya Hospital, Central South University, Xiangya Road 87, Hunan, Changsha, 410008, China.
- Hunan Intellectual and Development Disabilities Research Center, Hunan, Changsha, 410008, China.
| |
Collapse
|
4
|
Wen W, Turrigiano GG. Keeping Your Brain in Balance: Homeostatic Regulation of Network Function. Annu Rev Neurosci 2024; 47:41-61. [PMID: 38382543 DOI: 10.1146/annurev-neuro-092523-110001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2024]
Abstract
To perform computations with the efficiency necessary for animal survival, neocortical microcircuits must be capable of reconfiguring in response to experience, while carefully regulating excitatory and inhibitory connectivity to maintain stable function. This dynamic fine-tuning is accomplished through a rich array of cellular homeostatic plasticity mechanisms that stabilize important cellular and network features such as firing rates, information flow, and sensory tuning properties. Further, these functional network properties can be stabilized by different forms of homeostatic plasticity, including mechanisms that target excitatory or inhibitory synapses, or that regulate intrinsic neuronal excitability. Here we discuss which aspects of neocortical circuit function are under homeostatic control, how this homeostasis is realized on the cellular and molecular levels, and the pathological consequences when circuit homeostasis is impaired. A remaining challenge is to elucidate how these diverse homeostatic mechanisms cooperate within complex circuits to enable them to be both flexible and stable.
Collapse
Affiliation(s)
- Wei Wen
- Department of Biology, Brandeis University, Waltham, Massachusetts, USA;
| | - Gina G Turrigiano
- Department of Biology, Brandeis University, Waltham, Massachusetts, USA;
| |
Collapse
|
5
|
Ramirez-Franco J, Debreux K, Sangiardi M, Belghazi M, Kim Y, Lee SH, Lévêque C, Seagar M, El Far O. The downregulation of Kv 1 channels in Lgi1 -/-mice is accompanied by a profound modification of its interactome and a parallel decrease in Kv 2 channels. Neurobiol Dis 2024; 196:106513. [PMID: 38663634 DOI: 10.1016/j.nbd.2024.106513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 03/12/2024] [Accepted: 04/23/2024] [Indexed: 05/03/2024] Open
Abstract
In animal models of LGI1-dependent autosomal dominant lateral temporal lobe epilepsy, Kv1 channels are downregulated, suggesting their crucial involvement in epileptogenesis. The molecular basis of Kv1 channel-downregulation in LGI1 knock-out mice has not been elucidated and how the absence of this extracellular protein induces an important modification in the expression of Kv1 remains unknown. In this study we analyse by immunofluorescence the modifications in neuronal Kv1.1 and Kv1.2 distribution throughout the hippocampal formation of LGI1 knock-out mice. We show that Kv1 downregulation is not restricted to the axonal compartment, but also takes place in the somatodendritic region and is accompanied by a drastic decrease in Kv2 expression levels. Moreover, we find that the downregulation of these Kv channels is associated with a marked increase in bursting patterns. Finally, mass spectrometry uncovered key modifications in the Kv1 interactome that highlight the epileptogenic implication of Kv1 downregulation in LGI1 knock-out animals.
Collapse
Affiliation(s)
- Jorge Ramirez-Franco
- INSERM UMR_S 1072, Unité de Neurobiologie des canaux Ioniques et de la Synapse, Aix-Marseille Université, 13015 Marseille, France.
| | - Kévin Debreux
- INSERM UMR_S 1072, Unité de Neurobiologie des canaux Ioniques et de la Synapse, Aix-Marseille Université, 13015 Marseille, France
| | - Marion Sangiardi
- INSERM UMR_S 1072, Unité de Neurobiologie des canaux Ioniques et de la Synapse, Aix-Marseille Université, 13015 Marseille, France
| | - Maya Belghazi
- Marseille Protéomique (MaP), Plateforme Protéomique IMM, CNRS FR3479, Aix-Marseille Université, 31 Chemin Joseph Aiguier, 13009 Marseille, France
| | - Yujin Kim
- Department of Physiology, Cell Physiology Lab, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul 03080, South Korea
| | - Suk-Ho Lee
- Department of Physiology, Cell Physiology Lab, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul 03080, South Korea
| | - Christian Lévêque
- INSERM UMR_S 1072, Unité de Neurobiologie des canaux Ioniques et de la Synapse, Aix-Marseille Université, 13015 Marseille, France
| | - Michael Seagar
- INSERM UMR_S 1072, Unité de Neurobiologie des canaux Ioniques et de la Synapse, Aix-Marseille Université, 13015 Marseille, France
| | - Oussama El Far
- INSERM UMR_S 1072, Unité de Neurobiologie des canaux Ioniques et de la Synapse, Aix-Marseille Université, 13015 Marseille, France.
| |
Collapse
|
6
|
Ren L, Yu J, Chen H, Luo J, Lv F, Min S. Alteration of hyperpolarization-activated cation current-mediated metaplasticity contributes to electroconvulsive shock-induced learning and memory impairment in depressed rats. Front Psychiatry 2024; 15:1365119. [PMID: 38911706 PMCID: PMC11190359 DOI: 10.3389/fpsyt.2024.1365119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 05/27/2024] [Indexed: 06/25/2024] Open
Abstract
Background Accompanied by a rapid and effective antidepressant effect, electroconvulsive shock (ECS) can also induce learning and memory impairment. Our previous research reported that metaplasticity is involved in this process. However, the mechanisms still remain unclear. This study investigated the role of I h current in the metaplastic changes and learning and memory impairment induced by ECS in depressive rats. Methods Depressive rats received ECS after modelling using chronic unpredictable. ZD7288, a type of I h current inhibitor was used to verify the effect of I h current. The sucrose preference test and Morris water maze were used for behavior testing. Changes in metaplasticity was assessed with the LTD/LTP threshold by stimulation at different frequencies. Spontaneous and evoked action potentials (APs) were measured to confirm difference of neuronal excitability. Additionally, the amplitude of I h current was analyzed. Results ECS exerts antidepressant effect, but also induce spatial learning and memory dysfunction. ECS up-regulates the LTD/LTP threshold. In rats treated with ECS, the frequency of spontaneous and evoked APs is significantly reduced. In addition, ECS induces changes in the intrinsic properties of AP, including a decrease of AP-half width and peak amplitude, and an increase in AP time to peak and post-hyperpolarization potential amplitude. In particular, ECS increases both instantaneous and steady-state I h currents. However, Inhibition of I h current with ZD7288 results in a relief of learning and memory impairment and a decrease in threshold, as well as a significant reversal of whole-cell electrophysiological changes. Conclusion ECS-induced learning and memory impairment is caused by neuronal hypoexcitability mediated metaplasticity, and upregulation of LTD/LTP threshold by an increase in I h current.
Collapse
Affiliation(s)
- Li Ren
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jian Yu
- Department of Psychiatry, Shanghai 10th People’s Hospital, Anesthesia and Brain Research Institute, Tongji University, Shanghai, China
| | - Hengsheng Chen
- Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Jie Luo
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Feng Lv
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Su Min
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
7
|
Guarina L, Le JT, Griffith TN, Santana LF, Cudmore RH. SanPy: Software for the analysis and visualization of whole-cell current-clamp recordings. Biophys J 2024; 123:759-769. [PMID: 38419330 PMCID: PMC10995421 DOI: 10.1016/j.bpj.2024.02.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 01/16/2024] [Accepted: 02/26/2024] [Indexed: 03/02/2024] Open
Abstract
The analysis of action potentials and other membrane voltage fluctuations provides a powerful approach for interrogating the function of excitable cells. However, a major bottleneck in the interpretation of this critical data is the lack of intuitive, agreed-upon software tools for its analysis. Here, we present SanPy, an open-source and freely available software package for the analysis and exploration of whole-cell current-clamp recordings written in Python. SanPy provides a robust computational engine with an application programming interface. Using this, we have developed a cross-platform desktop application with a graphical user interface that does not require programming. SanPy is designed to extract common parameters from action potentials, including threshold time and voltage, peak, half-width, and interval statistics. In addition, several cardiac parameters are measured, including the early diastolic duration and rate. SanPy is built to be fully extensible by providing a plugin architecture for the addition of new file loaders, analysis, and visualizations. A key feature of SanPy is its focus on quality control and data exploration. In the desktop interface, all plots of the data and analysis are linked, allowing simultaneous data visualization from different dimensions with the goal of obtaining ground-truth analysis. We provide documentation for all aspects of SanPy, including several use cases and examples. To test SanPy, we performed analysis on current-clamp recordings from heart and brain cells. Taken together, SanPy is a powerful tool for whole-cell current-clamp analysis and lays the foundation for future extension by the scientific community.
Collapse
Affiliation(s)
- Laura Guarina
- Department of Physiology & Membrane Biology, University of California-Davis School of Medicine, Davis, California
| | - Johnson Tran Le
- Department of Physiology & Membrane Biology, University of California-Davis School of Medicine, Davis, California
| | - Theanne N Griffith
- Department of Physiology & Membrane Biology, University of California-Davis School of Medicine, Davis, California
| | - Luis Fernando Santana
- Department of Physiology & Membrane Biology, University of California-Davis School of Medicine, Davis, California
| | - Robert H Cudmore
- Department of Physiology & Membrane Biology, University of California-Davis School of Medicine, Davis, California.
| |
Collapse
|
8
|
Naderi S, Motamedi F, Pourbadie HG, Rafiei S, Khodagholi F, Naderi N, Janahmadi M. Neuroprotective Effects of Ferrostatin and Necrostatin Against Entorhinal Amyloidopathy-Induced Electrophysiological Alterations Mediated by voltage-gated Ca 2+ Channels in the Dentate Gyrus Granular Cells. Neurochem Res 2024; 49:99-116. [PMID: 37615884 DOI: 10.1007/s11064-023-04006-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Revised: 06/07/2023] [Accepted: 07/29/2023] [Indexed: 08/25/2023]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease that is the main form of dementia. Abnormal deposition of amyloid-beta (Aβ) peptides in neurons and synapses cause neuronal loss and cognitive deficits. We have previously reported that ferroptosis and necroptosis were implicated in Aβ25-35 neurotoxicity, and their specific inhibitors had attenuating effects on cognitive impairment induced by Aβ25-35 neurotoxicity. Here, we aimed to examine the impact of ferroptosis and necroptosis inhibition following the Aβ25-35 neurotoxicity on the neuronal excitability of dentate gyrus (DG) and the possible involvement of voltage-gated Ca2+ channels in their effects. After inducing Aβ25-35 neurotoxicity, electrophysiological alterations in the intrinsic properties and excitability were recorded by the whole-cell patch-clamp under current-clamp condition. Voltage-clamp recordings were also performed to shed light on the involvement of calcium channel currents. Aβ25-35 neurotoxicity induced a considerable reduction in input resistance (Rin), accompanied by a profoundly decreased excitability and a reduction in the amplitude of voltage-gated calcium channel currents in the DG granule cells. However, three days of administration of either ferrostatin-1 (Fer-1), a ferroptosis inhibitor, or Necrostatin-1 (Nec-1), a necroptosis inhibitor, in the entorhinal cortex could almost preserve the normal excitability and the Ca2+ currents. In conclusion, these findings suggest that ferroptosis and necroptosis involvement in EC amyloidopathy could be a potential candidate to prevent the suppressive effect of Aβ on the Ca2+ channel current and neuronal function, which might take place in neurons during the development of AD.
Collapse
Affiliation(s)
- Soudabeh Naderi
- School of Medicine, Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fereshteh Motamedi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Shahrbanoo Rafiei
- Neurobiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fariba Khodagholi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nima Naderi
- Department of Pharmacology and Toxicology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahyar Janahmadi
- Neuroscience Research Center, Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
9
|
Extrémet J, Ramirez-Franco J, Fronzaroli-Molinieres L, Boumedine-Guignon N, Ankri N, El Far O, Garrido JJ, Debanne D, Russier M. Rescue of Normal Excitability in LGI1-Deficient Epileptic Neurons. J Neurosci 2023; 43:8596-8606. [PMID: 37863654 PMCID: PMC10727174 DOI: 10.1523/jneurosci.0701-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 09/08/2023] [Accepted: 10/14/2023] [Indexed: 10/22/2023] Open
Abstract
Leucine-rich glioma inactivated 1 (LGI1) is a glycoprotein secreted by neurons, the deletion of which leads to autosomal dominant lateral temporal lobe epilepsy. We previously showed that LGI1 deficiency in a mouse model (i.e., knock-out for LGI1 or KO-Lgi1) decreased Kv1.1 channel density at the axon initial segment (AIS) and at presynaptic terminals, thus enhancing both intrinsic excitability and glutamate release. However, it is not known whether normal excitability can be restored in epileptic neurons. Here, we show that the selective expression of LGI1 in KO-Lgi1 neurons from mice of both sexes, using single-cell electroporation, reduces intrinsic excitability and restores both the Kv1.1-mediated D-type current and Kv1.1 channels at the AIS. In addition, we show that the homeostatic-like shortening of the AIS length observed in KO-Lgi1 neurons is prevented in neurons electroporated with the Lgi1 gene. Furthermore, we reveal a spatial gradient of intrinsic excitability that is centered on the electroporated neuron. We conclude that expression of LGI1 restores normal excitability through functional Kv1 channels at the AIS.SIGNIFICANCE STATEMENT The lack of leucine-rich glioma inactivated 1 (LGI1) protein induces severe epileptic seizures that leads to death. Enhanced intrinsic and synaptic excitation in KO-Lgi1 mice is because of the decrease in Kv1.1 channels in CA3 neurons. However, the conditions to restore normal excitability profile in epileptic neurons remain to be defined. We show here that the expression of LGI1 in KO-Lgi1 neurons in single neurons reduces intrinsic excitability, and restores both the Kv1.1-mediated D-type current and Kv1.1 channels at the axon initial segment (AIS). Furthermore, the homeostatic shortening of the AIS length observed in KO-Lgi1 neurons is prevented in neurons in which the Lgi1 gene has been rescued. We conclude that LGI1 constitutes a critical factor to restore normal excitability in epileptic neurons.
Collapse
Affiliation(s)
- Johanna Extrémet
- Unité de Neurobiologie des canaux Ioniques et de la Synapse, Unité Mixte de Recherche 1072, Institut National de la Santé et de la Recherche Médicale, Aix-Marseille Université, Marseille, 13015, France
| | - Jorge Ramirez-Franco
- Unité de Neurobiologie des canaux Ioniques et de la Synapse, Unité Mixte de Recherche 1072, Institut National de la Santé et de la Recherche Médicale, Aix-Marseille Université, Marseille, 13015, France
| | - Laure Fronzaroli-Molinieres
- Unité de Neurobiologie des canaux Ioniques et de la Synapse, Unité Mixte de Recherche 1072, Institut National de la Santé et de la Recherche Médicale, Aix-Marseille Université, Marseille, 13015, France
| | - Norah Boumedine-Guignon
- Unité de Neurobiologie des canaux Ioniques et de la Synapse, Unité Mixte de Recherche 1072, Institut National de la Santé et de la Recherche Médicale, Aix-Marseille Université, Marseille, 13015, France
| | - Norbert Ankri
- Unité de Neurobiologie des canaux Ioniques et de la Synapse, Unité Mixte de Recherche 1072, Institut National de la Santé et de la Recherche Médicale, Aix-Marseille Université, Marseille, 13015, France
| | - Oussama El Far
- Unité de Neurobiologie des canaux Ioniques et de la Synapse, Unité Mixte de Recherche 1072, Institut National de la Santé et de la Recherche Médicale, Aix-Marseille Université, Marseille, 13015, France
| | - Juan José Garrido
- Cajal Institute, Consejo Superior de Investigaciones Cientificas, Madrid, 28002, Spain
| | - Dominique Debanne
- Unité de Neurobiologie des canaux Ioniques et de la Synapse, Unité Mixte de Recherche 1072, Institut National de la Santé et de la Recherche Médicale, Aix-Marseille Université, Marseille, 13015, France
| | - Michaël Russier
- Unité de Neurobiologie des canaux Ioniques et de la Synapse, Unité Mixte de Recherche 1072, Institut National de la Santé et de la Recherche Médicale, Aix-Marseille Université, Marseille, 13015, France
| |
Collapse
|
10
|
Lu Y, Sciaccotta F, Kiely L, Bellanger B, Erisir A, Meliza CD. Rapid, Activity-Dependent Intrinsic Plasticity in the Developing Zebra Finch Auditory Cortex. J Neurosci 2023; 43:6872-6883. [PMID: 37648449 PMCID: PMC10573762 DOI: 10.1523/jneurosci.0354-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 07/14/2023] [Accepted: 08/23/2023] [Indexed: 09/01/2023] Open
Abstract
The acoustic environment an animal experiences early in life shapes the structure and function of its auditory system. This process of experience-dependent development is thought to be primarily orchestrated by potentiation and depression of synapses, but plasticity of intrinsic voltage dynamics may also contribute. Here, we show that in juvenile male and female zebra finches, neurons in a cortical-level auditory area, the caudal mesopallium (CM), can rapidly change their firing dynamics. This plasticity was only observed in birds that were reared in a complex acoustic and social environment, which also caused increased expression of the low-threshold potassium channel Kv1.1 in the plasma membrane and endoplasmic reticulum (ER). Intrinsic plasticity depended on activity, was reversed by blocking low-threshold potassium currents, and was prevented by blocking intracellular calcium signaling. Taken together, these results suggest that Kv1.1 is rapidly mobilized to the plasma membrane by activity-dependent elevation of intracellular calcium. This produces a shift in the excitability and temporal integration of CM neurons that may be permissive for auditory learning in complex acoustic environments during a crucial period for the development of vocal perception and production.SIGNIFICANCE STATEMENT Neurons can change not only the strength of their connections to other neurons, but also how they integrate synaptic currents to produce patterns of action potentials. In contrast to synaptic plasticity, the mechanisms and functional roles of intrinisic plasticity remain poorly understood. We found that neurons in the zebra finch auditory cortex can rapidly shift their spiking dynamics within a few minutes in response to intracellular stimulation. This plasticity involves increased conductance of a low-threshold potassium current associated with the Kv1.1 channel, but it only occurs in birds reared in a rich acoustic environment. Thus, auditory experience regulates a mechanism of neural plasticity that allows neurons to rapidly adapt their firing dynamics to stimulation.
Collapse
Affiliation(s)
| | | | | | | | - Alev Erisir
- Psychology Department
- Neuroscience Graduate Program, University of Virginia, Charlottesville, Virginia 22904
| | - C Daniel Meliza
- Psychology Department
- Neuroscience Graduate Program, University of Virginia, Charlottesville, Virginia 22904
| |
Collapse
|
11
|
Guarina L, Le JT, Griffith TN, Santana LF, Cudmore RH. SanPy: A whole-cell electrophysiology analysis pipeline. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.06.539660. [PMID: 37214972 PMCID: PMC10197560 DOI: 10.1101/2023.05.06.539660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
The analysis of action potentials and other membrane voltage fluctuations provide a powerful approach for interrogating the function of excitable cells. Yet, a major bottleneck in the interpretation of this critical data is the lack of intuitive, agreed upon software tools for its analysis. Here, we present SanPy, a Python-based open-source and freely available software pipeline for the analysis and exploration of whole-cell current-clamp recordings. SanPy provides a robust computational engine with an application programming interface. Using this, we have developed a cross-platform graphical user interface that does not require programming. SanPy is designed to extract common parameters from action potentials including threshold time and voltage, peak, half-width, and interval statistics. In addition, several cardiac parameters are measured including the early diastolic duration and rate. SanPy is built to be fully extensible by providing a plugin architecture for the addition of new file loaders, analysis, and visualizations. A key feature of SanPy is its focus on quality control and data exploration. In the desktop interface, all plots of the data and analysis are linked allowing simultaneous data visualization from different dimensions with the goal of obtaining ground truth analysis. We provide documentation for all aspects of SanPy including several use cases and examples. To test SanPy, we have performed analysis on current-clamp recordings from heart and brain cells. Taken together, SanPy is a powerful tool for whole-cell current-clamp analysis and lays the foundation for future extension by the scientific community.
Collapse
Affiliation(s)
- Laura Guarina
- Department of Physiology & Membrane Biology, University of California-Davis School of Medicine, Davis, California, 95616, USA
| | - Johnson Tran Le
- Department of Physiology & Membrane Biology, University of California-Davis School of Medicine, Davis, California, 95616, USA
| | - Theanne N Griffith
- Department of Physiology & Membrane Biology, University of California-Davis School of Medicine, Davis, California, 95616, USA
| | - Luis Fernando Santana
- Department of Physiology & Membrane Biology, University of California-Davis School of Medicine, Davis, California, 95616, USA
| | - Robert H Cudmore
- Department of Physiology & Membrane Biology, University of California-Davis School of Medicine, Davis, California, 95616, USA
| |
Collapse
|
12
|
Andrade-Talavera Y, Fisahn A, Rodríguez-Moreno A. Timing to be precise? An overview of spike timing-dependent plasticity, brain rhythmicity, and glial cells interplay within neuronal circuits. Mol Psychiatry 2023; 28:2177-2188. [PMID: 36991134 PMCID: PMC10611582 DOI: 10.1038/s41380-023-02027-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 02/27/2023] [Accepted: 03/01/2023] [Indexed: 03/31/2023]
Abstract
In the mammalian brain information processing and storage rely on the complex coding and decoding events performed by neuronal networks. These actions are based on the computational ability of neurons and their functional engagement in neuronal assemblies where precise timing of action potential firing is crucial. Neuronal circuits manage a myriad of spatially and temporally overlapping inputs to compute specific outputs that are proposed to underly memory traces formation, sensory perception, and cognitive behaviors. Spike-timing-dependent plasticity (STDP) and electrical brain rhythms are suggested to underlie such functions while the physiological evidence of assembly structures and mechanisms driving both processes continues to be scarce. Here, we review foundational and current evidence on timing precision and cooperative neuronal electrical activity driving STDP and brain rhythms, their interactions, and the emerging role of glial cells in such processes. We also provide an overview of their cognitive correlates and discuss current limitations and controversies, future perspectives on experimental approaches, and their application in humans.
Collapse
Affiliation(s)
- Yuniesky Andrade-Talavera
- Laboratory of Cellular Neuroscience and Plasticity, Department of Physiology, Anatomy and Cell Biology, Universidad Pablo de Olavide, ES-41013, Seville, Spain.
| | - André Fisahn
- Department of Biosciences and Nutrition and Department of Women's and Children's Health, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Antonio Rodríguez-Moreno
- Laboratory of Cellular Neuroscience and Plasticity, Department of Physiology, Anatomy and Cell Biology, Universidad Pablo de Olavide, ES-41013, Seville, Spain.
| |
Collapse
|
13
|
Berecki G, Bryson A, Polster T, Petrou S. Biophysical characterization and modelling of SCN1A gain-of-function predicts interneuron hyperexcitability and a predisposition to network instability through homeostatic plasticity. Neurobiol Dis 2023; 179:106059. [PMID: 36868483 DOI: 10.1016/j.nbd.2023.106059] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 02/11/2023] [Accepted: 02/27/2023] [Indexed: 03/05/2023] Open
Abstract
SCN1A gain-of-function variants are associated with early onset developmental and epileptic encephalopathies (DEEs) that possess distinct clinical features compared to Dravet syndrome caused by SCN1A loss-of-function. However, it is unclear how SCN1A gain-of-function may predispose to cortical hyper-excitability and seizures. Here, we first report the clinical features of a patient carrying a de novo SCN1A variant (T162I) associated with neonatal-onset DEE, and then characterize the biophysical properties of T162I and three other SCN1A variants associated with neonatal-onset DEE (I236V) and early infantile DEE (P1345S, R1636Q). In voltage clamp experiments, three variants (T162I, P1345S and R1636Q) exhibited changes in activation and inactivation properties that enhanced window current, consistent with gain-of-function. Dynamic action potential clamp experiments utilising model neurons incorporating Nav1.1. channels supported a gain-of-function mechanism for all four variants. Here, the T162I, I236V, P1345S, and R1636Q variants exhibited higher peak firing rates relative to wild type and the T162I and R1636Q variants produced a hyperpolarized threshold and reduced neuronal rheobase. To explore the impact of these variants upon cortical excitability, we used a spiking network model containing an excitatory pyramidal cell (PC) and parvalbumin positive (PV) interneuron population. SCN1A gain-of-function was modelled by enhancing the excitability of PV interneurons and then incorporating three simple forms of homeostatic plasticity that restored pyramidal cell firing rates. We found that homeostatic plasticity mechanisms exerted differential impact upon network function, with changes to PV-to-PC and PC-to-PC synaptic strength predisposing to network instability. Overall, our findings support a role for SCN1A gain-of-function and inhibitory interneuron hyperexcitability in early onset DEE. We propose a mechanism through which homeostatic plasticity pathways can predispose to pathological excitatory activity and contribute to phenotypic variability in SCN1A disorders.
Collapse
Affiliation(s)
- Géza Berecki
- Ion Channels and Disease Group, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC 3052, Australia.
| | - Alexander Bryson
- Ion Channels and Disease Group, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC 3052, Australia; Department of Neurology, Austin Health, Heidelberg, VIC 3084, Australia
| | - Tilman Polster
- Krankenhaus Mara, Bethel Epilepsy Centre, Department of Epileptology, Medical School, Bielefeld University, Campus Bielefeld-Bethel, Bielefeld, Germany
| | - Steven Petrou
- Ion Channels and Disease Group, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC 3052, Australia; Praxis Precision Medicines, Inc., Cambridge, MA 02142, USA; Department of the Florey Institute, University of Melbourne, Parkville, VIC 3050, Australia.
| |
Collapse
|
14
|
Extrémet J, El Far O, Ankri N, Irani SR, Debanne D, Russier M. An Epitope-Specific LGI1-Autoantibody Enhances Neuronal Excitability by Modulating Kv1.1 Channel. Cells 2022; 11:cells11172713. [PMID: 36078121 PMCID: PMC9454693 DOI: 10.3390/cells11172713] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/22/2022] [Accepted: 08/29/2022] [Indexed: 11/16/2022] Open
Abstract
Leucine-rich Glioma-Inactivated protein 1 (LGI1) is expressed in the central nervous system and its genetic loss of function is associated with epileptic disorders. Additionally, patients with LGI1-directed autoantibodies have frequent focal seizures as a key feature of their disease. LGI1 is composed of a Leucine-Rich Repeat (LRR) and an Epitempin (EPTP) domain. These domains are reported to interact with different members of the transsynaptic complex formed by LGI1 at excitatory synapses, including presynaptic Kv1 potassium channels. Patient-derived recombinant monoclonal antibodies (mAbs) are ideal reagents to study whether domain-specific LGI1-autoantibodies induce epileptiform activities in neurons and their downstream mechanisms. We measured the intrinsic excitability of CA3 pyramidal neurons in organotypic cultures from rat hippocampus treated with either an LRR- or an EPTP-reactive patient-derived mAb, or with IgG from control patients. We found an increase in intrinsic excitability correlated with a reduction of the sensitivity to a selective Kv1.1-channel blocker in neurons treated with the LRR mAb, but not in neurons treated with the EPTP mAb. Our findings suggest LRR mAbs are able to modulate neuronal excitability that could account for epileptiform activity observed in patients.
Collapse
Affiliation(s)
| | - Oussama El Far
- UNIS, INSERM, Aix-Marseille Université, 13015 Marseille, France
| | - Norbert Ankri
- UNIS, INSERM, Aix-Marseille Université, 13015 Marseille, France
| | - Sarosh R. Irani
- Oxford Autoimmune Neurology Group, Nuffield Department of Clinical Neurosciences, Oxford University, Oxford OX3 9DU, UK
| | - Dominique Debanne
- UNIS, INSERM, Aix-Marseille Université, 13015 Marseille, France
- Correspondence: (D.D.); (M.R.)
| | - Michaël Russier
- UNIS, INSERM, Aix-Marseille Université, 13015 Marseille, France
- Correspondence: (D.D.); (M.R.)
| |
Collapse
|
15
|
Kv1.1 channels inhibition in the rat motor cortex recapitulates seizures associated with anti-LGI1 encephalitis. Prog Neurobiol 2022; 213:102262. [DOI: 10.1016/j.pneurobio.2022.102262] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 02/03/2022] [Accepted: 03/08/2022] [Indexed: 12/29/2022]
|
16
|
Baudin P, Cousyn L, Navarro V. The LGI1 protein: molecular structure, physiological functions and disruption-related seizures. Cell Mol Life Sci 2021; 79:16. [PMID: 34967933 PMCID: PMC11072701 DOI: 10.1007/s00018-021-04088-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 12/07/2021] [Accepted: 12/09/2021] [Indexed: 01/16/2023]
Abstract
Leucine-rich, glioma inactivated 1 (LGI1) is a secreted glycoprotein, mainly expressed in the brain, and involved in central nervous system development and physiology. Mutations of LGI1 have been linked to autosomal dominant lateral temporal lobe epilepsy (ADLTE). Recently auto-antibodies against LGI1 have been described as the basis for an autoimmune encephalitis, associated with specific motor and limbic epileptic seizures. It is the second most common cause of autoimmune encephalitis. This review presents details on the molecular structure, expression and physiological functions of LGI1, and examines how their disruption underlies human pathologies. Knock-down of LGI1 in rodents reveals that this protein is necessary for normal brain development. In mature brains, LGI1 is associated with Kv1 channels and AMPA receptors, via domain-specific interaction with membrane anchoring proteins and contributes to regulation of the expression and function of these channels. Loss of function, due to mutations or autoantibodies, of this key protein in the control of neuronal activity is a common feature in the genesis of epileptic seizures in ADLTE and anti-LGI1 autoimmune encephalitis.
Collapse
Affiliation(s)
- Paul Baudin
- Sorbonne Université, Paris Brain Institute - Institut du Cerveau, ICM, INSERM, CNRS, AP-HP, Pitié-Salpêtrière Hospital, Paris, France
| | - Louis Cousyn
- Sorbonne Université, Paris Brain Institute - Institut du Cerveau, ICM, INSERM, CNRS, AP-HP, Pitié-Salpêtrière Hospital, Paris, France
- AP-HP, Epilepsy Unit, Pitié-Salpêtrière Hospital, DMU Neurosciences, Paris, France
| | - Vincent Navarro
- Sorbonne Université, Paris Brain Institute - Institut du Cerveau, ICM, INSERM, CNRS, AP-HP, Pitié-Salpêtrière Hospital, Paris, France.
- AP-HP, Epilepsy Unit, Pitié-Salpêtrière Hospital, DMU Neurosciences, Paris, France.
- AP-HP, Center of Reference for Rare Epilepsies, Pitié-Salpêtrière Hospital, 47-83 Boulevard de l'Hôpital, 75013, Paris, France.
| |
Collapse
|
17
|
Zbili M, Rama S, Benitez MJ, Fronzaroli-Molinieres L, Bialowas A, Boumedine-Guignon N, Garrido JJ, Debanne D. Homeostatic regulation of axonal Kv1.1 channels accounts for both synaptic and intrinsic modifications in the hippocampal CA3 circuit. Proc Natl Acad Sci U S A 2021; 118:e2110601118. [PMID: 34799447 PMCID: PMC8617510 DOI: 10.1073/pnas.2110601118] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/13/2021] [Indexed: 11/18/2022] Open
Abstract
Homeostatic plasticity of intrinsic excitability goes hand in hand with homeostatic plasticity of synaptic transmission. However, the mechanisms linking the two forms of homeostatic regulation have not been identified so far. Using electrophysiological, imaging, and immunohistochemical techniques, we show here that blockade of excitatory synaptic receptors for 2 to 3 d induces an up-regulation of both synaptic transmission at CA3-CA3 connections and intrinsic excitability of CA3 pyramidal neurons. Intrinsic plasticity was found to be mediated by a reduction of Kv1.1 channel density at the axon initial segment. In activity-deprived circuits, CA3-CA3 synapses were found to express a high release probability, an insensitivity to dendrotoxin, and a lack of depolarization-induced presynaptic facilitation, indicating a reduction in presynaptic Kv1.1 function. Further support for the down-regulation of axonal Kv1.1 channels in activity-deprived neurons was the broadening of action potentials measured in the axon. We conclude that regulation of the axonal Kv1.1 channel constitutes a major mechanism linking intrinsic excitability and synaptic strength that accounts for the functional synergy existing between homeostatic regulation of intrinsic excitability and synaptic transmission.
Collapse
Affiliation(s)
- Mickaël Zbili
- Unité de Neurobiologie des canaux Ioniques et de la Synapse (UNIS), UMR_S 1072, INSERM, Aix-Marseille Université, Marseille 13015, France
| | - Sylvain Rama
- Unité de Neurobiologie des canaux Ioniques et de la Synapse (UNIS), UMR_S 1072, INSERM, Aix-Marseille Université, Marseille 13015, France
| | - Maria-José Benitez
- Instituto Cajal, Consejo Superior de Investigaciones Cientificas (CSIC), Madrid 28002, Spain
- Departamento de Química Física Aplicada, Universidad Autónoma de Madrid, Madrid 28049, Spain
| | - Laure Fronzaroli-Molinieres
- Unité de Neurobiologie des canaux Ioniques et de la Synapse (UNIS), UMR_S 1072, INSERM, Aix-Marseille Université, Marseille 13015, France
| | - Andrzej Bialowas
- Unité de Neurobiologie des canaux Ioniques et de la Synapse (UNIS), UMR_S 1072, INSERM, Aix-Marseille Université, Marseille 13015, France
| | - Norah Boumedine-Guignon
- Unité de Neurobiologie des canaux Ioniques et de la Synapse (UNIS), UMR_S 1072, INSERM, Aix-Marseille Université, Marseille 13015, France
| | - Juan José Garrido
- Instituto Cajal, Consejo Superior de Investigaciones Cientificas (CSIC), Madrid 28002, Spain
| | - Dominique Debanne
- Unité de Neurobiologie des canaux Ioniques et de la Synapse (UNIS), UMR_S 1072, INSERM, Aix-Marseille Université, Marseille 13015, France;
| |
Collapse
|
18
|
Conventional measures of intrinsic excitability are poor estimators of neuronal activity under realistic synaptic inputs. PLoS Comput Biol 2021; 17:e1009378. [PMID: 34529674 PMCID: PMC8478185 DOI: 10.1371/journal.pcbi.1009378] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 09/28/2021] [Accepted: 08/24/2021] [Indexed: 11/19/2022] Open
Abstract
Activity-dependent regulation of intrinsic excitability has been shown to greatly contribute to the overall plasticity of neuronal circuits. Such neuroadaptations are commonly investigated in patch clamp experiments using current step stimulation and the resulting input-output functions are analyzed to quantify alterations in intrinsic excitability. However, it is rarely addressed, how such changes translate to the function of neurons when they operate under natural synaptic inputs. Still, it is reasonable to expect that a strong correlation and near proportional relationship exist between static firing responses and those evoked by synaptic drive. We challenge this view by performing a high-yield electrophysiological analysis of cultured mouse hippocampal neurons using both standard protocols and simulated synaptic inputs via dynamic clamp. We find that under these conditions the neurons exhibit vastly different firing responses with surprisingly weak correlation between static and dynamic firing intensities. These contrasting responses are regulated by two intrinsic K-currents mediated by Kv1 and Kir channels, respectively. Pharmacological manipulation of the K-currents produces differential regulation of the firing output of neurons. Static firing responses are greatly increased in stuttering type neurons under blocking their Kv1 channels, while the synaptic responses of the same neurons are less affected. Pharmacological blocking of Kir-channels in delayed firing type neurons, on the other hand, exhibit the opposite effects. Our subsequent computational model simulations confirm the findings in the electrophysiological experiments and also show that adaptive changes in the kinetic properties of such currents can even produce paradoxical regulation of the firing output.
Collapse
|
19
|
Neural excitability increases with axonal resistance between soma and axon initial segment. Proc Natl Acad Sci U S A 2021; 118:2102217118. [PMID: 34389672 DOI: 10.1073/pnas.2102217118] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The position of the axon initial segment (AIS) is thought to play a critical role in neuronal excitability. Previous experimental studies have found that a distal shift in AIS position correlates with a reduction in excitability. Yet theoretical work has suggested the opposite, because of increased electrical isolation. A distal shift in AIS position corresponds to an elevation of axial resistance R a We therefore examined how changes in R a at the axon hillock impact the voltage threshold (Vth) of the somatic action potential in L5 pyramidal neurons. Increasing R a by mechanically pinching the axon between the soma and the AIS was found to lower Vth by ∼6 mV. Conversely, decreasing R a by substituting internal ions with higher mobility elevated Vth All R a -dependent changes in Vth could be reproduced in a Hodgkin-Huxley compartmental model. We conclude that in L5 pyramidal neurons, excitability increases with axial resistance and therefore with a distal shift of the AIS.
Collapse
|
20
|
Robert V, Therreau L, Davatolhagh MF, Bernardo-Garcia FJ, Clements KN, Chevaleyre V, Piskorowski RA. The mechanisms shaping CA2 pyramidal neuron action potential bursting induced by muscarinic acetylcholine receptor activation. J Gen Physiol 2021; 152:133812. [PMID: 32069351 PMCID: PMC7141590 DOI: 10.1085/jgp.201912462] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 12/18/2019] [Accepted: 01/13/2020] [Indexed: 01/04/2023] Open
Abstract
Recent studies have revealed that hippocampal area CA2 plays an important role in hippocampal network function. Disruption of this region has been implicated in neuropsychiatric disorders. It is well appreciated that cholinergic input to the hippocampus plays an important role in learning and memory. While the effect of elevated cholinergic tone has been well studied in areas CA1 and CA3, it remains unclear how changes in cholinergic tone impact synaptic transmission and the intrinsic properties of neurons in area CA2. In this study, we applied the cholinergic agonist carbachol and performed on-cell, whole-cell, and extracellular recordings in area CA2. We observed that under conditions of high cholinergic tone, CA2 pyramidal neurons depolarized and rhythmically fired bursts of action potentials. This depolarization depended on the activation of M1 and M3 cholinergic receptors. Furthermore, we examined how the intrinsic properties and action-potential firing were altered in CA2 pyramidal neurons treated with 10 µM carbachol. While this intrinsic burst firing persisted in the absence of synaptic transmission, bursts were shaped by synaptic inputs in the intact network. We found that both excitatory and inhibitory synaptic transmission were reduced upon carbachol treatment. Finally, we examined the contribution of different channels to the cholinergic-induced changes in neuronal properties. We found that a conductance from Kv7 channels partially contributed to carbachol-induced changes in resting membrane potential and membrane resistance. We also found that D-type potassium currents contributed to controlling several properties of the bursts, including firing rate and burst kinetics. Furthermore, we determined that T-type calcium channels and small conductance calcium-activated potassium channels play a role in regulating bursting activity.
Collapse
Affiliation(s)
- Vincent Robert
- Université Paris Descartes, Inserm UMR1266, Institute of Psychiatry and Neuroscience of Paris, Team Synaptic Plasticity and Neural Networks, Paris, France
| | - Ludivine Therreau
- Université Paris Descartes, Inserm UMR1266, Institute of Psychiatry and Neuroscience of Paris, Team Synaptic Plasticity and Neural Networks, Paris, France
| | - M Felicia Davatolhagh
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - F Javier Bernardo-Garcia
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA
| | | | - Vivien Chevaleyre
- Université Paris Descartes, Inserm UMR1266, Institute of Psychiatry and Neuroscience of Paris, Team Synaptic Plasticity and Neural Networks, Paris, France
| | - Rebecca A Piskorowski
- Université Paris Descartes, Inserm UMR1266, Institute of Psychiatry and Neuroscience of Paris, Team Synaptic Plasticity and Neural Networks, Paris, France
| |
Collapse
|
21
|
Ahmadirad N, Fathollahi Y, Janahmadi M, Ghasemi Z, Shojaei A, Rezaei M, Barkley V, Mirnajafi-Zadeh J. The role of α adrenergic receptors in mediating the inhibitory effect of electrical brain stimulation on epileptiform activity in rat hippocampal slices. Brain Res 2021; 1765:147492. [PMID: 33887250 DOI: 10.1016/j.brainres.2021.147492] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 04/06/2021] [Accepted: 04/14/2021] [Indexed: 10/21/2022]
Abstract
The Inhibitory effect of electrical low-frequency stimulation (LFS) on neuronal excitability and seizure occurrence has been indicated in experimental models, but the precise mechanism has not established. This investigation was intended to figure out the role of α1 and α2 adrenergic receptors in LFS' inhibitory effect on neuronal excitability. Epileptiform activity induced in an in vitro rat hippocampal slice preparation by high K+ ACSF and LFS (900 square wave pulses at 1 Hz) was administered at the beginning of epileptiform activity to the Schaffer collaterals. In CA1 pyramidal neurons, the electrophysiological properties were measured at the baseline, before high K+ ACSF washout, and at 15 min after high K+ ACSF washout using whole-cell, patch-clamp recording. Results indicated that after high K+ ACSF washout, prazosine (10 µM; α1 adrenergic receptor antagonist) and yohimbine (5 µM; α2 adrenergic receptor antagonist) suppressed the LFS' effect of reducing rheobase current and utilization time following depolarizing ramp current, the latency to the first spike following a depolarizing current and latency to the first rebound action potential following hyperpolarizing current pulses. Thus, it may be proposed that LFS' inhibitory action on the neuronal hyperexcitability, in some way, is mediated by α1 and α2 adrenergic receptors.
Collapse
Affiliation(s)
- Nooshin Ahmadirad
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Yaghoub Fathollahi
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mahyar Janahmadi
- Department of Physiology, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Ghasemi
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran; Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Canada
| | - Amir Shojaei
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mahmoud Rezaei
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Victoria Barkley
- Krembil Research Institute, University Health Network, Toronto, Canada
| | - Javad Mirnajafi-Zadeh
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran; Institute for Brain Sciences and Cognition, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
22
|
Wason TD. A model integrating multiple processes of synchronization and coherence for information instantiation within a cortical area. Biosystems 2021; 205:104403. [PMID: 33746019 DOI: 10.1016/j.biosystems.2021.104403] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 03/05/2021] [Indexed: 12/14/2022]
Abstract
What is the form of dynamic, e.g., sensory, information in the mammalian cortex? Information in the cortex is modeled as a coherence map of a mixed chimera state of synchronous, phasic, and disordered minicolumns. The theoretical model is built on neurophysiological evidence. Complex spatiotemporal information is instantiated through a system of interacting biological processes that generate a synchronized cortical area, a coherent aperture. Minicolumn elements are grouped in macrocolumns in an array analogous to a phased-array radar, modeled as an aperture, a "hole through which radiant energy flows." Coherence maps in a cortical area transform inputs from multiple sources into outputs to multiple targets, while reducing complexity and entropy. Coherent apertures can assume extremely large numbers of different information states as coherence maps, which can be communicated among apertures with corresponding very large bandwidths. The coherent aperture model incorporates considerable reported research, integrating five conceptually and mathematically independent processes: 1) a damped Kuramoto network model, 2) a pumped area field potential, 3) the gating of nearly coincident spikes, 4) the coherence of activity across cortical lamina, and 5) complex information formed through functions in macrocolumns. Biological processes and their interactions are described in equations and a functional circuit such that the mathematical pieces can be assembled the same way the neurophysiological ones are. The model can be conceptually convolved over the specifics of local cortical areas within and across species. A coherent aperture becomes a node in a graph of cortical areas with a corresponding distribution of information.
Collapse
Affiliation(s)
- Thomas D Wason
- North Carolina State University, Department of Biological Sciences, Meitzen Laboratory, Campus Box 7617, 128 David Clark Labs, Raleigh, NC 27695-7617, USA.
| |
Collapse
|
23
|
Tian Y, Korn P, Tripathi P, Komnig D, Wiemuth D, Nikouee A, Classen A, Bolm C, Falkenburger BH, Lüscher B, Gründer S. The mono-ADP-ribosyltransferase ARTD10 regulates the voltage-gated K + channel Kv1.1 through protein kinase C delta. BMC Biol 2020; 18:143. [PMID: 33059680 PMCID: PMC7558731 DOI: 10.1186/s12915-020-00878-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 09/24/2020] [Indexed: 11/30/2022] Open
Abstract
Background ADP-ribosylation is a ubiquitous post-translational modification that involves both mono- and poly-ADP-ribosylation. ARTD10, also known as PARP10, mediates mono-ADP-ribosylation (MARylation) of substrate proteins. A previous screen identified protein kinase C delta (PKCδ) as a potential ARTD10 substrate, among several other kinases. The voltage-gated K+ channel Kv1.1 constitutes one of the dominant Kv channels in neurons of the central nervous system and the inactivation properties of Kv1.1 are modulated by PKC. In this study, we addressed the role of ARTD10-PKCδ as a regulator of Kv1.1. Results We found that ARTD10 inhibited PKCδ, which increased Kv1.1 current amplitude and the proportion of the inactivating current component in HeLa cells, indicating that ARTD10 regulates Kv1.1 in living cells. An inhibitor of ARTD10, OUL35, significantly decreased peak amplitude together with the proportion of the inactivating current component of Kv1.1-containing channels in primary hippocampal neurons, demonstrating that the ARTD10-PKCδ signaling cascade regulates native Kv1.1. Moreover, we show that the pharmacological blockade of ARTD10 increases excitability of hippocampal neurons. Conclusions Our results, for the first time, suggest that MARylation by ARTD10 controls neuronal excitability.
Collapse
Affiliation(s)
- Yuemin Tian
- Institute of Physiology, RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Patricia Korn
- Institute of Biochemistry and Molecular Biology, RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Priyanka Tripathi
- Institute of Physiology, RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany.,Present address: Institute of Neuropathology, RWTH Aachen University Medical School, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Daniel Komnig
- Department of Neurology, RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany.,JARA-Institute Molecular Neuroscience and Neuroimaging, Forschungszentrum Jülich, Jülich, Germany
| | - Dominik Wiemuth
- Institute of Physiology, RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Azadeh Nikouee
- Institute of Physiology, RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Arno Classen
- Institute of Organic Chemistry, RWTH Aachen University, Landoltweg 1, 52056, Aachen, Germany
| | - Carsten Bolm
- Institute of Organic Chemistry, RWTH Aachen University, Landoltweg 1, 52056, Aachen, Germany
| | - Björn H Falkenburger
- Department of Neurology, RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany.,JARA-Institute Molecular Neuroscience and Neuroimaging, Forschungszentrum Jülich, Jülich, Germany.,Present address: Department of Neurology, Dresden University Medical Center, Fetscherstraße 74, 01307, Dresden, Germany
| | - Bernhard Lüscher
- Institute of Biochemistry and Molecular Biology, RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Stefan Gründer
- Institute of Physiology, RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany.
| |
Collapse
|
24
|
Sun H, Zhang H, Ross A, Wang TT, Al-Chami A, Wu SH. Developmentally Regulated Rebound Depolarization Enhances Spike Timing Precision in Auditory Midbrain Neurons. Front Cell Neurosci 2020; 14:236. [PMID: 32848625 PMCID: PMC7424072 DOI: 10.3389/fncel.2020.00236] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 07/06/2020] [Indexed: 12/23/2022] Open
Abstract
The inferior colliculus (IC) is an auditory midbrain structure involved in processing biologically important temporal features of sounds. The responses of IC neurons to these temporal features reflect an interaction of synaptic inputs and neuronal biophysical properties. One striking biophysical property of IC neurons is the rebound depolarization produced following membrane hyperpolarization. To understand how the rebound depolarization is involved in spike timing, we made whole-cell patch clamp recordings from IC neurons in brain slices of P9-21 rats. We found that the percentage of rebound neurons was developmentally regulated. The precision of the timing of the first spike on the rebound increased when the neuron was repetitively injected with a depolarizing current following membrane hyperpolarization. The average jitter of the first spikes was only 0.5 ms. The selective T-type Ca2+ channel antagonist, mibefradil, significantly increased the jitter of the first spike of neurons in response to repetitive depolarization following membrane hyperpolarization. Furthermore, the rebound was potentiated by one to two preceding rebounds within a few hundred milliseconds. The first spike generated on the potentiated rebound was more precise than that on the non-potentiated rebound. With the addition of a calcium chelator, BAPTA, into the cell, the rebound potentiation no longer occurred, and the precision of the first spike on the rebound was not improved. These results suggest that the postinhibitory rebound mediated by T-type Ca2+ channel promotes spike timing precision in IC neurons. The rebound potentiation and precise spikes may be induced by increases in intracellular calcium levels.
Collapse
Affiliation(s)
- Hongyu Sun
- Department of Neuroscience, Carleton University, Ottawa, ON, Canada
| | - Hui Zhang
- Department of Neuroscience, Carleton University, Ottawa, ON, Canada
| | - Alysia Ross
- Department of Neuroscience, Carleton University, Ottawa, ON, Canada
| | - Ting Ting Wang
- Department of Neuroscience, Carleton University, Ottawa, ON, Canada
| | - Aycheh Al-Chami
- Department of Neuroscience, Carleton University, Ottawa, ON, Canada
| | - Shu Hui Wu
- Department of Neuroscience, Carleton University, Ottawa, ON, Canada
| |
Collapse
|
25
|
Lignani G, Baldelli P, Marra V. Homeostatic Plasticity in Epilepsy. Front Cell Neurosci 2020; 14:197. [PMID: 32676011 PMCID: PMC7333442 DOI: 10.3389/fncel.2020.00197] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 06/05/2020] [Indexed: 11/26/2022] Open
Abstract
In the healthy brain, neuronal excitability and synaptic strength are homeostatically regulated to keep neuronal network activity within physiological boundaries. Epilepsy is characterized by episodes of highly synchronized firing across in widespread neuronal populations, due to a failure in regulation of network activity. Here we consider epilepsy as a failure of homeostatic plasticity or as a maladaptive response to perturbations in the activity. How homeostatic compensation is involved in epileptogenic processes or in the chronic phase of epilepsy, is still debated. Although several theories have been proposed, there is relatively little experimental evidence to evaluate them. In this perspective, we will discuss recent results that shed light on the potential role of homeostatic plasticity in epilepsy. First, we will present some recent insights on how homeostatic compensations are probably active before and during epileptogenesis and how their actions are temporally regulated and closely dependent on the progression of pathology. Then, we will consider the dual role of transcriptional regulation during epileptogenesis, and finally, we will underline the importance of homeostatic plasticity in the context of therapeutic interventions for epilepsy. While classic pharmacological interventions may be counteracted by the epileptic brain to maintain its potentially dysfunctional set point, novel therapeutic approaches may provide the neuronal network with the tools necessary to restore its physiological balance.
Collapse
Affiliation(s)
- Gabriele Lignani
- Department of Clinical and Experimental Epilepsy, Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Pietro Baldelli
- Department of Experimental Medicine, University of Genoa, Genoa, Italy.,IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Vincenzo Marra
- Department of Neuroscience, Psychology and Behaviour, University of Leicester, Leicester, United Kingdom
| |
Collapse
|
26
|
Feria Pliego JA, Pedroarena CM. Kv1 potassium channels control action potential firing of putative GABAergic deep cerebellar nuclear neurons. Sci Rep 2020; 10:6954. [PMID: 32332769 PMCID: PMC7181752 DOI: 10.1038/s41598-020-63583-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 04/01/2020] [Indexed: 12/26/2022] Open
Abstract
Low threshold voltage activated Kv1 potassium channels play key roles in regulating action potential (AP) threshold, neural excitability, and synaptic transmission. Kv1 channels are highly expressed in the cerebellum and mutations of human Kv1 genes are associated to episodic forms of ataxia (EAT-1). Besides the well-established role of Kv1 channels in controlling the cerebellar basket-Purkinje cells synapses, Kv1 channels are expressed by the deep cerebellar nuclear neurons (DCNs) where they regulate the activity of principal DCNs carrying the cerebellar output. DCNs include as well GABAergic neurons serving important functions, such as those forming the inhibitory nucleo-olivary pathway, the nucleo-cortical DCNs providing feed-back inhibition to the cerebellar cortex, and those targeting principal DCNs, but whether their function is regulated by Kv1 channels remains unclear. Here, using cerebellar slices from mature GAD67-GFP mice to identify putative GABAergic-DCNs (GAD + DCN) we show that specific Kv1 channel blockers (dendrotoxin-alpha/I/K, DTXs) hyperpolarized the threshold of somatic action potentials, increased the spontaneous firing rate and hampered evoked high frequency repetitive responses of GAD + DCNs. Moreover, DTXs induced somatic depolarization and tonic firing in previously silent, putative nucleo-cortical DCNs. These results reveal a novel role of Kv1 channels in regulating GABAergic-DCNs activity and thereby, cerebellar function at multiple levels.
Collapse
Affiliation(s)
- Jessica Abigail Feria Pliego
- Graduate School of Cellular and Molecular Neurosciences, University of Tübingen, Tübingen, Germany.,Department for Cognitive Neurology, Hertie-Institute for Clinical Brain Research, 72076, Tübingen, Germany.,Systems Neurophysiology, Werner Reichardt Center for Integrative Neuroscience, University of Tübingen, 72076, Tübingen, Germany
| | - Christine M Pedroarena
- Department for Cognitive Neurology, Hertie-Institute for Clinical Brain Research, 72076, Tübingen, Germany. .,Systems Neurophysiology, Werner Reichardt Center for Integrative Neuroscience, University of Tübingen, 72076, Tübingen, Germany.
| |
Collapse
|
27
|
Kirschstein T, Sadkiewicz E, Hund-Göschel G, Becker J, Guli X, Müller S, Rohde M, Hübner DC, Brehme H, Kolbaske S, Porath K, Sellmann T, Großmann A, Wittstock M, Syrbe S, Storch A, Köhling R. Stereotactically Injected Kv1.2 and CASPR2 Antisera Cause Differential Effects on CA1 Synaptic and Cellular Excitability, but Both Enhance the Vulnerability to Pro-epileptic Conditions. Front Synaptic Neurosci 2020; 12:13. [PMID: 32269520 PMCID: PMC7110982 DOI: 10.3389/fnsyn.2020.00013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 03/06/2020] [Indexed: 12/12/2022] Open
Abstract
PURPOSE We present a case of voltage-gated potassium channel (VGKC) complex antibody-positive limbic encephalitis (LE) harboring autoantibodies against Kv1.2. Since the patient responded well to immunotherapy, the autoantibodies were regarded as pathogenic. We aimed to characterize the pathophysiological role of this antibody in comparison to an antibody against the VGKC-associated protein contactin-associated protein-2 (CASPR2). METHODS Stereotactic injection of patient sera (anti-Kv1.2-associated LE or anti-CASPR2 encephalopathy) and a control subject was performed into the hippocampus of the anesthetized rat in vivo, and hippocampal slices were prepared for electrophysiological purposes. Using extra- and intracellular techniques, synaptic transmission, long-term potentiation (LTP) and vulnerability to pro-epileptic conditions were analyzed. RESULTS We observed that the slope of the field excitatory postsynaptic potential (fEPSP) was significantly increased at Schaffer collateral-CA1 synapses in anti-Kv1.2-treated and anti-CASPR2-treated rats, but not at medial perforant path-dentate gyrus synapses. The increase of the fEPSP slope in CA1 was accompanied by a decrease of the paired-pulse ratio in anti-Kv1.2, but not in anti-CASPR2 tissue, indicating presynaptic site of anti-Kv1.2. In addition, anti-Kv1.2 tissue showed enhanced LTP in CA1, but dentate gyrus LTP remained unaltered. Importantly, LTP in slices from anti-CASPR2-treated animals did not differ from control values. Intracellular recordings from CA1 neurons revealed that the resting membrane potential and a single action potential were not different between anti-Kv1.2 and control tissue. However, when the depolarization was prolonged, the number of action potentials elicited was reduced in anti-Kv1.2-treated tissue compared to both control and anti-CASPR2 tissue. In contrast, polyspike discharges induced by removal of Mg2+ occurred earlier and more frequently in both patient sera compared to control. CONCLUSION Patient serum containing anti-Kv1.2 facilitates presynaptic transmitter release as well as postsynaptic depolarization at the Schaffer-collateral-CA1 synapse, but not in the dentate gyrus. As a consequence, both synaptic transmission and LTP in CA1 are facilitated and action potential firing is altered. In contrast, anti-CASPR2 leads to increased postsynaptic potentials, but without changing LTP or firing properties suggesting that anti-Kv1.2 and anti-CASPR2 differ in their cellular effects. Both patient sera alter susceptibility to epileptic conditions, but presumably by different mechanisms.
Collapse
Affiliation(s)
- Timo Kirschstein
- Oscar Langendorff Institute of Physiology, University of Rostock, Rostock, Germany
- Department of Neurology, University of Rostock, Rostock, Germany
- Center of Transdisciplinary Neurosciences Rostock, University of Rostock, Rostock, Germany
| | - Erika Sadkiewicz
- Oscar Langendorff Institute of Physiology, University of Rostock, Rostock, Germany
| | - Gerda Hund-Göschel
- Oscar Langendorff Institute of Physiology, University of Rostock, Rostock, Germany
| | - Juliane Becker
- Oscar Langendorff Institute of Physiology, University of Rostock, Rostock, Germany
| | - Xiati Guli
- Oscar Langendorff Institute of Physiology, University of Rostock, Rostock, Germany
| | - Steffen Müller
- Oscar Langendorff Institute of Physiology, University of Rostock, Rostock, Germany
| | - Marco Rohde
- Oscar Langendorff Institute of Physiology, University of Rostock, Rostock, Germany
| | | | - Hannes Brehme
- Department of Neurology, University of Rostock, Rostock, Germany
| | - Stephan Kolbaske
- Department of Neurology, University of Rostock, Rostock, Germany
| | - Katrin Porath
- Oscar Langendorff Institute of Physiology, University of Rostock, Rostock, Germany
| | - Tina Sellmann
- Oscar Langendorff Institute of Physiology, University of Rostock, Rostock, Germany
| | - Annette Großmann
- Institute of Diagnostic and Intervention Radiology, University of Rostock, Rostock, Germany
| | | | - Steffen Syrbe
- Clinik for Pediatric and Adolescent Medicine, University of Heidelberg, Heidelberg, Germany
| | - Alexander Storch
- Department of Neurology, University of Rostock, Rostock, Germany
- Center of Transdisciplinary Neurosciences Rostock, University of Rostock, Rostock, Germany
| | - Rüdiger Köhling
- Oscar Langendorff Institute of Physiology, University of Rostock, Rostock, Germany
- Center of Transdisciplinary Neurosciences Rostock, University of Rostock, Rostock, Germany
| |
Collapse
|
28
|
Yang F, Yang L, Wataya-Kaneda M, Teng L, Katayama I. Epilepsy in a melanocyte-lineage mTOR hyperactivation mouse model: A novel epilepsy model. PLoS One 2020; 15:e0228204. [PMID: 31978189 PMCID: PMC6980560 DOI: 10.1371/journal.pone.0228204] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 01/09/2020] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE To clarify the complex mechanism underlying epileptogeneis, a novel animal model was generated. METHODS In our previous research, we have generated a melanocyte-lineage mTOR hyperactivation mouse model (Mitf-M-Cre Tsc2 KO mice; cKO mice) to investigate mTOR pathway in melanogenesis regulation, markedly reduced skin pigmentation was observed. Very unexpectedly, spontaneous recurrent epilepsy was also developed in this mouse model. RESULTS Compared with control littermates, no change was found in either brain size or brain mass in cKO mice. Hematoxylin staining revealed no obvious aberrant histologic features in the whole brains of cKO mice. Histoimmunofluorescence staining and electron microscopy examination revealed markedly increased mTOR signaling and hyperproliferation of mitochondria in cKO mice, especially in the hippocampus. Furthermore, rapamycin treatment reversed these abnormalities. CONCLUSIONS This study suggests that our melanocyte-lineage mTOR hyperactivation mouse is a novel animal model of epilepsy, which may promote the progress of both epilepsy and neurophysiology research.
Collapse
Affiliation(s)
- Fei Yang
- Department of Dermatology, Course of Integrated Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Lingli Yang
- Department of Dermatology, Course of Integrated Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Mari Wataya-Kaneda
- Department of Dermatology, Course of Integrated Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Lanting Teng
- Department of Dermatology, Course of Integrated Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Ichiro Katayama
- Department of Dermatology, Course of Integrated Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| |
Collapse
|
29
|
Impaired Reliability and Precision of Spiking in Adults But Not Juveniles in a Mouse Model of Fragile X Syndrome. eNeuro 2019; 6:ENEURO.0217-19.2019. [PMID: 31685673 PMCID: PMC6917895 DOI: 10.1523/eneuro.0217-19.2019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 10/02/2019] [Accepted: 10/04/2019] [Indexed: 12/26/2022] Open
Abstract
Fragile X syndrome (FXS) is the most common source of intellectual disability and autism. Extensive studies have been performed on the network and behavioral correlates of the syndrome, but our knowledge about intrinsic conductance changes is still limited. In this study, we show a differential effect of FMRP knockout in different subsections of hippocampus using whole-cell patch clamp in mouse hippocampal slices. We observed no significant change in spike numbers in the CA1 region of hippocampus, but a significant increase in CA3, in juvenile mice. However, in adult mice we see a reduction in spike number in the CA1 with no significant difference in CA3. In addition, we see increased variability in spike numbers in CA1 cells following a variety of steady and modulated current step protocols. This effect emerges in adult mice (8 weeks) but not juvenile mice (4 weeks). This increased spiking variability was correlated with reduced spike number and with elevated AHP. The increased AHP arose from elevated SK currents (small conductance calcium-activated potassium channels), but other currents involved in medium AHP, such as Ih and M, were not significantly different. We obtained a partial rescue of the cellular variability phenotype when we blocked SK current using the specific blocker apamin. Our observations provide a single-cell correlate of the network observations of response variability and loss of synchronization, and suggest that the elevation of SK currents in FXS may provide a partial mechanistic explanation for this difference.
Collapse
|
30
|
Morgan PJ, Bourboulou R, Filippi C, Koenig-Gambini J, Epsztein J. Kv1.1 contributes to a rapid homeostatic plasticity of intrinsic excitability in CA1 pyramidal neurons in vivo. eLife 2019; 8:49915. [PMID: 31774395 PMCID: PMC6881145 DOI: 10.7554/elife.49915] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 11/08/2019] [Indexed: 12/12/2022] Open
Abstract
In area CA1 of the hippocampus, the selection of place cells to represent a new environment is biased towards neurons with higher excitability. However, different environments are represented by orthogonal cell ensembles, suggesting that regulatory mechanisms exist. Activity-dependent plasticity of intrinsic excitability, as observed in vitro, is an attractive candidate. Here, using whole-cell patch-clamp recordings of CA1 pyramidal neurons in anesthetized rats, we have examined how inducing theta-bursts of action potentials affects their intrinsic excitability over time. We observed a long-lasting, homeostatic depression of intrinsic excitability which commenced within minutes, and, in contrast to in vitro observations, was not mediated by dendritic Ih. Instead, it was attenuated by the Kv1.1 channel blocker dendrotoxin K, suggesting an axonal origin. Analysis of place cells’ out-of-field firing in mice navigating in virtual reality further revealed an experience-dependent reduction consistent with decreased excitability. We propose that this mechanism could reduce memory interference.
Collapse
Affiliation(s)
- Peter James Morgan
- Institute of Neurobiology of the Mediterranean Sea (INMED), Turing Center for Living Systems (CENTURI), Aix-Marseille University, INSERM, Marseille, France
| | - Romain Bourboulou
- Institute of Neurobiology of the Mediterranean Sea (INMED), Turing Center for Living Systems (CENTURI), Aix-Marseille University, INSERM, Marseille, France
| | - Caroline Filippi
- Institute of Neurobiology of the Mediterranean Sea (INMED), Turing Center for Living Systems (CENTURI), Aix-Marseille University, INSERM, Marseille, France
| | - Julie Koenig-Gambini
- Institute of Neurobiology of the Mediterranean Sea (INMED), Turing Center for Living Systems (CENTURI), Aix-Marseille University, INSERM, Marseille, France.,Institut Universitaire de France, Paris, France
| | - Jérôme Epsztein
- Institute of Neurobiology of the Mediterranean Sea (INMED), Turing Center for Living Systems (CENTURI), Aix-Marseille University, INSERM, Marseille, France
| |
Collapse
|
31
|
Soldado-Magraner S, Brandalise F, Honnuraiah S, Pfeiffer M, Moulinier M, Gerber U, Douglas R. Conditioning by subthreshold synaptic input changes the intrinsic firing pattern of CA3 hippocampal neurons. J Neurophysiol 2019; 123:90-106. [PMID: 31721636 DOI: 10.1152/jn.00506.2019] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Unlike synaptic strength, intrinsic excitability is assumed to be a stable property of neurons. For example, learning of somatic conductances is generally not incorporated into computational models, and the discharge pattern of neurons in response to test stimuli is frequently used as a basis for phenotypic classification. However, it is increasingly evident that signal processing properties of neurons are more generally plastic on the timescale of minutes. Here we demonstrate that the intrinsic firing patterns of CA3 neurons of the rat hippocampus in vitro undergo rapid long-term plasticity in response to a few minutes of only subthreshold synaptic conditioning. This plasticity on the spike timing could also be induced by intrasomatic injection of subthreshold depolarizing pulses and was blocked by kinase inhibitors, indicating that discharge dynamics are modulated locally. Cluster analysis of firing patterns before and after conditioning revealed systematic transitions toward adapting and intrinsic burst behaviors, irrespective of the patterns initially exhibited by the cells. We used a conductance-based model to decide appropriate pharmacological blockade and found that the observed transitions are likely due to recruitment of low-voltage calcium and Kv7 potassium conductances. We conclude that CA3 neurons adapt their conductance profile to the subthreshold activity of their input, so that their intrinsic firing pattern is not a static signature, but rather a reflection of their history of subthreshold activity. In this way, recurrent output from CA3 neurons may collectively shape the temporal dynamics of their embedding circuits.NEW & NOTEWORTHY Although firing patterns are widely conserved across the animal phyla, it is still a mystery why nerve cells present such diversity of discharge dynamics upon somatic step currents. Adding a new timing dimension to the intrinsic plasticity literature, here we show that CA3 neurons rapidly adapt through the space of known firing patterns in response to the subthreshold signals that they receive from their embedding circuit, potentially adjusting their network processing to the temporal statistics of their circuit.
Collapse
Affiliation(s)
| | - Federico Brandalise
- Brain Research Institute, University of Zurich, Switzerland.,Department of Fundamental Neurosciences, University of Geneva, Switzerland
| | - Suraj Honnuraiah
- Institute of Neuroinformatics, University of Zurich and ETH Zurich, Switzerland
| | - Michael Pfeiffer
- Institute of Neuroinformatics, University of Zurich and ETH Zurich, Switzerland
| | - Marie Moulinier
- Department of Fundamental Neurosciences, University of Geneva, Switzerland
| | - Urs Gerber
- Brain Research Institute, University of Zurich, Switzerland
| | - Rodney Douglas
- Institute of Neuroinformatics, University of Zurich and ETH Zurich, Switzerland
| |
Collapse
|
32
|
Debanne D, Russier M. The contribution of ion channels in input-output plasticity. Neurobiol Learn Mem 2019; 166:107095. [PMID: 31539624 DOI: 10.1016/j.nlm.2019.107095] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 09/04/2019] [Accepted: 09/16/2019] [Indexed: 12/20/2022]
Abstract
Persistent changes that occur in brain circuits are classically thought to be mediated by long-term modifications in synaptic efficacy. Yet, many studies have shown that voltage-gated ion channels located at the input and output side of the neurons are also the subject to persistent modifications. These channels are thus responsible for intrinsic plasticity that is expressed in many different neuronal types including glutamatergic principal neurons and GABAergic interneurons. As for synaptic plasticity, activation of synaptic glutamate receptors initiate persistent modification in neuronal excitability. We review here how synaptic input can be efficiently altered by activity-dependent modulation of ion channels that control EPSP amplification, spike threshold or resting membrane potential. We discuss the nature of the learning rules shared by intrinsic and synaptic plasticity, the mechanisms of ion channel regulation and the impact of intrinsic plasticity on induction of synaptic modifications.
Collapse
|
33
|
Plasticity of intrinsic neuronal excitability. Curr Opin Neurobiol 2019; 54:73-82. [DOI: 10.1016/j.conb.2018.09.001] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 08/17/2018] [Accepted: 09/06/2018] [Indexed: 11/20/2022]
|
34
|
Shim HG, Jang SS, Kim SH, Hwang EM, Min JO, Kim HY, Kim YS, Ryu C, Chung G, Kim Y, Yoon BE, Kim SJ. TNF-α increases the intrinsic excitability of cerebellar Purkinje cells through elevating glutamate release in Bergmann Glia. Sci Rep 2018; 8:11589. [PMID: 30072733 PMCID: PMC6072779 DOI: 10.1038/s41598-018-29786-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 07/18/2018] [Indexed: 11/17/2022] Open
Abstract
For decades, the glial function has been highlighted not only as the ‘structural glue’, but also as an ‘active participant’ in neural circuits. Here, we suggest that tumor necrosis factor α (TNF-α), a key inflammatory cytokine, alters the neural activity of the cerebellar Purkinje cells (PCs) by facilitating gliotransmission in the juvenile male rat cerebellum. A bath application of TNF-α (100 ng/ml) in acute cerebellar slices elevates spiking activity of PCs with no alterations in the regularity of PC firings. Interestingly, the effect of TNF-α on the intrinsic excitability of PCs was abolished under a condition in which the type1 TNF receptor (TNFR1) in Bergmann glia (BG) was genetically suppressed by viral delivery of an adeno-associated virus (AAV) containing TNFR1-shRNA. In addition, we measured the concentration of glutamate derived from dissociated cerebellar cortical astrocyte cultures treated with TNF-α and observed a progressive increase of glutamate in a time-dependent manner. We hypothesised that TNF-α-induced elevation of glutamate from BGs enveloping the synaptic cleft may directly activate metabotropic glutamate receptor1 (mGluR1). Pharmacological inhibition of mGluR1, indeed, prevented the TNF-α-mediated elevation of the intrinsic excitability in PCs. Taken together, our study reveals that TNF-α triggers glutamate release in BG, thereby increasing the intrinsic excitability of cerebellar PCs in a mGluR1-dependent manner.
Collapse
Affiliation(s)
- Hyun Geun Shim
- Department of Physiology, Seoul National University College of Medicine, Seoul, Korea.,Department of Biomedical Science, Seoul National University College of Medicine, Seoul, Korea
| | - Sung-Soo Jang
- Department of Physiology, Seoul National University College of Medicine, Seoul, Korea.,Neuroscience Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Seung Ha Kim
- Department of Physiology, Seoul National University College of Medicine, Seoul, Korea.,Department of Biomedical Science, Seoul National University College of Medicine, Seoul, Korea
| | - Eun Mi Hwang
- Center for Functional Connectomics, Korea Institute of Science and Technology, Seoul, Korea
| | - Joo Ok Min
- Department of Molecular biology, Dankook University, Chungnam, Korea
| | - Hye Yun Kim
- Department of Pharmacy and Integrated Science and Engineering Division, Yonsei University, Incheon, Korea
| | - Yoo Sung Kim
- Department of Molecular biology, Dankook University, Chungnam, Korea
| | - Changhyeon Ryu
- Department of Physiology, Seoul National University College of Medicine, Seoul, Korea.,Department of Biomedical Science, Seoul National University College of Medicine, Seoul, Korea.,Neuroscience Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Geehoon Chung
- Department of Physiology, Seoul National University College of Medicine, Seoul, Korea.,Department of Brain and Cognitive Science, College of Science, Seoul National University, Seoul, Korea
| | - YoungSoo Kim
- Department of Pharmacy and Integrated Science and Engineering Division, Yonsei University, Incheon, Korea
| | - Bo-Eun Yoon
- Department of Molecular biology, Dankook University, Chungnam, Korea
| | - Sang Jeong Kim
- Department of Physiology, Seoul National University College of Medicine, Seoul, Korea. .,Department of Biomedical Science, Seoul National University College of Medicine, Seoul, Korea. .,Neuroscience Research Institute, Seoul National University College of Medicine, Seoul, Korea.
| |
Collapse
|
35
|
Abstract
Exome and targeted sequencing have revolutionized clinical diagnosis. This has been particularly striking in epilepsy and neurodevelopmental disorders, for which new genes or new variants of preexisting candidate genes are being continuously identified at increasing rates every year. A surprising finding of these efforts is the recognition that gain of function potassium channel variants are actually associated with certain types of epilepsy, such as malignant migrating partial seizures of infancy or early-onset epileptic encephalopathy. This development has been difficult to understand as traditionally potassium channel loss-of-function, not gain-of-function, has been associated with hyperexcitability disorders. In this article, we describe the current state of the field regarding the gain-of-function potassium channel variants associated with epilepsy (KCNA2, KCNB1, KCND2, KCNH1, KCNH5, KCNJ10, KCNMA1, KCNQ2, KCNQ3, and KCNT1) and speculate on the possible cellular mechanisms behind the development of seizures and epilepsy in these patients. Understanding how potassium channel gain-of-function leads to epilepsy will provide new insights into the inner working of neural circuits and aid in developing new therapies.
Collapse
Affiliation(s)
- Zachary Niday
- Dept. of Physiology and Neurobiology, University of Connecticut, Storrs, CT 06269, USA
| | | |
Collapse
|
36
|
Myers TL, Gonzalez OC, Stein JB, Bazhenov M. Characterizing Concentration-Dependent Neural Dynamics of 4-Aminopyridine-Induced Epileptiform Activity. ACTA ACUST UNITED AC 2018; 4. [PMID: 30450487 PMCID: PMC6234984 DOI: 10.4172/2472-0895.1000128] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Epilepsy remains one of the most common neurological disorders. In patients, it is characterized by unprovoked, spontaneous, and recurrent seizures or ictal events. Typically, inter-ictal events or large bouts of population level activity can be measured between seizures and are generally asymptomatic. Decades of research have focused on understanding the mechanisms leading to the development of seizure-like activity using various pro-convulsive pharmacological agents, including 4-aimnopyridine (4AP). However, the lack of consistency in the concentrations used for studying 4AP-induced epileptiform activity in animal models may give rise to differences in results and interpretation thereof. Indeed, the range of 4AP concentration in both in vivo and in vitro studies varies from 3 μM to 40 mM. Here, we explored the effects of various 4AP concentrations on the development and characteristics of hippocampal epileptiform activity in acute mouse brain slices of either sex. Using multi-electrode array recordings, we show that 4AP induces hippocampal epileptiform activity for a broad range of concentrations. The frequency component and the spatiotemporal patterns of the epileptiform activity revealed a dose-dependent response. Finally, in the presence of 4AP, reduction of KCC2 co-transporter activity by KCC2 antagonist VU0240551 prevented the manifestation of the frequency component differences between different concentrations of 4AP. Overall, the study predicts that different concentrations of 4AP can result in the different mechanisms behind hippocampal epileptiform activity, of which some are dependent on the KCC2 co-transporter function.
Collapse
Affiliation(s)
- Timothy L Myers
- Neuroscience Graduate Program, University of California, Riverside, California, United States of America.,Department of Cell Biology and Neuroscience, University of California, Riverside, California, United States of America
| | - Oscar C Gonzalez
- Department of Medicine, University of California, San Diego, California, United States of America.,Neuroscience Graduate Program, University of California, San Diego, California, United States of America
| | - Jacob B Stein
- Department of Cell Biology and Neuroscience, University of California, Riverside, California, United States of America
| | - Maxim Bazhenov
- Department of Medicine, University of California, San Diego, California, United States of America
| |
Collapse
|
37
|
Shim HG, Lee YS, Kim SJ. The Emerging Concept of Intrinsic Plasticity: Activity-dependent Modulation of Intrinsic Excitability in Cerebellar Purkinje Cells and Motor Learning. Exp Neurobiol 2018; 27:139-154. [PMID: 30022866 PMCID: PMC6050419 DOI: 10.5607/en.2018.27.3.139] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 06/28/2018] [Accepted: 06/28/2018] [Indexed: 12/15/2022] Open
Abstract
What is memory? How does the brain process the sensory information and modify an organism's behavior? Many neuroscientists have focused on the activity- and experience-dependent modifications of synaptic functions in order to solve these fundamental questions in neuroscience. Recently, the plasticity of intrinsic excitability (called intrinsic plasticity) has emerged as an important element for information processing and storage in the brain. As the cerebellar Purkinje cells are the sole output neurons in the cerebellar cortex and the information is conveyed from a neuron to its relay neurons by forms of action potential firing, the modulation of the intrinsic firing activity may play a critical role in the cerebellar learning. Many voltage-gated and/or Ca2+-activated ion channels are involved in shaping the spiking output as well as integrating synaptic inputs to finely tune the cerebellar output. Recent studies suggested that the modulation of the intrinsic excitability and its plasticity in the cerebellar Purkinje cells might function as an integrator for information processing and memory formation. Moreover, the intrinsic plasticity might also determine the strength of connectivity to the sub-cortical areas such as deep cerebellar nuclei and vestibular nuclei to trigger the consolidation of the cerebellar-dependent memory by transferring the information.
Collapse
Affiliation(s)
- Hyun Geun Shim
- Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Korea
- Department of Biomedical Science, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Yong-Seok Lee
- Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Korea
- Department of Biomedical Science, Seoul National University College of Medicine, Seoul 03080, Korea
- Neuroscience Research Institute, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Sang Jeong Kim
- Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Korea
- Department of Biomedical Science, Seoul National University College of Medicine, Seoul 03080, Korea
- Neuroscience Research Institute, Seoul National University College of Medicine, Seoul 03080, Korea
| |
Collapse
|
38
|
Cyproheptadine Regulates Pyramidal Neuron Excitability in Mouse Medial Prefrontal Cortex. Neurosci Bull 2018; 34:759-768. [PMID: 29671217 DOI: 10.1007/s12264-018-0225-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 02/12/2018] [Indexed: 10/17/2022] Open
Abstract
Cyproheptadine (CPH), a first-generation antihistamine, enhances the delayed rectifier outward K+ current (IK) in mouse cortical neurons through a sigma-1 receptor-mediated protein kinase A pathway. In this study, we aimed to determine the effects of CPH on neuronal excitability in current-clamped pyramidal neurons in mouse medial prefrontal cortex slices. CPH (10 µmol/L) significantly reduced the current density required to generate action potentials (APs) and increased the instantaneous frequency evoked by a depolarizing current. CPH also depolarized the resting membrane potential (RMP), decreased the delay time to elicit an AP, and reduced the spike threshold potential. This effect of CPH was mimicked by a sigma-1 receptor agonist and eliminated by an antagonist. Application of tetraethylammonium (TEA) to block IK channels hyperpolarized the RMP and reduced the instantaneous frequency of APs. TEA eliminated the effects of CPH on AP frequency and delay time, but had no effect on spike threshold or RMP. The current-voltage relationship showed that CPH increased the membrane depolarization in response to positive current pulses and hyperpolarization in response to negative current pulses, suggesting that other types of membrane ion channels might also be affected by CPH. These results suggest that CPH increases the excitability of medial prefrontal cortex neurons by regulating TEA-sensitive IK channels as well as other TEA-insensitive K+ channels, probably ID and inward-rectifier Kir channels. This effect of CPH may explain its apparent clinical efficacy as an antidepressant and antipsychotic.
Collapse
|
39
|
Guan D, Pathak D, Foehring RC. Functional roles of Kv1-mediated currents in genetically identified subtypes of pyramidal neurons in layer 5 of mouse somatosensory cortex. J Neurophysiol 2018; 120:394-408. [PMID: 29641306 DOI: 10.1152/jn.00691.2017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
We used voltage-clamp recordings from somatic outside-out macropatches to determine the amplitude and biophysical properties of putative Kv1-mediated currents in layer 5 pyramidal neurons (PNs) from mice expressing EGFP under the control of promoters for etv1 or glt. We then used whole cell current-clamp recordings and Kv1-specific peptide blockers to test the hypothesis that Kv1 channels differentially regulate action potential (AP) voltage threshold, repolarization rate, and width as well as rheobase and repetitive firing in these two PN types. We found that Kv1-mediated currents make up a similar percentage of whole cell K+ current in both cell types, and only minor biophysical differences were observed between PN types or between currents sensitive to different Kv1 blockers. Putative Kv1 currents contributed to AP voltage threshold in both PN types, but AP width and rate of repolarization were only affected in etv1 PNs. Kv1 currents regulate rheobase, delay to the first AP, and firing rate similarly in both cell types, but the frequency-current slope was much more sensitive to Kv1 block in etv1 PNs. In both cell types, Kv1 block shifted the current required to elicit an onset doublet of action potentials to lower currents. Spike frequency adaptation was also affected differently by Kv1 block in the two PN types. Thus, despite similar expression levels and minimal differences in biophysical properties, Kv1 channels differentially regulate APs and repetitive firing in etv1 and glt PNs. This may reflect differences in subcellular localization of channel subtypes or differences in the other K+ channels expressed. NEW & NOTEWORTHY In two types of genetically identified layer 5 pyramidal neurons, α-dendrotoxin blocked approximately all of the putative Kv1 current (on average). We used outside-out macropatches and whole cell recordings at 33°C to show that despite similar expression levels and minimal differences in biophysical properties, Kv1 channels differentially regulate action potentials and repetitive firing in etv1 and glt pyramidal neurons. This may reflect differences in subcellular localization of channel subtypes or differences in the other K+ channels expressed.
Collapse
Affiliation(s)
- Dongxu Guan
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center , Memphis, Tennessee
| | - Dhruba Pathak
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center , Memphis, Tennessee
| | - Robert C Foehring
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center , Memphis, Tennessee
| |
Collapse
|
40
|
Abstract
Axons link distant brain regions and are usually considered as simple transmission cables in which reliable propagation occurs once an action potential has been generated. Safe propagation of action potentials relies on specific ion channel expression at strategic points of the axon such as nodes of Ranvier or axonal branch points. However, while action potentials are generally considered as the quantum of neuronal information, their signaling is not entirely digital. In fact, both their shape and their conduction speed have been shown to be modulated by activity, leading to regulations of synaptic latency and synaptic strength. We report here newly identified mechanisms of (1) safe spike propagation along the axon, (2) compartmentalization of action potential shape in the axon, (3) analog modulation of spike-evoked synaptic transmission and (4) alteration in conduction time after persistent regulation of axon morphology in central neurons. We discuss the contribution of these regulations in information processing.
Collapse
Affiliation(s)
- Sylvain Rama
- UNIS, UMR_S 1072, INSERM, Aix-Marseille Université, 13015 Marseille, France; Department of Clinical and Experimental Epilepsy, Institute of Neurology, University College London, Queen Square, London WC1N 3BG, UK
| | - Mickaël Zbili
- UNIS, UMR_S 1072, INSERM, Aix-Marseille Université, 13015 Marseille, France
| | - Dominique Debanne
- UNIS, UMR_S 1072, INSERM, Aix-Marseille Université, 13015 Marseille, France.
| |
Collapse
|
41
|
Ghasemi Z, Naderi N, Shojaei A, Raoufy MR, Ahmadirad N, Mirnajafi-Zadeh J. Effect of Low-Frequency Electrical Stimulation on the High-K+-Induced Neuronal Hyperexcitability in Rat Hippocampal Slices. Neuroscience 2018; 369:87-96. [DOI: 10.1016/j.neuroscience.2017.11.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 11/04/2017] [Accepted: 11/06/2017] [Indexed: 12/23/2022]
|
42
|
Plasticity of intrinsic excitability during LTD is mediated by bidirectional changes in h-channel activity. Sci Rep 2017; 7:14418. [PMID: 29089586 PMCID: PMC5663755 DOI: 10.1038/s41598-017-14874-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 10/18/2017] [Indexed: 11/12/2022] Open
Abstract
The polarity of excitability changes associated with induction of Long-Term synaptic Depression (LTD) in CA1 pyramidal neurons is a contentious issue. Postsynaptic neuronal excitability after LTD induction is found to be reduced in certain cases (i.e. synergistic changes) but enhanced in others (i.e. compensatory or homeostatic). We examined here whether these divergent findings could result from the activation of two separate mechanisms converging onto a single learning rule linking synergistic and homeostatic plasticity. We show that the magnitude of LTD induced with low frequency stimulation (LFS) of the Schaffer collaterals determines the polarity of intrinsic changes in CA1 pyramidal neurons. Apparent input resistance (Rin) is reduced following induction of moderate LTD (<20–30%). In contrast, Rin is increased after induction of large LTD (>40%) induced by repetitive episodes of LFS. The up-regulation of Ih observed after moderate LTD results from the activation of NMDA receptors whereas the down-regulation of Ih is due to activation of mGluR1 receptors. These changes in Rin were associated with changes in intrinsic excitability. In conclusion, our study indicates that changes in excitability after LTD induction follow a learning rule describing a continuum linking synergistic and compensatory changes in excitability.
Collapse
|
43
|
Pelkey KA, Chittajallu R, Craig MT, Tricoire L, Wester JC, McBain CJ. Hippocampal GABAergic Inhibitory Interneurons. Physiol Rev 2017; 97:1619-1747. [PMID: 28954853 DOI: 10.1152/physrev.00007.2017] [Citation(s) in RCA: 538] [Impact Index Per Article: 67.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 05/16/2017] [Accepted: 05/26/2017] [Indexed: 12/11/2022] Open
Abstract
In the hippocampus GABAergic local circuit inhibitory interneurons represent only ~10-15% of the total neuronal population; however, their remarkable anatomical and physiological diversity allows them to regulate virtually all aspects of cellular and circuit function. Here we provide an overview of the current state of the field of interneuron research, focusing largely on the hippocampus. We discuss recent advances related to the various cell types, including their development and maturation, expression of subtype-specific voltage- and ligand-gated channels, and their roles in network oscillations. We also discuss recent technological advances and approaches that have permitted high-resolution, subtype-specific examination of their roles in numerous neural circuit disorders and the emerging therapeutic strategies to ameliorate such pathophysiological conditions. The ultimate goal of this review is not only to provide a touchstone for the current state of the field, but to help pave the way for future research by highlighting where gaps in our knowledge exist and how a complete appreciation of their roles will aid in future therapeutic strategies.
Collapse
Affiliation(s)
- Kenneth A Pelkey
- Porter Neuroscience Center, Eunice Kennedy-Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland; Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Hatherly Laboratories, University of Exeter, Exeter, United Kingdom; and Sorbonne Universités, UPMC University of Paris, INSERM, CNRS, Neurosciences Paris Seine-Institut de Biologie Paris Seine, Paris, France
| | - Ramesh Chittajallu
- Porter Neuroscience Center, Eunice Kennedy-Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland; Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Hatherly Laboratories, University of Exeter, Exeter, United Kingdom; and Sorbonne Universités, UPMC University of Paris, INSERM, CNRS, Neurosciences Paris Seine-Institut de Biologie Paris Seine, Paris, France
| | - Michael T Craig
- Porter Neuroscience Center, Eunice Kennedy-Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland; Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Hatherly Laboratories, University of Exeter, Exeter, United Kingdom; and Sorbonne Universités, UPMC University of Paris, INSERM, CNRS, Neurosciences Paris Seine-Institut de Biologie Paris Seine, Paris, France
| | - Ludovic Tricoire
- Porter Neuroscience Center, Eunice Kennedy-Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland; Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Hatherly Laboratories, University of Exeter, Exeter, United Kingdom; and Sorbonne Universités, UPMC University of Paris, INSERM, CNRS, Neurosciences Paris Seine-Institut de Biologie Paris Seine, Paris, France
| | - Jason C Wester
- Porter Neuroscience Center, Eunice Kennedy-Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland; Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Hatherly Laboratories, University of Exeter, Exeter, United Kingdom; and Sorbonne Universités, UPMC University of Paris, INSERM, CNRS, Neurosciences Paris Seine-Institut de Biologie Paris Seine, Paris, France
| | - Chris J McBain
- Porter Neuroscience Center, Eunice Kennedy-Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland; Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Hatherly Laboratories, University of Exeter, Exeter, United Kingdom; and Sorbonne Universités, UPMC University of Paris, INSERM, CNRS, Neurosciences Paris Seine-Institut de Biologie Paris Seine, Paris, France
| |
Collapse
|
44
|
Adrenergic Gate Release for Spike Timing-Dependent Synaptic Potentiation. Neuron 2017; 93:394-408. [PMID: 28103480 DOI: 10.1016/j.neuron.2016.12.039] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 07/08/2016] [Accepted: 12/21/2016] [Indexed: 12/11/2022]
Abstract
Spike timing-dependent synaptic plasticity (STDP) serves as a key cellular correlate of associative learning, which is facilitated by elevated attentional and emotional states involving activation of adrenergic signaling. At cellular levels, adrenergic signaling increases dendrite excitability, but the underlying mechanisms remain elusive. Here we show that activation of β2-adrenoceptors promoted STD long-term synaptic potentiation at mouse hippocampal excitatory synapses by inactivating dendritic Kv1.1-containing potassium channels, which increased dendrite excitability and facilitated dendritic propagation of postsynaptic depolarization, potentially improving coincidental activation of pre- and postsynaptic terminals. We further demonstrate that adrenergic modulation of Kv1.1 was mediated by the signaling scaffold SAP97, which, through direct protein-protein interactions, escorts β2 signaling to remove Kv1.1 from the dendrite surface. These results reveal a mechanism through which the postsynaptic signaling scaffolds bridge the aroused brain state to promote induction of synaptic plasticity and potentially to enhance spike timing and memory encoding.
Collapse
|
45
|
LGI1 tunes intrinsic excitability by regulating the density of axonal Kv1 channels. Proc Natl Acad Sci U S A 2017; 114:7719-7724. [PMID: 28673977 DOI: 10.1073/pnas.1618656114] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Autosomal dominant epilepsy with auditory features results from mutations in leucine-rich glioma-inactivated 1 (LGI1), a soluble glycoprotein secreted by neurons. Animal models of LGI1 depletion display spontaneous seizures, however, the function of LGI1 and the mechanisms by which deficiency leads to epilepsy are unknown. We investigated the effects of pure recombinant LGI1 and genetic depletion on intrinsic excitability, in the absence of synaptic input, in hippocampal CA3 neurons, a classical focus for epileptogenesis. Our data indicate that LGI1 is expressed at the axonal initial segment and regulates action potential firing by setting the density of the axonal Kv1.1 channels that underlie dendrotoxin-sensitive D-type potassium current. LGI1 deficiency incurs a >50% down-regulation of the expression of Kv1.1 and Kv1.2 via a posttranscriptional mechanism, resulting in a reduction in the capacity of axonal D-type current to limit glutamate release, thus contributing to epileptogenesis.
Collapse
|
46
|
The role of axonal Kv1 channels in CA3 pyramidal cell excitability. Sci Rep 2017; 7:315. [PMID: 28331203 PMCID: PMC5428268 DOI: 10.1038/s41598-017-00388-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 02/22/2017] [Indexed: 01/24/2023] Open
Abstract
Axonal ion channels control spike initiation and propagation along the axon and determine action potential waveform. We show here that functional suppression of axonal Kv1 channels with local puff of dendrotoxin (DTx), laser or mechanical axotomy significantly increased excitability measured in the cell body. Importantly, the functional effect of DTx puffing or axotomy was not limited to the axon initial segment but was also seen on axon collaterals. In contrast, no effects were observed when DTx was puffed on single apical dendrites or after single dendrotomy. A simple model with Kv1 located in the axon reproduced the experimental observations and showed that the distance at which the effects of axon collateral cuts are seen depends on the axon space constant. In conclusion, Kv1 channels located in the axon proper greatly participate in intrinsic excitability of CA3 pyramidal neurons. This finding stresses the importance of the axonal compartment in the regulation of intrinsic neuronal excitability.
Collapse
|
47
|
Weng W, Chen Y, Wang M, Zhuang Y, Behnisch T. Potentiation of Schaffer-Collateral CA1 Synaptic Transmission by eEF2K and p38 MAPK Mediated Mechanisms. Front Cell Neurosci 2016; 10:247. [PMID: 27826228 PMCID: PMC5078695 DOI: 10.3389/fncel.2016.00247] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 10/10/2016] [Indexed: 12/20/2022] Open
Abstract
The elongation factor 2 kinase (eEF2K), likewise known as CaMKIII, has been demonstrated to be involved in antidepressant responses of NMDA receptor antagonists. Even so, it remains open whether direct inhibition of eEF2K without altering up-stream or other signaling pathways affects hippocampal synaptic transmission and neuronal network synchrony. Inhibition of eEF2K by the selective and potent eEF2K inhibitor A-484954 induced a fast pre-synaptically mediated enhancement of synaptic transmission and synchronization of neural network activity. The eEF2K-inhibition mediated potentiation of synaptic transmission of hippocampal CA1 neurons is most notably independent of protein synthesis and does not rely on protein kinase C, protein kinase A or mitogen-activated protein kinase (MAPK)/extracellular signal-regulated protein kinase 1/2. Moreover, the strengthening of synaptic transmission in the response to the inhibition of eEF2K was strongly attenuated by the inhibition of p38 MAPK. In addition, we show the involvement of barium-sensitive and more specific the TWIK-related potassium-1 (TREK-1) channels in the eEF2K-inhibition mediated potentiation of synaptic transmission. These findings reveal a novel pathway of eEF2K mediated regulation of hippocampal synaptic transmission. Further research is required to study whether such compounds could be beneficial for the development of mood disorder treatments with a fast-acting antidepressant response.
Collapse
Affiliation(s)
- Weiguang Weng
- The Institutes of Brain Science, The State Key Laboratory of Medical Neurobiology, and the Collaborative Innovation Center for Brain Science, Fudan University Shanghai, China
| | - Ying Chen
- The Institutes of Brain Science, The State Key Laboratory of Medical Neurobiology, and the Collaborative Innovation Center for Brain Science, Fudan University Shanghai, China
| | - Man Wang
- The Institutes of Brain Science, The State Key Laboratory of Medical Neurobiology, and the Collaborative Innovation Center for Brain Science, Fudan University Shanghai, China
| | - Yinghan Zhuang
- The Institutes of Brain Science, The State Key Laboratory of Medical Neurobiology, and the Collaborative Innovation Center for Brain Science, Fudan University Shanghai, China
| | - Thomas Behnisch
- The Institutes of Brain Science, The State Key Laboratory of Medical Neurobiology, and the Collaborative Innovation Center for Brain Science, Fudan University Shanghai, China
| |
Collapse
|
48
|
|
49
|
Gasselin C, Inglebert Y, Debanne D. Homeostatic regulation of h-conductance controls intrinsic excitability and stabilizes the threshold for synaptic modification in CA1 neurons. J Physiol 2015; 593:4855-69. [PMID: 26316265 DOI: 10.1113/jp271369] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 08/15/2015] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS We determined the contribution of the hyperpolarization-activated cationic (h) current (Ih ) to the homeostatic regulation of CA1 pyramidal cells in vitro using chronic treatments (48 h) that either increase (picrotoxin) or decrease (kynurenate) neuronal activity. The h-conductance was found to be up- or down-regulated following chronic activity enhancement or activity deprivation, respectively. This bidirectional plasticity of Ih was found to subsequently alter both apparent input resistance and intrinsic neuronal excitability. Bidirectional homeostatic plasticity of Ih also determined EPSP waveform and EPSP summation tested at 5-30 Hz. Long-term synaptic modification induced by repetitive stimulation of the Schaffer collaterals was found to be constant across treatments in the presence of Ih but not when Ih was blocked pharmacologically. Thus, bidirectional homeostatic regulation of Ih stabilizes induction of long-term synaptic modification in CA1 pyramidal neurons that depends on EPSP summation. ABSTRACT The hyperpolarization-activated cationic (h) current is a voltage-shock absorber, highly expressed in the dendrites of CA1 pyramidal neurons. Up-regulation of Ih has been reported following episodes of intense network activity but the effect of activity deprivation on Ih and the functional consequence of homeostatic regulation of Ih remain unclear. We determined here the contribution of Ih to the homeostatic regulation of CA1 pyramidal cell excitability. Intrinsic neuronal excitability was decreased in neurons treated for 2-3 days with the GABAA channel blocker picrotoxin (PiTx) but increased in neurons treated (2-3 days) with the glutamate receptor antagonist kynurenate (Kyn). Membrane capacitance remained unchanged after treatment but the apparent input resistance was reduced for PiTx-treated neurons and enhanced for Kyn-treated neurons. Maximal Ih conductance was up-regulated after chronic hyperactivity but down-regulated following chronic hypoactivity. Up-regulation of Ih in PiTx-treated cultures was found to accelerate EPSP kinetics and reduce temporal summation of EPSPs whereas opposite effects were observed in Kyn-treated cultures, indicating that homeostatic regulation of Ih may control the induction of synaptic modification depending on EPSP summation. In fact, stimulation of the Schaffer collaterals at 3-10 Hz induced differential levels of plasticity in PiTx-treated and Kyn-treated neurons when Ih was blocked pharmacologically but not in control conditions. These data indicate that homeostatic regulation of Ih normalizes the threshold for long-term synaptic modification that depends on EPSP summation. In conclusion, bidirectional homeostatic regulation of Ih not only controls spiking activity but also stabilizes the threshold for long-term potentiation induced in CA1 pyramidal neurons by repetitive stimulation.
Collapse
Affiliation(s)
- Célia Gasselin
- INSERM, U-1072, Marseille, France.,Aix-Marseille University, Marseille, France.,Neurobiology of ion channels (UNIS), Marseille, France
| | - Yanis Inglebert
- INSERM, U-1072, Marseille, France.,Aix-Marseille University, Marseille, France.,Neurobiology of ion channels (UNIS), Marseille, France
| | - Dominique Debanne
- INSERM, U-1072, Marseille, France.,Aix-Marseille University, Marseille, France.,Neurobiology of ion channels (UNIS), Marseille, France
| |
Collapse
|
50
|
Sokolova IV, Schneider CJ, Bezaire M, Soltesz I, Vlkolinsky R, Nelson GA. Proton Radiation Alters Intrinsic and Synaptic Properties of CA1 Pyramidal Neurons of the Mouse Hippocampus. Radiat Res 2015; 183:208-18. [DOI: 10.1667/rr13785.1] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Affiliation(s)
- Irina V. Sokolova
- Department of Basic Sciences, Division of Radiation Research, School of Medicine, Loma Linda University, Loma Linda, California
| | - Calvin J. Schneider
- Department of Anatomy and Neurobiology, University of California, Irvine, Irvine, California
| | - Marianne Bezaire
- Department of Anatomy and Neurobiology, University of California, Irvine, Irvine, California
| | - Ivan Soltesz
- Department of Anatomy and Neurobiology, University of California, Irvine, Irvine, California
| | - Roman Vlkolinsky
- Department of Basic Sciences, Division of Radiation Research, School of Medicine, Loma Linda University, Loma Linda, California
| | - Gregory A. Nelson
- Department of Basic Sciences, Division of Radiation Research, School of Medicine, Loma Linda University, Loma Linda, California
| |
Collapse
|