1
|
Connolly JG, Plant LD. SUMO Regulation of Ion Channels in Health and Disease. Physiology (Bethesda) 2025; 40:0. [PMID: 39499247 DOI: 10.1152/physiol.00034.2024] [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: 07/15/2024] [Revised: 10/10/2024] [Accepted: 10/30/2024] [Indexed: 11/07/2024] Open
Abstract
The small ubiquitin-like modifier (SUMO) protein pathway governs a panoply of vital biological processes including cell death, proliferation, differentiation, metabolism, and signal transduction by diversifying the functions, half-lives, and partnerships of target proteins in situ. More recently, SUMOylation has emerged as a key regulator of ion homeostasis and excitability across multiple tissues due to the regulation of a plethora of ion channels expressed in a range of tissue subtypes. Altogether, the balance of SUMOylation states among relevant ion channels can result in graded biophysical effects that tune excitability and contribute to a range of disease states including cardiac arrhythmia, epilepsy, pain transmission, and inflammation. Here, we consolidate these concepts by focusing on the role of ion channel SUMOylation in the central nervous system, peripheral nervous system, and cardiovascular system. In addition, we review what is known about the enigmatic factors that regulate the SUMO pathway and consider the emerging role of small molecule SUMO modulators as potential therapeutics in a range of diseases.
Collapse
Affiliation(s)
- Jenna G Connolly
- Department of Pharmaceutical Sciences and the Center for Drug Discovery, The School of Pharmacy and Pharmaceutical SciencesBouvé College of Health Sciences, Northeastern University, Boston, Massachusetts, United States
| | - Leigh D Plant
- Department of Pharmaceutical Sciences and the Center for Drug Discovery, The School of Pharmacy and Pharmaceutical SciencesBouvé College of Health Sciences, Northeastern University, Boston, Massachusetts, United States
| |
Collapse
|
2
|
Lin B, Li F, Hui J, Xing Z, Fu J, Li S, Shi H, Liu C, Mao H, Wu Z. Modular Reconfigurable Approach Toward Noninvasive Wearable Body Net for Monitoring Sweat and Physiological Signals. ACS Sens 2025; 10:225-235. [PMID: 39576944 DOI: 10.1021/acssensors.4c02141] [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: 11/24/2024]
Abstract
In the realm of wearable technology, strategically placing sensors at various body locations enhances the detection of diverse physiological indicators crucial for remote medical care. However, current devices often focus on a single body part for specific physical parameters, which hinders the seamless integration of sensors across multiple body parts and necessitates redesign for new detection capabilities. Here, we propose a modular, reconfigurable circuit assembly method that can be adaptable for multiple body locations to construct the body net. By simply reassembling different child modules with the base module using flexible printed circuit board connectors, we can efficiently detect various parameters including sweat ion indicators, electrocardiogram signals, electromyography signals, motion data, heart rate, blood oxygen saturation, and skin temperature. These data can be transmitted to a mobile phone app via a Bluetooth Low Energy protocol for further evaluation. Comparative evaluations against established commercial devices substantiate the viability of our sensor technology. In addition, results from wearable body network detections using reconfigurable sensors across multiple body parts of volunteers also indicate promising application prospects, demonstrating the extensive potential for regular health monitoring and clinical applications.
Collapse
Affiliation(s)
- Bo Lin
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai 200050, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Fangqi Li
- Institute of Microelectronics of the Chinese Academy of Science, Beijing 100029, China
| | - Jianan Hui
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai 200050, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhe Xing
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai 200050, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jie Fu
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Shuang Li
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Haotian Shi
- China Three Gorges Renewables (Group) Company Limited, Harbin 150000, China
| | - Chaoran Liu
- Ministry of Education Engineering Research Center of Smart Microsensors and Microsystems, College of Electronics and Information, Hangzhou Danzi University, Hangzhou 310018, China
| | - Hongju Mao
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai 200050, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhenhua Wu
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai 200050, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
3
|
Tuna T, Banks T, Glickert G, Sevinc C, Nair SS, Unal G. Basal forebrain innervation of the amygdala: an anatomical and computational exploration. Brain Struct Funct 2025; 230:30. [PMID: 39805973 PMCID: PMC11729089 DOI: 10.1007/s00429-024-02886-1] [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: 02/21/2024] [Accepted: 12/05/2024] [Indexed: 01/16/2025]
Abstract
Theta oscillations of the mammalian amygdala are associated with processing, encoding and retrieval of aversive memories. In the hippocampus, the power of the network theta oscillation is modulated by basal forebrain (BF) GABAergic projections. Here, we combine anatomical and computational approaches to investigate if similar BF projections to the amygdaloid complex provide an analogous modulation of local network activity. We used retrograde tracing with fluorescent immunohistochemistry to identify cholinergic and non-cholinergic parvalbumin- or calbindin-immunoreactive BF neuronal subgroups targeting the input (lateral and basolateral nuclei) and output (central nucleus and the central bed nucleus of the stria terminalis) regions of the amygdaloid complex. We observed a dense non-cholinergic, putative GABAergic projection from the ventral pallidum (VP) and the substantia innominata (SI) to the basolateral amygdala (BLA). The VP/SI axonal projections to the BLA were confirmed using viral anterograde tracing and transsynaptic labeling. We tested the potential function of this VP/SI-BLA pathway in a 1000-cell biophysically realistic network model, which incorporated principal neurons and three major interneuron groups of the BLA, together with extrinsic glutamatergic, cholinergic, and VP/SI GABAergic inputs. We observed in silico that theta-modulation of VP/SI GABAergic projections enhanced theta oscillations in the BLA via their selective innervation of the parvalbumin-expressing local interneurons. Ablation of parvalbumin-, but not somatostatin- or calretinin-expressing, interneurons reduced theta power in the BLA model. These results suggest that long-range BF GABAergic projections may modulate network activity at their target regions through the formation of a common interneuron-type and oscillatory phase-specific disinhibitory motif.
Collapse
Affiliation(s)
- Tuğçe Tuna
- Behavioral Neuroscience Laboratory, Department of Psychology, Boğaziçi University, Bebek, 34342, Istanbul, Turkey
| | - Tyler Banks
- Neural Engineering Laboratory, Department of Electrical Engineering and Computer Science, University of Missouri, Columbia, MO, 65211, USA
| | - Gregory Glickert
- Neural Engineering Laboratory, Department of Electrical Engineering and Computer Science, University of Missouri, Columbia, MO, 65211, USA
| | - Cem Sevinc
- Behavioral Neuroscience Laboratory, Department of Psychology, Boğaziçi University, Bebek, 34342, Istanbul, Turkey
| | - Satish S Nair
- Neural Engineering Laboratory, Department of Electrical Engineering and Computer Science, University of Missouri, Columbia, MO, 65211, USA
| | - Gunes Unal
- Behavioral Neuroscience Laboratory, Department of Psychology, Boğaziçi University, Bebek, 34342, Istanbul, Turkey.
| |
Collapse
|
4
|
Espino CM, Nagaraja C, Ortiz S, Dayton JR, Murali AR, Ma Y, Mann EL, Garlapalli S, Wohlgemuth RP, Brashear SE, Smith LR, Wilkinson KA, Griffith TN. Differential encoding of mammalian proprioception by voltage-gated sodium channels. SCIENCE ADVANCES 2025; 11:eads6660. [PMID: 39772670 PMCID: PMC11708877 DOI: 10.1126/sciadv.ads6660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 12/02/2024] [Indexed: 01/11/2025]
Abstract
Animals requiring purposeful movement for survival are endowed with mechanoreceptors, called proprioceptors, that provide essential sensory feedback from muscles and joints to spinal cord circuits, which modulates motor output. Despite the essential nature of proprioceptive signaling in daily life, the mechanisms governing proprioceptor activity are poorly understood. Here, we identified nonredundant roles for two voltage-gated sodium channels (NaVs), NaV1.1 and NaV1.6, in mammalian proprioception. Deletion of NaV1.6 in somatosensory neurons (NaV1.6cKO mice) causes severe motor deficits accompanied by loss of proprioceptive transmission, which contrasts with our previous findings using similar mouse models to target NaV1.1 (NaV1.1cKO). In NaV1.6cKO animals, we observed impairments in proprioceptor end-organ structure and a marked reduction in skeletal muscle myofiber size that were absent in NaV1.1cKO mice. We attribute the differential contributions of NaV1.1 and NaV1.6 to distinct cellular localization patterns. Collectively, we provide evidence that NaVs uniquely shape neural signaling within a somatosensory modality.
Collapse
Affiliation(s)
- Cyrrus M. Espino
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA, USA
| | - Chetan Nagaraja
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA, USA
| | - Serena Ortiz
- Department of Biological Sciences, San José State University, San Jose, CA, USA
| | - Jacquelyn R. Dayton
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA, USA
| | - Akash R. Murali
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA, USA
- Undergraduate Program in Neurobiology, Physiology and Behavior, University of California, Davis, Davis, CA, USA
| | - Yanki Ma
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA, USA
- Undergraduate Program in Neurobiology, Physiology and Behavior, University of California, Davis, Davis, CA, USA
| | - Emari L. Mann
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA, USA
- Postbaccalaureate Research Education Program at UC Davis, University of California, Davis, Davis, CA, USA
| | - Snigdha Garlapalli
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA, USA
- Undergraduate Program in Psychology, University of California, Davis, Davis, CA, USA
| | - Ross P. Wohlgemuth
- Department of Physiology, Neurobiology, and Behavior, University of California, Davis, Davis, CA, USA
| | - Sarah E. Brashear
- Department of Physiology, Neurobiology, and Behavior, University of California, Davis, Davis, CA, USA
| | - Lucas R. Smith
- Department of Physiology, Neurobiology, and Behavior, University of California, Davis, Davis, CA, USA
| | | | - Theanne N. Griffith
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA, USA
| |
Collapse
|
5
|
Baumgartner TJ, Dvorak NM, Goode NA, Haghighijoo Z, Marosi M, Singh J, Singh AK, Laezza F. Axin-binding domain of glycogen synthase kinase 3β facilitates functional interactions with voltage-gated Na+ channel Nav1.6. J Biol Chem 2025:108162. [PMID: 39793889 DOI: 10.1016/j.jbc.2025.108162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 12/09/2024] [Accepted: 01/02/2025] [Indexed: 01/13/2025] Open
Abstract
Voltage-gated Na+ (Nav) channels are the primary determinants of the action potential in excitable cells. Nav channels rely on a wide and diverse array of intracellular protein-protein interactions (PPIs) to achieve their full function. Glycogen synthase kinase 3 β (GSK3β) has been previously identified as a modulator of Nav1.6-encoded currents and neuronal excitability through PPI formation with Nav1.6 and phosphorylation of its C-terminal domain (CTD). Here, we hypothesized that GSK3β functions as a scaffold in a regulatory PPI complex with Nav1.6 CTD. Mutagenesis screening using the split-luciferase complementation assay indicated that the axin-binding domain (ABD) of GSK3β (262-299) is necessary for complex formation between the Nav1.6 CTD and GSK3β, and that residues within this domain are drivers of GSK3β-mediated regulation of the channel. Overexpression of an ABD-GFP fusion construct in HEK293 cells stably expressing Nav1.6 significantly reduced Nav1.6 nanocluster density compared to GFP alone. In addition, overexpression of the ABD-GFP fusion construct ablates GSK3β-mediated potentiation of Nav1.6 encoded currents and alters channel kinetics. Finally, in vivo AAV-mediated overexpression of the ABD-GFP construct in the CA1 hippocampal region induced a reduction in maximal action potential firing and an increase in action potential current threshold in a manner resembling previously reported effects of GSK3β silencing in neurons. Taken together, these results not only suggest that GSK3β-mediated regulation of Nav1.6 extends beyond transient phosphorylation, but also implicates the ABD as a critical regulatory domain that facilitates GSK3β's functional effects on Nav1.6 and neuronal excitability.
Collapse
Affiliation(s)
- T J Baumgartner
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555
| | - N M Dvorak
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555
| | - N A Goode
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555
| | - Z Haghighijoo
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555
| | - M Marosi
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555
| | - J Singh
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555
| | - A K Singh
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555
| | - F Laezza
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555.
| |
Collapse
|
6
|
Han Y, Hacker D, Donders BC, Parperis C, Thuenauer R, Leterrier C, Grünewald K, Mikhaylova M. Unveiling the cell biology of hippocampal neurons with dendritic axon origin. J Cell Biol 2025; 224:e202403141. [PMID: 39495320 PMCID: PMC11536041 DOI: 10.1083/jcb.202403141] [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: 03/26/2024] [Revised: 08/01/2024] [Accepted: 09/23/2024] [Indexed: 11/05/2024] Open
Abstract
In mammalian axon-carrying-dendrite (AcD) neurons, the axon emanates from a basal dendrite, instead of the soma, to create a privileged route for action potential generation at the axon initial segment (AIS). However, it is unclear how such unusual morphology is established and whether the structure and function of the AIS in AcD neurons are preserved. By using dissociated hippocampal cultures as a model, we show that the development of AcD morphology can occur prior to synaptogenesis and independently of the in vivo environment. A single precursor neurite first gives rise to the axon and then to the AcD. The AIS possesses a similar cytoskeletal architecture as the soma-derived AIS and similarly functions as a trafficking barrier to retain axon-specific molecular composition. However, it does not undergo homeostatic plasticity, contains lesser cisternal organelles, and receives fewer inhibitory inputs. Our findings reveal insights into AcD neuron biology and underscore AIS structural differences based on axon onset.
Collapse
Affiliation(s)
- Yuhao Han
- AG Optobiology, Institute of Biology, Humboldt Universität zu Berlin, Berlin, Germany
- AG “Neuronal Protein Transport”, Centre for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Centre for Structural Systems Biology, Hamburg, Germany
- Structural Cell Biology of Viruses, Leibniz Institute of Virology (LIV), Hamburg, Germany
| | - Daniela Hacker
- AG Optobiology, Institute of Biology, Humboldt Universität zu Berlin, Berlin, Germany
| | | | | | - Roland Thuenauer
- Advanced Light and Fluorescence Microscopy (ALFM) Facility, Centre for Structural Systems Biology, Hamburg, Germany
- Technology Platform Light Microscopy, University of Hamburg, Hamburg, Germany
- Technology Platform Microscopy and Image Analysis (TP MIA), Leibniz Institute of Virology (LIV), Hamburg, Germany
| | | | - Kay Grünewald
- Centre for Structural Systems Biology, Hamburg, Germany
- Structural Cell Biology of Viruses, Leibniz Institute of Virology (LIV), Hamburg, Germany
- Department of Chemistry, University of Hamburg, Hamburg, Germany
| | - Marina Mikhaylova
- AG Optobiology, Institute of Biology, Humboldt Universität zu Berlin, Berlin, Germany
- AG “Neuronal Protein Transport”, Centre for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
7
|
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
|
8
|
Berling D, Baroni L, Chaffiol A, Gauvain G, Picaud S, Antolík J. Optogenetic Stimulation Recruits Cortical Neurons in a Morphology-Dependent Manner. J Neurosci 2024; 44:e1215242024. [PMID: 39424369 PMCID: PMC11622177 DOI: 10.1523/jneurosci.1215-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 09/20/2024] [Accepted: 09/22/2024] [Indexed: 10/21/2024] Open
Abstract
Single-photon optogenetics enables precise, cell-type-specific modulation of neuronal circuits, making it a crucial tool in neuroscience. Its miniaturization in the form of fully implantable wide-field stimulator arrays enables long-term interrogation of cortical circuits and bears promise for brain-machine interfaces for sensory and motor function restoration. However, achieving selective activation of functional cortical representations poses a challenge, as studies show that targeted optogenetic stimulation results in activity spread beyond one functional domain. While recurrent network mechanisms contribute to activity spread, here we demonstrate with detailed simulations of isolated pyramidal neurons from cats of unknown sex that already neuron morphology causes a complex spread of optogenetic activity at the scale of one cortical column. Since the shape of a neuron impacts its optogenetic response, we find that a single stimulator at the cortical surface recruits a complex spatial distribution of neurons that can be inhomogeneous and vary with stimulation intensity and neuronal morphology across layers. We explore strategies to enhance stimulation precision, finding that optimizing stimulator optics may offer more significant improvements than the preferentially somatic expression of the opsin through genetic targeting. Our results indicate that, with the right optical setup, single-photon optogenetics can precisely activate isolated neurons at the scale of functional cortical domains spanning several hundred micrometers.
Collapse
Affiliation(s)
- David Berling
- Faculty of Mathematics and Physics, Charles University, Prague 118 00, Czechia
| | - Luca Baroni
- Faculty of Mathematics and Physics, Charles University, Prague 118 00, Czechia
| | | | - Gregory Gauvain
- Institut de la Vision, Sorbonne Université, Paris 75012, France
| | - Serge Picaud
- Institut de la Vision, Sorbonne Université, Paris 75012, France
| | - Ján Antolík
- Faculty of Mathematics and Physics, Charles University, Prague 118 00, Czechia
| |
Collapse
|
9
|
Senko K, Saddoris KL, Baus E, Soe K, Vaughn SE. Catatonia responsive to corticosteroids in a patient with an SCN2A variant. AMERICAN JOURNAL OF MEDICAL GENETICS. PART C, SEMINARS IN MEDICAL GENETICS 2024; 196:e32101. [PMID: 39056510 DOI: 10.1002/ajmg.c.32101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/07/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024]
Abstract
Variants in SCN2A are a known risk factor for developing autism spectrum disorder (ASD). Catatonia is a complex neuropsychiatric syndrome, which occurs at a higher rate in individuals with ASD. Catatonia has also been associated with COVID-19 infection, though the majority of these cases are associated with increased serum inflammatory markers. We present a case of a 15-year-old female with ASD and corticosteroid responsive stuporous catatonia to explore the relationship between SCN2A variants, ASD, COVID-19 exposure, and treatment refractory catatonia. Despite a lack of significantly elevated serum or CSF inflammatory markers, this patient showed significant improvement following initiation of corticosteroid therapy. This case presents a novel approach to the work-up and treatment of catatonia in individuals with SCN2A variants independent of elevated inflammatory markers.
Collapse
Affiliation(s)
- Kimberly Senko
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Division of Child & Adolescent Psychiatry, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Division of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Kelsey L Saddoris
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Division of Child & Adolescent Psychiatry, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Division of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Ella Baus
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Division of Child & Adolescent Psychiatry, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Division of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Katherine Soe
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Division of Child & Adolescent Psychiatry, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Division of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Samuel E Vaughn
- Division of Child & Adolescent Psychiatry, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Division of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| |
Collapse
|
10
|
Rusina E, Simonti M, Duprat F, Cestèle S, Mantegazza M. Voltage-gated sodium channels in genetic epilepsy: up and down of excitability. J Neurochem 2024; 168:3872-3890. [PMID: 37654020 PMCID: PMC11591406 DOI: 10.1111/jnc.15947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 08/11/2023] [Accepted: 08/13/2023] [Indexed: 09/02/2023]
Abstract
The past two decades have witnessed a wide range of studies investigating genetic variants of voltage-gated sodium (NaV) channels, which are involved in a broad spectrum of diseases, including several types of epilepsy. We have reviewed here phenotypes and pathological mechanisms of genetic epilepsies caused by variants in NaV α and β subunits, as well as of some relevant interacting proteins (FGF12/FHF1, PRRT2, and Ankyrin-G). Notably, variants of all these genes can induce either gain- or loss-of-function of NaV leading to either neuronal hyperexcitability or hypoexcitability. We present the results of functional studies obtained with different experimental models, highlighting that they should be interpreted considering the features of the experimental system used. These systems are models, but they have allowed us to better understand pathophysiological issues, ameliorate diagnostics, orientate genetic counseling, and select/develop therapies within a precision medicine framework. These studies have also allowed us to gain insights into the physiological roles of different NaV channels and of the cells that express them. Overall, our review shows the progress that has been made, but also the need for further studies on aspects that have not yet been clarified. Finally, we conclude by highlighting some significant themes of general interest that can be gleaned from the results of the work of the last two decades.
Collapse
Affiliation(s)
- Evgeniia Rusina
- University Cote d'AzurValbonne‐Sophia AntipolisFrance
- CNRS UMR 7275Institute of Molecular and Cellular Pharmacology (IPMC)Valbonne‐Sophia AntipolisFrance
| | - Martina Simonti
- University Cote d'AzurValbonne‐Sophia AntipolisFrance
- CNRS UMR 7275Institute of Molecular and Cellular Pharmacology (IPMC)Valbonne‐Sophia AntipolisFrance
| | - Fabrice Duprat
- University Cote d'AzurValbonne‐Sophia AntipolisFrance
- CNRS UMR 7275Institute of Molecular and Cellular Pharmacology (IPMC)Valbonne‐Sophia AntipolisFrance
- InsermValbonne‐Sophia AntipolisFrance
| | - Sandrine Cestèle
- University Cote d'AzurValbonne‐Sophia AntipolisFrance
- CNRS UMR 7275Institute of Molecular and Cellular Pharmacology (IPMC)Valbonne‐Sophia AntipolisFrance
| | - Massimo Mantegazza
- University Cote d'AzurValbonne‐Sophia AntipolisFrance
- CNRS UMR 7275Institute of Molecular and Cellular Pharmacology (IPMC)Valbonne‐Sophia AntipolisFrance
- InsermValbonne‐Sophia AntipolisFrance
| |
Collapse
|
11
|
Jia L, Li M, Pachernegg S, Sedo A, Jancovski N, Burbano LE, Dalby K, Nemiroff A, Reid C, Maljevic S, Petrou S. Variant-specific in vitro neuronal network phenotypes and drug sensitivity in SCN2A developmental and epileptic encephalopathy. J Neurochem 2024; 168:3950-3961. [PMID: 38544375 DOI: 10.1111/jnc.16103] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 03/06/2024] [Accepted: 03/08/2024] [Indexed: 11/27/2024]
Abstract
De novo variants in the NaV1.2 voltage-gated sodium channel gene SCN2A are among the major causes of developmental and epileptic encephalopathies (DEE). Based on their biophysical impact on channel conductance and gating, SCN2A DEE variants can be classified into gain-of-function (GoF) or loss-of-function (LoF). Clinical and functional data have linked early seizure onset DEE to the GoF SCN2A variants, whereas late seizure onset DEE is associated with the loss of SCN2A function. This study aims to assess the impact of GoF and LoF SCN2A variants on cultured neuronal network activity and explore their modulation by selected antiseizure medications (ASM). To this end, primary cortical cultures were generated from two knock-in mouse lines carrying variants corresponding to human GoF SCN2A p.R1882Q and LoF p.R853Q DEE variant. In vitro neuronal network activity and responses to ASM were analyzed using multielectrode array (MEA) between 2 and 4 weeks in culture. The SCN2A p.R1882Q neuronal cultures showed significantly greater mean firing and burst firing. Their network synchronicity was also higher. In contrast, the SCN2A p.R853Q cultures showed lower mean firing rate, and burst firing events were less frequent. The network synchronicity was also lower. Phenytoin and levetiracetam reduced the excitability of GoF cultures, while retigabine showed differential and potentially beneficial effects on cultures with both GoF and LoF variants. We conclude that in vitro neuronal networks harboring SCN2A GoF or LoF DEE variants present with distinctive phenotypes and responses to ASM.
Collapse
Affiliation(s)
- Linghan Jia
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Melody Li
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Svenja Pachernegg
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Alicia Sedo
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Nikola Jancovski
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Lisseth Estefania Burbano
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Kelley Dalby
- RogCon Biosciences, San Diego, California, USA
- Praxis Precision Medicines, Boston, Massachusetts, USA
| | - Alex Nemiroff
- RogCon Biosciences, San Diego, California, USA
- Praxis Precision Medicines, Boston, Massachusetts, USA
| | - Christopher Reid
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Snezana Maljevic
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Steven Petrou
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
- RogCon Biosciences, San Diego, California, USA
- Praxis Precision Medicines, Boston, Massachusetts, USA
| |
Collapse
|
12
|
Qi Y, Zhao R, Tian J, Lu J, He M, Tai Y. Specific and Plastic: Chandelier Cell-to-Axon Initial Segment Connections in Shaping Functional Cortical Network. Neurosci Bull 2024; 40:1774-1788. [PMID: 39080101 PMCID: PMC11607270 DOI: 10.1007/s12264-024-01266-3] [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: 01/12/2024] [Accepted: 03/19/2024] [Indexed: 11/30/2024] Open
Abstract
Axon initial segment (AIS) is the most excitable subcellular domain of a neuron for action potential initiation. AISs of cortical projection neurons (PNs) receive GABAergic synaptic inputs primarily from chandelier cells (ChCs), which are believed to regulate action potential generation and modulate neuronal excitability. As individual ChCs often innervate hundreds of PNs, they may alter the activity of PN ensembles and even impact the entire neural network. During postnatal development or in response to changes in network activity, the AISs and axo-axonic synapses undergo dynamic structural and functional changes that underlie the wiring, refinement, and adaptation of cortical microcircuits. Here we briefly introduce the history of ChCs and review recent research advances employing modern genetic and molecular tools. Special attention will be attributed to the plasticity of the AIS and the ChC-PN connections, which play a pivotal role in shaping the dynamic network under both physiological and pathological conditions.
Collapse
Affiliation(s)
- Yanqing Qi
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Neurobiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Rui Zhao
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Neurobiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Jifeng Tian
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Neurobiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Jiangteng Lu
- Songjiang Research Institute, Shanghai Songjiang District Central Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China
- Center for Brain Science of Shanghai Children's Medical Center, Department of Anatomy and Physiology, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Miao He
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Neurobiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| | - Yilin Tai
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Neurobiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
13
|
Li M, Jin Y, Wu J, Zhao M, Yu K, Yu H. Arbidol, an antiviral drug, identified as a sodium channel blocker with anticonvulsant activity. Br J Pharmacol 2024; 181:4311-4327. [PMID: 38982721 DOI: 10.1111/bph.16496] [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: 10/06/2023] [Revised: 05/24/2024] [Accepted: 05/30/2024] [Indexed: 07/11/2024] Open
Abstract
BACKGROUND AND PURPOSE Sodium channel blockers (SCBs) have traditionally been utilized as anti-seizure medications by primarily targeting the inactivation process. In a drug discovery project aiming at finding potential anticonvulsants, we have identified arbidol, originally an antiviral drug, as a potent SCB. In order to evaluate its anticonvulsant potential, we have thoroughly examined its biophysical properties as well as its effects on animal seizure models. EXPERIMENTAL APPROACH Patch clamp recording was used to investigate the electrophysiological properties of arbidol, as well as the binding and unbinding kinetics of arbidol, carbamazepine and lacosamide. Furthermore, we evaluated the anticonvulsant effects of arbidol using three different seizure models in male mice. KEY RESULTS Arbidol effectively suppressed neuronal epileptiform activity by blocking sodium channels. Arbidol demonstrated a distinct mode of action by interacting with both the fast and slow inactivation of Nav1.2 channels compared with carbamazepine and lacosamide. A kinetic study suggested that the binding and unbinding rates might be associated with the specific characteristics of these three drugs. Arbidol targeted the classical binding site of local anaesthetics, effectively inhibited the gain-of-function effects of Nav1.2 epileptic mutations and exhibited varying degrees of anticonvulsant effects in the maximal electroshock model and subcutaneous pentylenetetrazol model but had no effect in the pilocarpine-induced status epilepticus model. CONCLUSIONS AND IMPLICATIONS Arbidol shows promising potential as an anticonvulsant agent, providing a unique mode of action that sets it apart from existing SCBs.
Collapse
Affiliation(s)
- Min Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Yuchen Jin
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Jun Wu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Miao Zhao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Kexin Yu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Haibo Yu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| |
Collapse
|
14
|
Smeralda CL, Pandit S, Turrini S, Reilly J, Palmisano A, Sprugnoli G, Hampel H, Benussi A, Borroni B, Press D, Rotenberg A, El Fakhri G, Koch G, Rossi S, Santarnecchi E. The role of parvalbumin interneuron dysfunction across neurodegenerative dementias. Ageing Res Rev 2024; 101:102509. [PMID: 39306248 DOI: 10.1016/j.arr.2024.102509] [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: 07/05/2024] [Revised: 09/15/2024] [Accepted: 09/15/2024] [Indexed: 10/04/2024]
Abstract
Parvalbumin-positive (PV+) basket neurons are fast-spiking, non-adapting inhibitory interneurons whose oscillatory activity is essential for regulating cortical excitation/inhibition balance. Their dysfunction results in cortical hyperexcitability and gamma rhythm disruption, which have recently gained substantial traction as contributing factors as well as potential therapeutic targets for the treatment of Alzheimer's Disease (AD). Recent evidence indicates that PV+ cells are also impaired in Frontotemporal Dementia (FTD) and Dementia with Lewy bodies (DLB). However, no attempt has been made to integrate these findings into a coherent pathophysiological framework addressing the contribution of PV+ interneuron dysfunction to the generation of cortical hyperexcitability and gamma rhythm disruption in FTD and DLB. To fill this gap, we epitomized the most recent evidence on PV+ interneuron impairment in AD, FTD, and DLB, focusing on its contribution to the generation of cortical hyperexcitability and gamma oscillatory disruption and their interplay with misfolded protein accumulation, neuronal death, and clinical symptoms' onset. Our work deepens the current understanding concerning the role of PV+ interneuron dysfunction across neurodegenerative dementias, highlighting commonalities and differences among AD, FTD, and DLB, thus paving the way for identifying novel biomarkers and potential therapeutic targets for the treatment of these diseases.
Collapse
Affiliation(s)
- Carmelo Luca Smeralda
- Precision Neuroscience & Neuromodulation Program, Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Siena Brain Investigation & Neuromodulation Lab (Si-BIN Lab), Unit of Neurology and Clinical Neurophysiology, Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Siddhartha Pandit
- Precision Neuroscience & Neuromodulation Program, Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Sonia Turrini
- Precision Neuroscience & Neuromodulation Program, Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Centro Studi e Ricerche in Neuroscienze Cognitive, Dipartimento di Psicologia, University of Bologna, Italy
| | - Julianne Reilly
- Precision Neuroscience & Neuromodulation Program, Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Annalisa Palmisano
- Chair of Lifespan Developmental Neuroscience, TUD Dresden University of Technology, Dresden, Germany
| | - Giulia Sprugnoli
- Precision Neuroscience & Neuromodulation Program, Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Harald Hampel
- Sorbonne University, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France
| | - Alberto Benussi
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy; Neurology Unit, Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy
| | - Barbara Borroni
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Daniel Press
- Cognitive Neurology Unit, Beth Israel Deaconess Medical Center, 330 Brookline Avenue, Brookline, MA, USA
| | - Alexander Rotenberg
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Georges El Fakhri
- Gordon Center for Medical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Giacomo Koch
- Human Physiology Unit, Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy; Experimental Neuropsychophysiology Laboratory, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Simone Rossi
- Siena Brain Investigation & Neuromodulation Lab (Si-BIN Lab), Unit of Neurology and Clinical Neurophysiology, Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Emiliano Santarnecchi
- Precision Neuroscience & Neuromodulation Program, Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
15
|
Procopio R, Gagliardi M, Talarico M, Fortunato F, Sammarra I, Procopio AC, Roncada P, Malanga D, Annesi G, Gambardella A. Two Novel Variants in the CHRNA2 and SCN2A Genes in Italian Patients with Febrile Seizures. Genes (Basel) 2024; 15:1407. [PMID: 39596607 PMCID: PMC11593345 DOI: 10.3390/genes15111407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 10/28/2024] [Accepted: 10/28/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Febrile seizures (FSs) are the most common form of epilepsy in children aged between six months and five years. The exact cause is unknown, but several studies have demonstrated the importance of genetic predisposition, with increasing involvement of receptors and ion channels. The present study aims to identify novel pathogenic variants in Italian patients with FSs. METHODS We performed targeted panel sequencing in a cohort of 21 patients with FSs. In silico analysis was performed to predict the pathogenic role of the resulting variants. RESULTS We found two novel variants segregating in two families with FSs: c.1021C>G (p.Leu341Val) in the CHRNA2 gene and c.140A>G (p.Glu47Gly) in SCN2A. CONCLUSIONS The c.1021C>G (p.Leu341Val) variant leads to a codon change of highly conserved leucine to valine at position 341 and is located in segments M3 of the subunit, which is important for channel gating. The c.140A>G (p.Glu47Gly) variant causes a substitution of glutamic acid with glycine at position 47 of the protein, which is highly conserved across the species. Moreover, it is located in the N-terminal domain, a region commonly affected in ASD, which impacts the inactivation kinetics and voltage dependence of steady-state activation. Further analyses are needed to better explain the role of CHRNA2 and SCN2A in the development of febrile seizures.
Collapse
Affiliation(s)
- Radha Procopio
- Department of Medical and Surgical Sciences, Neuroscience Research Center, Magna Graecia University, 88100 Catanzaro, Italy;
| | - Monica Gagliardi
- Department of Medical and Surgical Sciences, Neuroscience Research Center, Magna Graecia University, 88100 Catanzaro, Italy;
| | - Mariagrazia Talarico
- Institute of Neurology, Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy; (M.T.); (F.F.); (I.S.); (A.G.)
| | - Francesco Fortunato
- Institute of Neurology, Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy; (M.T.); (F.F.); (I.S.); (A.G.)
| | - Ilaria Sammarra
- Institute of Neurology, Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy; (M.T.); (F.F.); (I.S.); (A.G.)
| | - Anna Caterina Procopio
- Department of Health Sciences, Magna Graecia University, 88100 Catanzaro, Italy; (A.C.P.); (P.R.)
| | - Paola Roncada
- Department of Health Sciences, Magna Graecia University, 88100 Catanzaro, Italy; (A.C.P.); (P.R.)
| | - Donatella Malanga
- Laboratory of Molecular Oncology, Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy;
- Interdepartmental Center of Services (CIS), Magna Graecia University, 88100 Catanzaro, Italy
| | - Grazia Annesi
- Institute for Biomedical Research and Innovation, National Research Council, 87050 Mangone, Italy;
| | - Antonio Gambardella
- Institute of Neurology, Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy; (M.T.); (F.F.); (I.S.); (A.G.)
| |
Collapse
|
16
|
Li Z, Luo J, Li C, Zhu H. Upregulation of Nav1.6 expression in the ventral posterolateral nucleus of thalamus contributes to hyperalgesia in a model of Parkinson's disease. Exp Neurol 2024; 383:115032. [PMID: 39490625 DOI: 10.1016/j.expneurol.2024.115032] [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: 08/15/2024] [Revised: 10/23/2024] [Accepted: 10/24/2024] [Indexed: 11/05/2024]
Abstract
Pain is the most common non-motor manifestation of Parkinson's disease (PD), affecting the quality of life for patients. Nav1.6 is the most abundant subtype of voltage-gated sodium channels (VGSCs) in the brain of adult mammals. Here we investigated the expression patterns of Nav1.6 in the ventral posterolateral (VPL) nucleus of the thalamus and its involvement in the development of hyperalgesia in 6-hydroxydopamine (6-OHDA)-lesioned rats. The results showed a significant increase in Nav1.6 expression in reactive astrocytes of the ipsilateral VPL in 6-OHDA-lesioned rats at 4 weeks post-injection. Moreover, 6-OHDA-lesioned rats exhibited mechanical hyperalgesia, but did not display thermal hyperalgesia in the ipsilateral paw at the same time point. The down-regulation of Nav1.6 in the ipsilateral VPL can reduce mechanical hyperalgesia and improve sensorimotor impairments in 6-OHDA- lesioned rats. Furthermore, the analysis of local field potentials (LFPs) revealed that the increased Nav1.6 may participate in abnormal synchronized oscillations within the thalamocortical loop in 6-OHDA-lesioned rats. These findings suggest that the altered expression of Nav1.6 in astrocytes of the VPL may play an important role in the abnormal processing of pain within the thalamocortical circuit, contributing to the formation of mechanical hyperalgesia in animal models of PD.
Collapse
Affiliation(s)
- Zhiwei Li
- School of Life Science, Shanghai University, China
| | - Jiamin Luo
- School of Life Science, Shanghai University, China
| | | | - Hongyan Zhu
- School of Life Science, Shanghai University, China.
| |
Collapse
|
17
|
Shabani K, Krupp J, Lemesre E, Lévy N, Tran H. Voltage-Gated Ion Channel Compensatory Effect in DEE: Implications for Future Therapies. Cells 2024; 13:1763. [PMID: 39513870 PMCID: PMC11544952 DOI: 10.3390/cells13211763] [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: 09/09/2024] [Revised: 10/02/2024] [Accepted: 10/11/2024] [Indexed: 11/16/2024] Open
Abstract
Developmental and Epileptic Encephalopathies (DEEs) represent a clinically and genetically heterogeneous group of rare and severe epilepsies. DEEs commonly begin early in infancy with frequent seizures of various types associated with intellectual disability and leading to a neurodevelopmental delay or regression. Disease-causing genomic variants have been identified in numerous genes and are implicated in over 100 types of DEEs. In this context, genes encoding voltage-gated ion channels (VGCs) play a significant role, and part of the large phenotypic variability observed in DEE patients carrying VGC mutations could be explained by the presence of genetic modifier alleles that can compensate for these mutations. This review will focus on the current knowledge of the compensatory effect of DEE-associated voltage-gated ion channels and their therapeutic implications in DEE. We will enter into detailed considerations regarding the sodium channels SCN1A, SCN2A, and SCN8A; the potassium channels KCNA1, KCNQ2, and KCNT1; and the calcium channels CACNA1A and CACNA1G.
Collapse
Affiliation(s)
- Khadijeh Shabani
- Institut de Recherches Servier, Rue Francis Perrin, 91190 Gif-sur-Yvette, France; (J.K.); (E.L.); (N.L.)
| | | | | | | | - Helene Tran
- Institut de Recherches Servier, Rue Francis Perrin, 91190 Gif-sur-Yvette, France; (J.K.); (E.L.); (N.L.)
| |
Collapse
|
18
|
Wang W, Williams DJ, Teoh JJ, Soundararajan D, Zuberi A, Lutz CM, Frankel WN, Makinson CD. Impaired axon initial segment structure and function in a model of ARHGEF9 developmental and epileptic encephalopathy. Proc Natl Acad Sci U S A 2024; 121:e2400709121. [PMID: 39374387 PMCID: PMC11494352 DOI: 10.1073/pnas.2400709121] [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: 01/15/2024] [Accepted: 07/25/2024] [Indexed: 10/09/2024] Open
Abstract
Developmental and epileptic encephalopathies (DEE) are rare but devastating and largely intractable childhood epilepsies. Genetic variants in ARHGEF9, encoding a scaffolding protein important for the organization of the postsynaptic density of inhibitory synapses, are associated with DEE accompanied by complex neurological phenotypes. In a mouse model carrying a patient-derived ARHGEF9 variant associated with severe disease, we observed aggregation of postsynaptic proteins and loss of functional inhibitory synapses at the axon initial segment (AIS), altered axo-axonic synaptic inhibition, disrupted action potential generation, and complex seizure phenotypes consistent with clinical observations. These results illustrate diverse roles of ARHGEF9 that converge on regulation of the structure and function of the AIS, thus revealing a pathological mechanism for ARHGEF9-associated DEE. This unique example of a neuropathological condition associated with multiple AIS dysfunctions may inform strategies for treating neurodevelopmental diseases.
Collapse
Affiliation(s)
- Wanqi Wang
- Department of Neurology, Center for Translational Research in Neurodevelopmental Disease, Columbia University Irving Medical Center, New York, NY10032
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY10032
| | - Damian J. Williams
- Department of Neurology, Center for Translational Research in Neurodevelopmental Disease, Columbia University Irving Medical Center, New York, NY10032
- Department of Neuroscience, Columbia University Irving Medical Center, New York, NY10032
| | - Jia Jie Teoh
- Department of Neurology, Center for Translational Research in Neurodevelopmental Disease, Columbia University Irving Medical Center, New York, NY10032
- Department of Neurology, Columbia University Irving Medical Center, New York, NY10032
| | - Divyalakshmi Soundararajan
- Department of Neurology, Center for Translational Research in Neurodevelopmental Disease, Columbia University Irving Medical Center, New York, NY10032
- Department of Neurology, Columbia University Irving Medical Center, New York, NY10032
| | - Aamir Zuberi
- The Rare and Orphan Disease Center, The Jackson Laboratory, Bar Harbor, ME04609
| | - Cathleen M. Lutz
- The Rare and Orphan Disease Center, The Jackson Laboratory, Bar Harbor, ME04609
| | - Wayne N. Frankel
- Department of Neurology, Center for Translational Research in Neurodevelopmental Disease, Columbia University Irving Medical Center, New York, NY10032
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY10032
- The Rare and Orphan Disease Center, The Jackson Laboratory, Bar Harbor, ME04609
| | - Christopher D. Makinson
- Department of Neurology, Center for Translational Research in Neurodevelopmental Disease, Columbia University Irving Medical Center, New York, NY10032
- Department of Neuroscience, Columbia University Irving Medical Center, New York, NY10032
- Department of Neurology, Columbia University Irving Medical Center, New York, NY10032
| |
Collapse
|
19
|
Müller P, Draguhn A, Egorov AV. Persistent sodium currents in neurons: potential mechanisms and pharmacological blockers. Pflugers Arch 2024; 476:1445-1473. [PMID: 38967655 PMCID: PMC11381486 DOI: 10.1007/s00424-024-02980-7] [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: 05/04/2024] [Revised: 06/07/2024] [Accepted: 06/11/2024] [Indexed: 07/06/2024]
Abstract
Persistent sodium current (INaP) is an important activity-dependent regulator of neuronal excitability. It is involved in a variety of physiological and pathological processes, including pacemaking, prolongation of sensory potentials, neuronal injury, chronic pain and diseases such as epilepsy and amyotrophic lateral sclerosis. Despite its importance, neither the molecular basis nor the regulation of INaP are sufficiently understood. Of particular significance is a solid knowledge and widely accepted consensus about pharmacological tools for analysing the function of INaP and for developing new therapeutic strategies. However, the literature on INaP is heterogeneous, with varying definitions and methodologies used across studies. To address these issues, we provide a systematic review of the current state of knowledge on INaP, with focus on mechanisms and effects of this current in the central nervous system. We provide an overview of the specificity and efficacy of the most widely used INaP blockers: amiodarone, cannabidiol, carbamazepine, cenobamate, eslicarbazepine, ethosuximide, gabapentin, GS967, lacosamide, lamotrigine, lidocaine, NBI-921352, oxcarbazepine, phenytoine, PRAX-562, propofol, ranolazine, riluzole, rufinamide, topiramate, valproaic acid and zonisamide. We conclude that there is strong variance in the pharmacological effects of these drugs, and in the available information. At present, GS967 and riluzole can be regarded bona fide INaP blockers, while phenytoin and lacosamide are blockers that only act on the slowly inactivating component of sodium currents.
Collapse
Affiliation(s)
- Peter Müller
- Department Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tuebingen , Hoppe-Seyler-Straße 3, 72076, Tübingen, Germany.
| | - Andreas Draguhn
- Institute for Physiology and Pathophysiology, Medical Faculty, Heidelberg University, Im Neuenheimer Feld 326, 69120, Heidelberg, Germany
| | - Alexei V Egorov
- Institute for Physiology and Pathophysiology, Medical Faculty, Heidelberg University, Im Neuenheimer Feld 326, 69120, Heidelberg, Germany
| |
Collapse
|
20
|
Turovskaya MV, Gavrish MS, Tarabykin VS, Babaev AA. Overexpression of BDNF Suppresses the Epileptiform Activity in Cortical Neurons of Heterozygous Mice with a Transcription Factor Sip1 Deletion. Int J Mol Sci 2024; 25:10537. [PMID: 39408863 PMCID: PMC11476396 DOI: 10.3390/ijms251910537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 09/21/2024] [Accepted: 09/26/2024] [Indexed: 10/20/2024] Open
Abstract
Since genetic mutations during brain development play a significant role in the genesis of epilepsy, and such genetically determined epilepsies are the most difficult to treat, there is a need to study the mechanisms of epilepsy development with deletions of various transcription factors. We utilized heterozygous mice (Sip1wt/fl) with a neuronal deletion of the transcription factor Sip1 (Smad interacting protein 1) in the cerebral cortex. These mice are characterized by cognitive impairment and are prone to epilepsy. It is known that the brain-derived neurotrophic factor (BDNF) has a neuroprotective effect in various neurodegenerative diseases. Therefore, we created and applied an adeno-associated construct carrying the BDNF sequence selectively in neurons. Using in vitro and in vivo research models, we were able to identify a key gen, the disruption of whose expression accompanies the deletion of Sip1 and contributes to hyperexcitation of neurons in the cerebral cortex. Overexpression of BDNF in cortical neurons eliminated epileptiform activity in neurons obtained from heterozygous Sip1 mice in a magnesium-free model of epileptiform activity (in vitro). Using PCR analysis, it was possible to identify correlations in the expression profile of genes encoding key proteins responsible for neurotransmission and neuronal survival. The effects of BDNF overexpression on the expression profiles of these genes were also revealed. Using BDNF overexpression in cortical neurons of heterozygous Sip1 mice, it was possible to achieve 100% survival in the pilocarpine model of epilepsy. At the level of gene expression in the cerebral cortex, patterns were established that may be involved in the protection of brain cells from epileptic seizures and the restoration of cognitive functions in mice with Sip1 deletion.
Collapse
Affiliation(s)
- Maria V. Turovskaya
- Institute of Neuroscience, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Ave, 603022 Nizhny Novgorod, Russia; (M.S.G.); (A.A.B.)
- Institute of Cell Biophysics of the Russian Academy of Sciences, Federal Research Center “Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences”, Institutskaya st. building 3, 142290 Pushchino, Russia
| | - Maria S. Gavrish
- Institute of Neuroscience, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Ave, 603022 Nizhny Novgorod, Russia; (M.S.G.); (A.A.B.)
| | - Viktor S. Tarabykin
- Institute of Cell Biology and Neurobiology, Charité—Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Alexei A. Babaev
- Institute of Neuroscience, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Ave, 603022 Nizhny Novgorod, Russia; (M.S.G.); (A.A.B.)
| |
Collapse
|
21
|
Espino CM, Nagaraja C, Ortiz S, Dayton JR, Murali AR, Ma Y, Mann EL, Garlapalli S, Wohlgemuth RP, Brashear SE, Smith LR, Wilkinson KA, Griffith TN. Differential encoding of mammalian proprioception by voltage-gated sodium channels. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.27.609982. [PMID: 39253497 PMCID: PMC11383322 DOI: 10.1101/2024.08.27.609982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
Animals that require purposeful movement for survival are endowed with mechanosensory neurons called proprioceptors that provide essential sensory feedback from muscles and joints to spinal cord circuits, which modulates motor output. Despite the essential nature of proprioceptive signaling in daily life, the mechanisms governing proprioceptor activity are poorly understood. Here, we have identified distinct and nonredundant roles for two voltage-gated sodium channels (NaVs), NaV1.1 and NaV1.6, in mammalian proprioception. Deletion of NaV1.6 in somatosensory neurons (NaV1.6cKO mice) causes severe motor deficits accompanied by complete loss of proprioceptive transmission, which contrasts with our previous findings using similar mouse models to target NaV1.1 (NaV1.1cKO). In NaV1.6cKO animals, loss of proprioceptive feedback caused non-cell-autonomous impairments in proprioceptor end-organs and skeletal muscle that were absent in NaV1.1cKO mice. We attribute the differential contribution of NaV1.1 and NaV1.6 in proprioceptor function to distinct cellular localization patterns. Collectively, these data provide the first evidence that NaV subtypes uniquely shape neurotransmission within a somatosensory modality.
Collapse
Affiliation(s)
- Cyrrus M. Espino
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA, USA
| | - Chetan Nagaraja
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA, USA
| | - Serena Ortiz
- Department of Biological Sciences, San José State University, San Jose, CA, USA
| | - Jacquelyn R. Dayton
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA, USA
| | - Akash R. Murali
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA, USA
- Undergraduate Program in Neurobiology, Physiology and Behavior, University of California, Davis, Davis, CA, USA
| | - Yanki Ma
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA, USA
- Undergraduate Program in Neurobiology, Physiology and Behavior, University of California, Davis, Davis, CA, USA
| | - Emari L. Mann
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA, USA
- Postbaccalaureate Research Education Program at UC Davis, University of California, Davis, Davis, CA, USA
| | - Snigdha Garlapalli
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA, USA
- Undergraduate Program in Psychology, University of California, Davis, Davis, CA, USA
| | - Ross P. Wohlgemuth
- Department of Physiology, Neurobiology, and Behavior, University of California, Davis, Davis, CA, USA
| | - Sarah E. Brashear
- Department of Physiology, Neurobiology, and Behavior, University of California, Davis, Davis, CA, USA
| | - Lucas R. Smith
- Department of Physiology, Neurobiology, and Behavior, University of California, Davis, Davis, CA, USA
| | | | - Theanne N. Griffith
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA, USA
| |
Collapse
|
22
|
Zhao R, Ren B, Xiao Y, Tian J, Zou Y, Wei J, Qi Y, Hu A, Xie X, Huang ZJ, Shu Y, He M, Lu J, Tai Y. Axo-axonic synaptic input drives homeostatic plasticity by tuning the axon initial segment structurally and functionally. SCIENCE ADVANCES 2024; 10:eadk4331. [PMID: 39093969 PMCID: PMC11296346 DOI: 10.1126/sciadv.adk4331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 06/27/2024] [Indexed: 08/04/2024]
Abstract
Homeostatic plasticity maintains the stability of functional brain networks. The axon initial segment (AIS), where action potentials start, undergoes dynamic adjustment to exert powerful control over neuronal firing properties in response to network activity changes. However, it is poorly understood whether this plasticity involves direct synaptic input to the AIS. Here, we show that changes of GABAergic synaptic input from chandelier cells (ChCs) drive homeostatic tuning of the AIS of principal neurons (PNs) in the prelimbic (PL) region, while those from parvalbumin-positive basket cells do not. This tuning is evident in AIS morphology, voltage-gated sodium channel expression, and PN excitability. Moreover, the impact of this homeostatic plasticity can be reflected in animal behavior. Social behavior, inversely linked to PL PN activity, shows time-dependent alterations tightly coupled to changes in AIS plasticity and PN excitability. Thus, AIS-originated homeostatic plasticity in PNs may counteract deficits elicited by imbalanced ChC presynaptic input at cellular and behavioral levels.
Collapse
Affiliation(s)
- Rui Zhao
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Neurobiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Baihui Ren
- Center for Brain Science of Shanghai Children’s Medical Center, Department of Anatomy and Physiology, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Yujie Xiao
- Department of Neurology, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan University, Shanghai 200032, China
| | - Jifeng Tian
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Neurobiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yi Zou
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Neurobiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jiafan Wei
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Neurobiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yanqing Qi
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Neurobiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Ankang Hu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Xiaoying Xie
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Neurobiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Z. Josh Huang
- Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA
- Department of Biomedical Engineering, Duke University Pratt School of Engineering, Durham, NC 27708, USA
| | - Yousheng Shu
- Department of Neurology, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan University, Shanghai 200032, China
| | - Miao He
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Neurobiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jiangteng Lu
- Center for Brain Science of Shanghai Children’s Medical Center, Department of Anatomy and Physiology, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
- Songjiang Research Institute, Shanghai Songjiang District Central Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai 200025, China
| | - Yilin Tai
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Neurobiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| |
Collapse
|
23
|
Wang Y, Yang H, Li N, Wang L, Guo C, Ma W, Liu S, Peng C, Chen J, Song H, Chen H, Ma X, Yi J, Lian J, Kong W, Dong J, Tu X, Shah M, Tian X, Huang Z. A Novel Ubiquitin Ligase Adaptor PTPRN Suppresses Seizure Susceptibility through Endocytosis of Na V1.2 Sodium Channels. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400560. [PMID: 38874331 PMCID: PMC11304301 DOI: 10.1002/advs.202400560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 05/06/2024] [Indexed: 06/15/2024]
Abstract
Intrinsic plasticity, a fundamental process enabling neurons to modify their intrinsic properties, plays a crucial role in shaping neuronal input-output function and is implicated in various neurological and psychiatric disorders. Despite its importance, the underlying molecular mechanisms of intrinsic plasticity remain poorly understood. In this study, a new ubiquitin ligase adaptor, protein tyrosine phosphatase receptor type N (PTPRN), is identified as a regulator of intrinsic neuronal excitability in the context of temporal lobe epilepsy. PTPRN recruits the NEDD4 Like E3 Ubiquitin Protein Ligase (NEDD4L) to NaV1.2 sodium channels, facilitating NEDD4L-mediated ubiquitination, and endocytosis of NaV1.2. Knockout of PTPRN in hippocampal granule cells leads to augmented NaV1.2-mediated sodium currents and higher intrinsic excitability, resulting in increased seizure susceptibility in transgenic mice. Conversely, adeno-associated virus-mediated delivery of PTPRN in the dentate gyrus region decreases intrinsic excitability and reduces seizure susceptibility. Moreover, the present findings indicate that PTPRN exerts a selective modulation effect on voltage-gated sodium channels. Collectively, PTPRN plays a significant role in regulating intrinsic excitability and seizure susceptibility, suggesting a potential strategy for precise modulation of NaV1.2 channels' function.
Collapse
Affiliation(s)
- Yifan Wang
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
- IDG/McGovern Institute for Brain ResearchPeking UniversityBeijing100871China
| | - Hui Yang
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
- IDG/McGovern Institute for Brain ResearchPeking UniversityBeijing100871China
| | - Na Li
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
- IDG/McGovern Institute for Brain ResearchPeking UniversityBeijing100871China
| | - Lili Wang
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
- IDG/McGovern Institute for Brain ResearchPeking UniversityBeijing100871China
| | - Chang Guo
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
- IDG/McGovern Institute for Brain ResearchPeking UniversityBeijing100871China
| | - Weining Ma
- Department of NeurologyShengjing Hospital Affiliated to China Medical UniversityShenyang110022China
| | - Shiqi Liu
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
- IDG/McGovern Institute for Brain ResearchPeking UniversityBeijing100871China
| | - Chao Peng
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
- IDG/McGovern Institute for Brain ResearchPeking UniversityBeijing100871China
| | - Jiexin Chen
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
- IDG/McGovern Institute for Brain ResearchPeking UniversityBeijing100871China
| | - Huifang Song
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
- IDG/McGovern Institute for Brain ResearchPeking UniversityBeijing100871China
| | - Hedan Chen
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
- IDG/McGovern Institute for Brain ResearchPeking UniversityBeijing100871China
| | - Xinyue Ma
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
- IDG/McGovern Institute for Brain ResearchPeking UniversityBeijing100871China
| | - Jingyun Yi
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
- IDG/McGovern Institute for Brain ResearchPeking UniversityBeijing100871China
| | - Jingjing Lian
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
- IDG/McGovern Institute for Brain ResearchPeking UniversityBeijing100871China
| | - Weikaixin Kong
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
- IDG/McGovern Institute for Brain ResearchPeking UniversityBeijing100871China
| | - Jie Dong
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
- IDG/McGovern Institute for Brain ResearchPeking UniversityBeijing100871China
| | - Xinyu Tu
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
- IDG/McGovern Institute for Brain ResearchPeking UniversityBeijing100871China
| | - Mala Shah
- UCL School of PharmacyUniversity College LondonLondonWC1N 1AXUK
| | - Xin Tian
- Department of NeurologyThe First Affiliated Hospital of Chongqing Medical UniversityChongqing Key Laboratory of NeurologyChongqing400016China
| | - Zhuo Huang
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
- IDG/McGovern Institute for Brain ResearchPeking UniversityBeijing100871China
| |
Collapse
|
24
|
Singh AK, Singh J, Goode NA, Laezza F. Crosstalk among WEE1 Kinase, AKT, and GSK3 in Nav1.2 Channelosome Regulation. Int J Mol Sci 2024; 25:8069. [PMID: 39125637 PMCID: PMC11311446 DOI: 10.3390/ijms25158069] [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: 05/30/2024] [Revised: 07/05/2024] [Accepted: 07/19/2024] [Indexed: 08/12/2024] Open
Abstract
The signaling complex around voltage-gated sodium (Nav) channels includes accessory proteins and kinases crucial for regulating neuronal firing. Previous studies showed that one such kinase, WEE1-critical to the cell cycle-selectively modulates Nav1.2 channel activity through the accessory protein fibroblast growth factor 14 (FGF14). Here, we tested whether WEE1 exhibits crosstalk with the AKT/GSK3 kinase pathway for coordinated regulation of FGF14/Nav1.2 channel complex assembly and function. Using the in-cell split luciferase complementation assay (LCA), we found that the WEE1 inhibitor II and GSK3 inhibitor XIII reduce the FGF14/Nav1.2 complex formation, while the AKT inhibitor triciribine increases it. However, combining WEE1 inhibitor II with either one of the other two inhibitors abolished its effect on the FGF14/Nav1.2 complex formation. Whole-cell voltage-clamp recordings of sodium currents (INa) in HEK293 cells co-expressing Nav1.2 channels and FGF14-GFP showed that WEE1 inhibitor II significantly suppresses peak INa density, both alone and in the presence of triciribine or GSK3 inhibitor XIII, despite the latter inhibitor's opposite effects on INa. Additionally, WEE1 inhibitor II slowed the tau of fast inactivation and caused depolarizing shifts in the voltage dependence of activation and inactivation. These phenotypes either prevailed or were additive when combined with triciribine but were outcompeted when both WEE1 inhibitor II and GSK3 inhibitor XIII were present. Concerted regulation by WEE1 inhibitor II, triciribine, and GSK3 inhibitor XIII was also observed in long-term inactivation and use dependency of Nav1.2 currents. Overall, these findings suggest a complex role for WEE1 kinase-in concert with the AKT/GSK3 pathway-in regulating the Nav1.2 channelosome.
Collapse
Affiliation(s)
- Aditya K. Singh
- Department of Pharmacology & Toxicology, The University of Texas Medical Branch, Galveston, TX 77555, USA; (J.S.); (N.A.G.); (F.L.)
| | | | | | | |
Collapse
|
25
|
He G, Zheng Y, Chang S, Wang L, Yang X, Hao H, Li J, Zhang X, Tian F, Liang X, Xu H, Wang P, Chen X, Cao Z, Fang S, Gao Z, Liu H. Discovery of Novel Pyrimidine-Based Derivatives as Nav1.2 Inhibitors with Efficacy in Mouse Models of Epilepsy. J Med Chem 2024. [PMID: 39037114 DOI: 10.1021/acs.jmedchem.4c00861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/23/2024]
Abstract
Dysfunction of voltage-gated sodium channel Nav1.2 causes various epileptic disorders, and inhibition of the channel has emerged as an attractive therapeutic strategy. However, currently available Nav1.2 inhibitors exhibit low potency and limited structural diversity. In this study, a novel series of pyrimidine-based derivatives with Nav1.2 inhibitory activity were designed, synthesized, and evaluated. Compounds 14 and 35 exhibited potent activity against Nav1.2, boasting IC50 values of 120 and 65 nM, respectively. Compound 14 displayed favorable pharmacokinetics (F = 43%) following intraperitoneal injection and excellent brain penetration potency (B/P = 3.6). Compounds 14 and 35 exhibited robust antiepileptic activities in the maximal electroshock test, with ED50 values of 3.2 and 11.1 mg/kg, respectively. Compound 35 also demonstrated potent antiepileptic activity in a 6 Hz (32 mA) model, with an ED50 value of 18.5 mg/kg. Overall, compounds 14 and 35 are promising leads for the development of new small-molecule therapeutics for epilepsy.
Collapse
Affiliation(s)
- Guoxue He
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yueming Zheng
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shunzhen Chang
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Long Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Pharmacophenomics Laboratory, Human Phenome Institute, Fudan University, Shanghai 201203, China
| | - Xiaohao Yang
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Haishuang Hao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jiyuan Li
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xian Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Fuyun Tian
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xuewu Liang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Haiyan Xu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Pei Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xueqin Chen
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Zeyu Cao
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Sui Fang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Zhaobing Gao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Hong Liu
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| |
Collapse
|
26
|
Chow CY, King GF. Shining a Light on Venom-Peptide Receptors: Venom Peptides as Targeted Agents for In Vivo Molecular Imaging. Toxins (Basel) 2024; 16:307. [PMID: 39057947 PMCID: PMC11281729 DOI: 10.3390/toxins16070307] [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: 05/13/2024] [Revised: 06/27/2024] [Accepted: 07/02/2024] [Indexed: 07/28/2024] Open
Abstract
Molecular imaging has revolutionised the field of biomedical research by providing a non-invasive means to visualise and understand biochemical processes within living organisms. Optical fluorescent imaging in particular allows researchers to gain valuable insights into the dynamic behaviour of a target of interest in real time. Ion channels play a fundamental role in cellular signalling, and they are implicated in diverse pathological conditions, making them an attractive target in the field of molecular imaging. Many venom peptides exhibit exquisite selectivity and potency towards ion channels, rendering them ideal agents for molecular imaging applications. In this review, we illustrate the use of fluorescently-labelled venom peptides for disease diagnostics and intraoperative imaging of brain tumours and peripheral nerves. Finally, we address challenges for the development and clinical translation of venom peptides as nerve-targeted imaging agents.
Collapse
Affiliation(s)
- Chun Yuen Chow
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072, Australia
- Australia Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, St. Lucia, QLD 4072, Australia
| | - Glenn F. King
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072, Australia
- Australia Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, St. Lucia, QLD 4072, Australia
| |
Collapse
|
27
|
Mittal D, Narayanan R. Network motifs in cellular neurophysiology. Trends Neurosci 2024; 47:506-521. [PMID: 38806296 DOI: 10.1016/j.tins.2024.04.008] [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: 01/15/2024] [Revised: 04/08/2024] [Accepted: 04/29/2024] [Indexed: 05/30/2024]
Abstract
Concepts from network science and graph theory, including the framework of network motifs, have been frequently applied in studying neuronal networks and other biological complex systems. Network-based approaches can also be used to study the functions of individual neurons, where cellular elements such as ion channels and membrane voltage are conceptualized as nodes within a network, and their interactions are denoted by edges. Network motifs in this context provide functional building blocks that help to illuminate the principles of cellular neurophysiology. In this review we build a case that network motifs operating within neurons provide tools for defining the functional architecture of single-neuron physiology and neuronal adaptations. We highlight the presence of such computational motifs in the cellular mechanisms underlying action potential generation, neuronal oscillations, dendritic integration, and neuronal plasticity. Future work applying the network motifs perspective may help to decipher the functional complexities of neurons and their adaptation during health and disease.
Collapse
Affiliation(s)
- Divyansh Mittal
- Centre for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Rishikesh Narayanan
- Cellular Neurophysiology Laboratory, Molecular Biophysics Unit, Indian Institute of Science, Bangalore, India.
| |
Collapse
|
28
|
Mahon S. Variation and convergence in the morpho-functional properties of the mammalian neocortex. Front Syst Neurosci 2024; 18:1413780. [PMID: 38966330 PMCID: PMC11222651 DOI: 10.3389/fnsys.2024.1413780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 06/03/2024] [Indexed: 07/06/2024] Open
Abstract
Man's natural inclination to classify and hierarchize the living world has prompted neurophysiologists to explore possible differences in brain organisation between mammals, with the aim of understanding the diversity of their behavioural repertoires. But what really distinguishes the human brain from that of a platypus, an opossum or a rodent? In this review, we compare the structural and electrical properties of neocortical neurons in the main mammalian radiations and examine their impact on the functioning of the networks they form. We discuss variations in overall brain size, number of neurons, length of their dendritic trees and density of spines, acknowledging their increase in humans as in most large-brained species. Our comparative analysis also highlights a remarkable consistency, particularly pronounced in marsupial and placental mammals, in the cell typology, intrinsic and synaptic electrical properties of pyramidal neuron subtypes, and in their organisation into functional circuits. These shared cellular and network characteristics contribute to the emergence of strikingly similar large-scale physiological and pathological brain dynamics across a wide range of species. These findings support the existence of a core set of neural principles and processes conserved throughout mammalian evolution, from which a number of species-specific adaptations appear, likely allowing distinct functional needs to be met in a variety of environmental contexts.
Collapse
Affiliation(s)
- Séverine Mahon
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital de la Pitié Salpêtrière, Paris, France
| |
Collapse
|
29
|
Thome C, Janssen JM, Karabulut S, Acuna C, D’Este E, Soyka SJ, Baum K, Bock M, Lehmann N, Roos J, Stevens NA, Hasegawa M, Ganea DA, Benoit CM, Gründemann J, Min L, Bird KM, Schultz C, Bennett V, Jenkins PM, Engelhardt M. Live imaging of excitable axonal microdomains in ankyrin-G-GFP mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.02.01.525891. [PMID: 38948770 PMCID: PMC11212890 DOI: 10.1101/2023.02.01.525891] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
The axon initial segment (AIS) constitutes not only the site of action potential initiation, but also a hub for activity-dependent modulation of output generation. Recent studies shedding light on AIS function used predominantly post-hoc approaches since no robust murine in vivo live reporters exist. Here, we introduce a reporter line in which the AIS is intrinsically labeled by an ankyrin-G-GFP fusion protein activated by Cre recombinase, tagging the native Ank3 gene. Using confocal, superresolution, and two-photon microscopy as well as whole-cell patch-clamp recordings in vitro, ex vivo, and in vivo, we confirm that the subcellular scaffold of the AIS and electrophysiological parameters of labeled cells remain unchanged. We further uncover rapid AIS remodeling following increased network activity in this model system, as well as highly reproducible in vivo labeling of AIS over weeks. This novel reporter line allows longitudinal studies of AIS modulation and plasticity in vivo in real-time and thus provides a unique approach to study subcellular plasticity in a broad range of applications.
Collapse
Affiliation(s)
- Christian Thome
- Institute of Anatomy and Cell Biology, Johannes Kepler University, 4020 Linz, Austria
- Clinical Research Institute of Neuroscience, Johannes Kepler University, 4020 Linz, Austria
- Institute of Physiology and Pathophysiology, Heidelberg University, 69120 Heidelberg, Germany
| | - Jan Maximilian Janssen
- Institute of Anatomy and Cell Biology, Johannes Kepler University, 4020 Linz, Austria
- Clinical Research Institute of Neuroscience, Johannes Kepler University, 4020 Linz, Austria
- Institute of Neuroanatomy, Mannheim Center for Translational Neuroscience (MCTN), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Seda Karabulut
- Institute of Neuroanatomy, Mannheim Center for Translational Neuroscience (MCTN), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Claudio Acuna
- Chica and Heinz Schaller Research Group, Institute of Anatomy and Cell Biology, Heidelberg University, 69120 Heidelberg, Germany
| | - Elisa D’Este
- Optical Microscopy Facility, Max Planck Institute for Medical Research, 69120 Heidelberg, Germany
| | - Stella J. Soyka
- Institute of Anatomy and Cell Biology, Dept. of Functional Neuroanatomy, Heidelberg University, 69120 Heidelberg, Germany
| | - Konrad Baum
- Institute of Anatomy and Cell Biology, Johannes Kepler University, 4020 Linz, Austria
- Clinical Research Institute of Neuroscience, Johannes Kepler University, 4020 Linz, Austria
| | - Michael Bock
- Institute of Neuroanatomy, Mannheim Center for Translational Neuroscience (MCTN), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Nadja Lehmann
- Institute of Neuroanatomy, Mannheim Center for Translational Neuroscience (MCTN), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Johannes Roos
- Institute of Anatomy and Cell Biology, Johannes Kepler University, 4020 Linz, Austria
- Clinical Research Institute of Neuroscience, Johannes Kepler University, 4020 Linz, Austria
- Institute of Neuroanatomy, Mannheim Center for Translational Neuroscience (MCTN), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Nikolas A. Stevens
- Institute of Physiology and Pathophysiology, Heidelberg University, 69120 Heidelberg, Germany
| | - Masashi Hasegawa
- German Center for Neurodegenerative Disease (DZNE), Neural Circuit Computations, 53127 Bonn, Germany
| | - Dan A. Ganea
- University of Basel, Department of Biomedicine, 4031 Basel, Switzerland
| | - Chloé M. Benoit
- German Center for Neurodegenerative Disease (DZNE), Neural Circuit Computations, 53127 Bonn, Germany
- University of Basel, Department of Biomedicine, 4031 Basel, Switzerland
| | - Jan Gründemann
- German Center for Neurodegenerative Disease (DZNE), Neural Circuit Computations, 53127 Bonn, Germany
- University of Basel, Department of Biomedicine, 4031 Basel, Switzerland
| | - Lia Min
- Departments of Pharmacology and Psychiatry, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Kalynn M. Bird
- Departments of Pharmacology and Psychiatry, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Christian Schultz
- Institute of Neuroanatomy, Mannheim Center for Translational Neuroscience (MCTN), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Vann Bennett
- Department of Biochemistry, Duke University Medical Center, Durham, NC 27710, USA
| | - Paul M. Jenkins
- Departments of Pharmacology and Psychiatry, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Maren Engelhardt
- Institute of Anatomy and Cell Biology, Johannes Kepler University, 4020 Linz, Austria
- Clinical Research Institute of Neuroscience, Johannes Kepler University, 4020 Linz, Austria
- Institute of Neuroanatomy, Mannheim Center for Translational Neuroscience (MCTN), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| |
Collapse
|
30
|
Buccino AP, Damart T, Bartram J, Mandge D, Xue X, Zbili M, Gänswein T, Jaquier A, Emmenegger V, Markram H, Hierlemann A, Van Geit W. A Multimodal Fitting Approach to Construct Single-Neuron Models With Patch Clamp and High-Density Microelectrode Arrays. Neural Comput 2024; 36:1286-1331. [PMID: 38776965 DOI: 10.1162/neco_a_01672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 02/20/2024] [Indexed: 05/25/2024]
Abstract
In computational neuroscience, multicompartment models are among the most biophysically realistic representations of single neurons. Constructing such models usually involves the use of the patch-clamp technique to record somatic voltage signals under different experimental conditions. The experimental data are then used to fit the many parameters of the model. While patching of the soma is currently the gold-standard approach to build multicompartment models, several studies have also evidenced a richness of dynamics in dendritic and axonal sections. Recording from the soma alone makes it hard to observe and correctly parameterize the activity of nonsomatic compartments. In order to provide a richer set of data as input to multicompartment models, we here investigate the combination of somatic patch-clamp recordings with recordings of high-density microelectrode arrays (HD-MEAs). HD-MEAs enable the observation of extracellular potentials and neural activity of neuronal compartments at subcellular resolution. In this work, we introduce a novel framework to combine patch-clamp and HD-MEA data to construct multicompartment models. We first validate our method on a ground-truth model with known parameters and show that the use of features extracted from extracellular signals, in addition to intracellular ones, yields models enabling better fits than using intracellular features alone. We also demonstrate our procedure using experimental data by constructing cell models from in vitro cell cultures. The proposed multimodal fitting procedure has the potential to augment the modeling efforts of the computational neuroscience community and provide the field with neuronal models that are more realistic and can be better validated.
Collapse
Affiliation(s)
- Alessio Paolo Buccino
- Bio Engineering Laboratory, Department of Biosystems Science and Engineering, ETH Zurich, 4056 Basel, Switzerland
| | - Tanguy Damart
- Blue Brain Project, École polytechnique fédérale de Lausanne, Campus Biotech, 1202 Geneva, Switzerland
| | - Julian Bartram
- Bio Engineering Laboratory, Department of Biosystems Science and Engineering, ETH Zurich, 4056 Basel, Switzerland
| | - Darshan Mandge
- Blue Brain Project, École polytechnique fédérale de Lausanne, Campus Biotech, 1202 Geneva, Switzerland
| | - Xiaohan Xue
- Bio Engineering Laboratory, Department of Biosystems Science and Engineering, ETH Zurich, 4056 Basel, Switzerland
| | - Mickael Zbili
- Blue Brain Project, École polytechnique fédérale de Lausanne, Campus Biotech, 1202 Geneva, Switzerland
| | - Tobias Gänswein
- Bio Engineering Laboratory, Department of Biosystems Science and Engineering, ETH Zurich, 4056 Basel, Switzerland
| | - Aurélien Jaquier
- Blue Brain Project, École polytechnique fédérale de Lausanne, Campus Biotech, 1202 Geneva, Switzerland
| | - Vishalini Emmenegger
- Bio Engineering Laboratory, Department of Biosystems Science and Engineering, ETH Zurich, 4056 Basel, Switzerland
| | - Henry Markram
- Blue Brain Project, École polytechnique fédérale de Lausanne, Campus Biotech, 1202 Geneva, Switzerland
| | - Andreas Hierlemann
- Bio Engineering Laboratory, Department of Biosystems Science and Engineering, ETH Zurich, 4056 Basel, Switzerland
| | - Werner Van Geit
- Blue Brain Project, École polytechnique fédérale de Lausanne, Campus Biotech, 1202 Geneva, Switzerland Present address: Foundation for Research on Information Technologies in Society (IT'IS), Zurich 8004, Switzerland
| |
Collapse
|
31
|
Zhao R, Ren B, Xiao Y, Tian J, Zou Y, Wei J, Qi Y, Hu A, Xie X, Huang ZJ, Shu Y, He M, Lu J, Tai Y. Axo-axonic synaptic input drives homeostatic plasticity by tuning the axon initial segment structurally and functionally. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.11.589005. [PMID: 38659885 PMCID: PMC11042219 DOI: 10.1101/2024.04.11.589005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
The stability of functional brain network is maintained by homeostatic plasticity, which restores equilibrium following perturbation. As the initiation site of action potentials, the axon initial segment (AIS) of glutamatergic projection neurons (PyNs) undergoes dynamic adjustment that exerts powerful control over neuronal firing properties in response to changes in network states. Although AIS plasticity has been reported to be coupled with the changes of network activity, it is poorly understood whether it involves direct synaptic input to the AIS. Here we show that changes of GABAergic synaptic input to the AIS of cortical PyNs, specifically from chandelier cells (ChCs), are sufficient to drive homeostatic tuning of the AIS within 1-2 weeks, while those from parvalbumin-positive basket cells do not. This tuning is reflected in the morphology of the AIS, the expression level of voltage-gated sodium channels, and the intrinsic neuronal excitability of PyNs. Interestingly, the timing of AIS tuning in PyNs of the prefrontal cortex corresponds to the recovery of changes in social behavior caused by alterations of ChC synaptic transmission. Thus, homeostatic plasticity of the AIS at postsynaptic PyNs may counteract deficits elicited by imbalanced ChC presynaptic input. Teaser Axon initial segment dynamically responds to changes in local input from chandelier cells to prevent abnormal neuronal functions.
Collapse
|
32
|
Ng ACH, Chahine M, Scantlebury MH, Appendino JP. Channelopathies in epilepsy: an overview of clinical presentations, pathogenic mechanisms, and therapeutic insights. J Neurol 2024; 271:3063-3094. [PMID: 38607431 DOI: 10.1007/s00415-024-12352-x] [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: 03/17/2024] [Revised: 03/24/2024] [Accepted: 03/25/2024] [Indexed: 04/13/2024]
Abstract
Pathogenic variants in genes encoding ion channels are causal for various pediatric and adult neurological conditions. In particular, several epilepsy syndromes have been identified to be caused by specific channelopathies. These encompass a spectrum from self-limited epilepsies to developmental and epileptic encephalopathies spanning genetic and acquired causes. Several of these channelopathies have exquisite responses to specific antiseizure medications (ASMs), while others ASMs may prove ineffective or even worsen seizures. Some channelopathies demonstrate phenotypic pleiotropy and can cause other neurological conditions outside of epilepsy. This review aims to provide a comprehensive exploration of the pathophysiology of seizure generation, ion channels implicated in epilepsy, and several genetic epilepsies due to ion channel dysfunction. We outline the clinical presentation, pathogenesis, and the current state of basic science and clinical research for these channelopathies. In addition, we briefly look at potential precision therapy approaches emerging for these disorders.
Collapse
Affiliation(s)
- Andy Cheuk-Him Ng
- Clinical Neuroscience and Pediatric Neurology, Department of Pediatrics, Cumming School of Medicine, Alberta Children's Hospital, University of Calgary, 28 Oki Drive NW, Calgary, AB, T3B 6A8, Canada
- Division of Neurology, Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta and Stollery Children's Hospital, Edmonton, AB, Canada
| | - Mohamed Chahine
- Department of Medicine, Faculty of Medicine, Université Laval, Quebec City, QC, Canada
- CERVO, Brain Research Centre, Quebec City, Canada
| | - Morris H Scantlebury
- Clinical Neuroscience and Pediatric Neurology, Department of Pediatrics, Cumming School of Medicine, Alberta Children's Hospital, University of Calgary, 28 Oki Drive NW, Calgary, AB, T3B 6A8, Canada
- Hotchkiss Brain Institute and Alberta Children's Hospital Research Institute, Calgary, Canada
| | - Juan P Appendino
- Clinical Neuroscience and Pediatric Neurology, Department of Pediatrics, Cumming School of Medicine, Alberta Children's Hospital, University of Calgary, 28 Oki Drive NW, Calgary, AB, T3B 6A8, Canada.
| |
Collapse
|
33
|
Ransdell JL, Carrasquillo Y, Bosch MK, Mellor RL, Ornitz DM, Nerbonne JM. Loss of Intracellular Fibroblast Growth Factor 14 (iFGF14) Increases the Excitability of Mature Hippocampal and Cortical Pyramidal Neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.04.592532. [PMID: 38746081 PMCID: PMC11092765 DOI: 10.1101/2024.05.04.592532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Mutations in FGF14 , which encodes intracellular fibroblast growth factor 14 (iFGF14), have been linked to spinocerebellar ataxia type 27 (SCA27), a multisystem disorder associated with progressive deficits in motor coordination and cognitive function. Mice ( Fgf14 -/- ) lacking iFGF14 display similar phenotypes, and we have previously shown that the deficits in motor coordination reflect reduced excitability of cerebellar Purkinje neurons, owing to the loss of iFGF14-mediated regulation of the voltage-dependence of inactivation of the fast transient component of the voltage-gated Na + (Nav) current, I NaT . Here, we present the results of experiments designed to test the hypothesis that loss of iFGF14 also attenuates the intrinsic excitability of mature hippocampal and cortical pyramidal neurons. Current-clamp recordings from adult mouse hippocampal CA1 pyramidal neurons in acute in vitro slices, however, revealed that repetitive firing rates were higher in Fgf14 -/- , than in wild type (WT), cells. In addition, the waveforms of individual action potentials were altered in Fgf14 -/- hippocampal CA1 pyramidal neurons, and the loss of iFGF14 reduced the time delay between the initiation of axonal and somal action potentials. Voltage-clamp recordings revealed that the loss of iFGF14 altered the voltage-dependence of activation, but not inactivation, of I NaT in CA1 pyramidal neurons. Similar effects of the loss of iFGF14 on firing properties were evident in current-clamp recordings from layer 5 visual cortical pyramidal neurons. Additional experiments demonstrated that the loss of iFGF14 does not alter the distribution of anti-Nav1.6 or anti-ankyrin G immunofluorescence labeling intensity along the axon initial segments (AIS) of mature hippocampal CA1 or layer 5 visual cortical pyramidal neurons in situ . Taken together, the results demonstrate that, in contrast with results reported for neonatal (rat) hippocampal pyramidal neurons in dissociated cell culture, the loss of iFGF14 does not disrupt AIS architecture or Nav1.6 localization/distribution along the AIS of mature hippocampal (or cortical) pyramidal neurons in situ .
Collapse
|
34
|
Wang C, Derderian KD, Hamada E, Zhou X, Nelson AD, Kyoung H, Ahituv N, Bouvier G, Bender KJ. Impaired cerebellar plasticity hypersensitizes sensory reflexes in SCN2A-associated ASD. Neuron 2024; 112:1444-1455.e5. [PMID: 38412857 PMCID: PMC11065582 DOI: 10.1016/j.neuron.2024.01.029] [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: 05/22/2023] [Revised: 01/03/2024] [Accepted: 01/29/2024] [Indexed: 02/29/2024]
Abstract
Children diagnosed with autism spectrum disorder (ASD) commonly present with sensory hypersensitivity or abnormally strong reactions to sensory stimuli. Such hypersensitivity can be overwhelming, causing high levels of distress that contribute markedly to the negative aspects of the disorder. Here, we identify a mechanism that underlies hypersensitivity in a sensorimotor reflex found to be altered in humans and in mice with loss of function in the ASD risk-factor gene SCN2A. The cerebellum-dependent vestibulo-ocular reflex (VOR), which helps maintain one's gaze during movement, was hypersensitized due to deficits in cerebellar synaptic plasticity. Heterozygous loss of SCN2A-encoded NaV1.2 sodium channels in granule cells impaired high-frequency transmission to Purkinje cells and long-term potentiation, a form of synaptic plasticity important for modulating VOR gain. VOR plasticity could be rescued in mice via a CRISPR-activator approach that increases Scn2a expression, demonstrating that evaluation of a simple reflex can be used to assess and quantify successful therapeutic intervention.
Collapse
Affiliation(s)
- Chenyu Wang
- Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA, USA; Department of Neurology, University of California, San Francisco, San Francisco, CA, USA; Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Kimberly D Derderian
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Elizabeth Hamada
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Xujia Zhou
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Andrew D Nelson
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA; Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Henry Kyoung
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Nadav Ahituv
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA; Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, USA; Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, USA
| | - Guy Bouvier
- Department of Physiology, University of California, San Francisco, San Francisco, CA, USA; Université Paris-Saclay, CNRS, Institut des Neurosciences Paris-Saclay, 91400 Saclay, France.
| | - Kevin J Bender
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA; Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
35
|
Eltokhi A, Lundstrom BN, Li J, Zweifel LS, Catterall WA, Gamal El-Din TM. Pathogenic gating pore current conducted by autism-related mutations in the Na V1.2 brain sodium channel. Proc Natl Acad Sci U S A 2024; 121:e2317769121. [PMID: 38564633 PMCID: PMC11009634 DOI: 10.1073/pnas.2317769121] [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: 10/23/2023] [Accepted: 02/26/2024] [Indexed: 04/04/2024] Open
Abstract
Autism spectrum disorder (ASD) is a complex neurodevelopmental condition characterized by social and communication deficits and repetitive behaviors. The genetic heterogeneity of ASD presents a challenge to the development of an effective treatment targeting the underlying molecular defects. ASD gating charge mutations in the KCNQ/KV7 potassium channel cause gating pore currents (Igp) and impair action potential (AP) firing of dopaminergic neurons in brain slices. Here, we investigated ASD gating charge mutations of the voltage-gated SCN2A/NaV1.2 brain sodium channel, which ranked high among the ion channel genes with mutations in individuals with ASD. Our results show that ASD mutations in the gating charges R2 in Domain-II (R853Q), and R1 (R1626Q) and R2 (R1629H) in Domain-IV of NaV1.2 caused Igp in the resting state of ~0.1% of the amplitude of central pore current. The R1626Q mutant also caused significant changes in the voltage dependence of fast inactivation, and the R1629H mutant conducted proton-selective Igp. These potentially pathogenic Igp were exacerbated by the absence of the extracellular Mg2+ and Ca2+. In silico simulation of the effects of these mutations in a conductance-based single-compartment cortical neuron model suggests that the inward Igp reduces the time to peak for the first AP in a train, increases AP rates during a train of stimuli, and reduces the interstimulus interval between consecutive APs, consistent with increased neural excitability and altered input/output relationships. Understanding this common pathophysiological mechanism among different voltage-gated ion channels at the circuit level will give insights into the underlying mechanisms of ASD.
Collapse
Affiliation(s)
- Ahmed Eltokhi
- Department of Pharmacology, University of Washington, Seattle, WA98195
| | - Brian Nils Lundstrom
- Department of Neurology in the Division of Epilepsy, Mayo Clinic, Rochester, MN55905
| | - Jin Li
- Department of Pharmacology, University of Washington, Seattle, WA98195
| | - Larry S. Zweifel
- Department of Pharmacology, University of Washington, Seattle, WA98195
- Department of Psychiatry & Behavioral Sciences, University of Washington, Seattle, WA98195
| | | | | |
Collapse
|
36
|
Nelson AD, Catalfio AM, Gupta JP, Min L, Caballero-Florán RN, Dean KP, Elvira CC, Derderian KD, Kyoung H, Sahagun A, Sanders SJ, Bender KJ, Jenkins PM. Physical and functional convergence of the autism risk genes Scn2a and Ank2 in neocortical pyramidal cell dendrites. Neuron 2024; 112:1133-1149.e6. [PMID: 38290518 PMCID: PMC11097922 DOI: 10.1016/j.neuron.2024.01.003] [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: 06/08/2022] [Revised: 04/26/2023] [Accepted: 01/03/2024] [Indexed: 02/01/2024]
Abstract
Dysfunction in sodium channels and their ankyrin scaffolding partners have both been implicated in neurodevelopmental disorders, including autism spectrum disorder (ASD). In particular, the genes SCN2A, which encodes the sodium channel NaV1.2, and ANK2, which encodes ankyrin-B, have strong ASD association. Recent studies indicate that ASD-associated haploinsufficiency in Scn2a impairs dendritic excitability and synaptic function in neocortical pyramidal cells, but how NaV1.2 is anchored within dendritic regions is unknown. Here, we show that ankyrin-B is essential for scaffolding NaV1.2 to the dendritic membrane of mouse neocortical neurons and that haploinsufficiency of Ank2 phenocopies intrinsic dendritic excitability and synaptic deficits observed in Scn2a+/- conditions. These results establish a direct, convergent link between two major ASD risk genes and reinforce an emerging framework suggesting that neocortical pyramidal cell dendritic dysfunction can contribute to neurodevelopmental disorder pathophysiology.
Collapse
Affiliation(s)
- Andrew D Nelson
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA; Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Amanda M Catalfio
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Julie P Gupta
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Lia Min
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, USA
| | | | - Kendall P Dean
- Cellular and Molecular Biology Program, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Carina C Elvira
- Cellular and Molecular Biology Program, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Kimberly D Derderian
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA; Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Henry Kyoung
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA; Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Atehsa Sahagun
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA; Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Stephan J Sanders
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA; Department of Psychiatry, University of California, San Francisco, San Francisco, CA, USA
| | - Kevin J Bender
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA; Department of Neurology, University of California, San Francisco, San Francisco, CA, USA.
| | - Paul M Jenkins
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, USA; Department of Psychiatry, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
37
|
Abstract
Repetitive transcranial magnetic stimulation (rTMS) has become an increasingly popular tool to modulate neural excitability and induce neural plasticity in clinical and preclinical models; however, the physiological mechanisms in which it exerts these effects remain largely unknown. To date, studies have primarily focused on characterizing rTMS-induced changes occurring at the synapse, with little attention given to changes in intrinsic membrane properties. However, accumulating evidence suggests that rTMS may induce its effects, in part, via intrinsic plasticity mechanisms, suggesting a new and potentially complementary understanding of how rTMS alters neural excitability and neural plasticity. In this review, we provide an overview of several intrinsic plasticity mechanisms before reviewing the evidence for rTMS-induced intrinsic plasticity. In addition, we discuss a select number of neurological conditions where rTMS-induced intrinsic plasticity has therapeutic potential before speculating on the temporal relationship between rTMS-induced intrinsic and synaptic plasticity.
Collapse
Affiliation(s)
- Emily S King
- Experimental and Regenerative Neurosciences, School of Biological Sciences, The University of Western Australia, Perth, Australia
- Perron Institute for Neurological and Translational Science, Perth, Australia
| | - Alexander D Tang
- Experimental and Regenerative Neurosciences, School of Biological Sciences, The University of Western Australia, Perth, Australia
- Perron Institute for Neurological and Translational Science, Perth, Australia
| |
Collapse
|
38
|
Hill SF, Yu W, Ziobro J, Chalasani S, Reger F, Meisler MH. Long-Term Downregulation of the Sodium Channel Gene Scn8a Is Therapeutic in Mouse Models of SCN8A Epilepsy. Ann Neurol 2024; 95:754-759. [PMID: 38113311 PMCID: PMC11170564 DOI: 10.1002/ana.26861] [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: 07/05/2023] [Revised: 11/06/2023] [Accepted: 12/14/2023] [Indexed: 12/21/2023]
Abstract
OBJECTIVE De novo mutations of the voltage-gated sodium channel gene SCN8A cause developmental and epileptic encephalopathy (DEE). Most pathogenic variants result in gain-of-function changes in activity of the sodium channel Nav1.6, poorly controlled seizures, and significant comorbidities. In previous work, an antisense oligonucleotide (ASO) reduced Scn8a transcripts and increased lifespan after neonatal administration to a mouse model. Here, we tested long-term ASO treatment initiated after seizure onset, as required for clinical application. METHODS ASO treatment was initiated after observation of a convulsive seizure and repeated at 4 to 6 week intervals for 1 year. We also tested the long-term efficacy of an AAV10-short hairpin RNA (shRNA) virus administered on P1. RESULTS Repeated treatment with the Scn8a ASO initiated after seizure onset provided long-term survival and reduced seizure frequency during a 12 month observation period. A single treatment with viral shRNA was also protective during 12 months of observation. INTERPRETATION Downregulation of Scn8a expression that is initiated after the onset of seizures is effective for long-term treatment in a model of SCN8A-DEE. Repeated ASO administration or a single dose of viral shRNA prevented seizures and extended survival through 12 months of observation. ANN NEUROL 2024;95:754-759.
Collapse
Affiliation(s)
- Sophie F Hill
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, United States
- Department of Human Genetics, University of Michigan, Ann Arbor, MI
| | - Wenxi Yu
- Department of Human Genetics, University of Michigan, Ann Arbor, MI
| | - Julie Ziobro
- Department of Pediatrics, University of Michigan, Ann Arbor, MI
| | - Sanjna Chalasani
- Department of Human Genetics, University of Michigan, Ann Arbor, MI
| | - Faith Reger
- Department of Human Genetics, University of Michigan, Ann Arbor, MI
| | - Miriam H Meisler
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, United States
- Department of Human Genetics, University of Michigan, Ann Arbor, MI
- Department of Neurology, University of Michigan, Ann Arbor, MI
| |
Collapse
|
39
|
Yang Y, Chen Z, Zhou J, Jiang S, Wang G, Wan L, Yu J, Jiang M, Wang Y, Hu J, Liu X, Wang Y. Anti-PD-1 treatment protects against seizure by suppressing sodium channel function. CNS Neurosci Ther 2024; 30:e14504. [PMID: 37904722 PMCID: PMC11017438 DOI: 10.1111/cns.14504] [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: 06/14/2023] [Revised: 10/05/2023] [Accepted: 10/06/2023] [Indexed: 11/01/2023] Open
Abstract
AIMS Although programmed cell death protein 1 (PD-1) typically serves as a target for immunotherapies, a few recent studies have found that PD-1 is expressed in the nervous system and that neuronal PD-1 might play a crucial role in regulating neuronal excitability. However, whether brain-localized PD-1 is involved in seizures and epileptogenesis is still unknown and worthy of in-depth exploration. METHODS The existence of PD-1 in human neurons was confirmed by immunohistochemistry, and PD-1 expression levels were measured by real-time quantitative PCR (RT-qPCR) and western blotting. Chemoconvulsants, pentylenetetrazol (PTZ) and cyclothiazide (CTZ), were applied for the establishment of in vivo (rodents) and in vitro (primary hippocampal neurons) models of seizure, respectively. SHR-1210 (a PD-1 monoclonal antibody) and sodium stibogluconate (SSG, a validated inhibitor of SH2-containing protein tyrosine phosphatase-1 [SHP-1]) were administrated to investigate the impact of PD-1 pathway blockade on epileptic behaviors of rodents and epileptiform discharges of neurons. A miRNA strategy was applied to determine the impact of PD-1 knockdown on neuronal excitability. The electrical activities and sodium channel function of neurons were determined by whole-cell patch-clamp recordings. The interaction between PD-1 and α-6 subunit of human voltage-gated sodium channel (Nav1.6) was validated by performing co-immunostaining and co-immunoprecipitation (co-IP) experiments. RESULTS Our results reveal that PD-1 protein and mRNA levels were upregulated in lesion cores compared with perifocal tissues of surgically resected specimens from patients with intractable epilepsy. Furthermore, we show that anti-PD-1 treatment has anti-seizure effects both in vivo and in vitro. Then, we reveal that PD-1 blockade can alter the electrophysiological properties of sodium channels. Moreover, we reveal that PD-1 acts together with downstream SHP-1 to regulate sodium channel function and hence neuronal excitability. Further investigation suggests that there is a direct interaction between neuronal PD-1 and Nav1.6. CONCLUSION Our study reveals that neuronal PD-1 plays an important role in epilepsy and that anti-PD-1 treatment protects against seizures by suppressing sodium channel function, identifying anti-PD-1 treatment as a novel therapeutic strategy for epilepsy.
Collapse
Affiliation(s)
- Yuling Yang
- Department of Neurology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Zhongshan HospitalFudan UniversityShanghaiChina
| | - Zhiyun Chen
- Department of Neurology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Zhongshan HospitalFudan UniversityShanghaiChina
| | - Jing Zhou
- Department of Neurology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Zhongshan HospitalFudan UniversityShanghaiChina
- Rehabilitation CenterShenzhen Second People's Hospital/The First Affiliated Hospital of Shenzhen University Health Science CenterShenzhenChina
| | - Shize Jiang
- Department of Neurosurgery, Huashan HospitalFudan UniversityShanghaiChina
| | - Guoxiang Wang
- Department of Neurology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Zhongshan HospitalFudan UniversityShanghaiChina
| | - Li Wan
- Department of Neurology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Zhongshan HospitalFudan UniversityShanghaiChina
- Rehabilitation CenterShenzhen Second People's Hospital/The First Affiliated Hospital of Shenzhen University Health Science CenterShenzhenChina
| | - Jiangning Yu
- Department of Neurology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Zhongshan HospitalFudan UniversityShanghaiChina
| | - Min Jiang
- Department of Neurology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Zhongshan HospitalFudan UniversityShanghaiChina
| | - Yulong Wang
- Rehabilitation CenterShenzhen Second People's Hospital/The First Affiliated Hospital of Shenzhen University Health Science CenterShenzhenChina
| | - Jie Hu
- Department of Neurosurgery, Huashan HospitalFudan UniversityShanghaiChina
| | - Xu Liu
- Department of Neurology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Zhongshan HospitalFudan UniversityShanghaiChina
| | - Yun Wang
- Department of Neurology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Zhongshan HospitalFudan UniversityShanghaiChina
| |
Collapse
|
40
|
Zhang L, Tian J, Lin Z, Dong Z. Efficient Sodium Transmembrane Permeation through Helically Folded Nanopores with Natural Channel-Like Ion Selectivity. J Am Chem Soc 2024; 146:8500-8507. [PMID: 38483183 DOI: 10.1021/jacs.3c14736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/28/2024]
Abstract
The selective transmembrane permeation of sodium ions achieved by biomimetic chemistry shows great potential to solve the problem of sodium ion transport blockade in diseases, but its implementation faces enormous difficulties. Herein, we design and synthesize a series of helically folded nanopores by employing a quinoline-oxadiazole structural sequence to finely replicate the pentahydrate structure of sodium ions. Surprisingly, these nanopores are capable of achieving sodium transmembrane permeation with ion selectivity at the level of natural sodium channels, as observed in rationally designed nanopores (M1-M5) with Na+/K+ ion selectivity ratio of up to 20.4. Moreover, slight structural variations in nanopore structures can switch ion transport modes between the channel and carrier. We found that, compared to the carrier mode, the channel mode not only transports ions faster but also has higher ion selectivity during transmembrane conduction, clearly illustrating that the trade-off phenomenon between ion selectivity and transport activity does not occur between the two transport modes of channel and carrier. At the same time, we also found that the spatial position and numbers of coordination sites are crucial for the sodium ion selectivity of the nanopores. Moreover, carrier M1 reported in this work is totally superior to the commercial Na+ carrier ETH2120, especially in terms of Na+/K+ ion selectivity, thus being a potentially practical Na+ carrier. Our study provides a new paradigm on the rational design of sodium-specific synthetic nanopores, which will open up the possibility for the application of artificial sodium-specific transmembrane permeation in biomedicine and disease treatment.
Collapse
Affiliation(s)
- Lei Zhang
- State Key Laboratory of Supramolecular Structure and Materials, and Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun 130012, China
| | - Jun Tian
- State Key Laboratory of Supramolecular Structure and Materials, and Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun 130012, China
| | - Ze Lin
- State Key Laboratory of Supramolecular Structure and Materials, and Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun 130012, China
| | - Zeyuan Dong
- State Key Laboratory of Supramolecular Structure and Materials, and Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun 130012, China
| |
Collapse
|
41
|
Wu J, Quraishi IH, Zhang Y, Bromwich M, Kaczmarek LK. Disease-causing Slack potassium channel mutations produce opposite effects on excitability of excitatory and inhibitory neurons. Cell Rep 2024; 43:113904. [PMID: 38457342 PMCID: PMC11013952 DOI: 10.1016/j.celrep.2024.113904] [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: 04/18/2023] [Revised: 12/18/2023] [Accepted: 02/16/2024] [Indexed: 03/10/2024] Open
Abstract
The KCNT1 gene encodes the sodium-activated potassium channel Slack (KCNT1, KNa1.1), a regulator of neuronal excitability. Gain-of-function mutations in humans cause cortical network hyperexcitability, seizures, and severe intellectual disability. Using a mouse model expressing the Slack-R455H mutation, we find that Na+-dependent K+ (KNa) and voltage-dependent sodium (NaV) currents are increased in both excitatory and inhibitory cortical neurons. These increased currents, however, enhance the firing of excitability neurons but suppress that of inhibitory neurons. We further show that the expression of NaV channel subunits, particularly that of NaV1.6, is upregulated and that the length of the axon initial segment and of axonal NaV immunostaining is increased in both neuron types. Our study on the coordinate regulation of KNa currents and the expression of NaV channels may provide an avenue for understanding and treating epilepsies and other neurological disorders.
Collapse
Affiliation(s)
- Jing Wu
- Department of Pharmacology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Imran H Quraishi
- Department of Neurology, Yale Comprehensive Epilepsy Center, Yale School of Medicine, New Haven, CT 06520, USA
| | - Yalan Zhang
- Department of Pharmacology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Mark Bromwich
- Department of Pharmacology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Leonard K Kaczmarek
- Department of Pharmacology, Yale School of Medicine, New Haven, CT 06520, USA; Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT 06520, USA.
| |
Collapse
|
42
|
Goodchild SJ, Shuart NG, Williams AD, Ye W, Parrish RR, Soriano M, Thouta S, Mezeyova J, Waldbrook M, Dean R, Focken T, Ghovanloo MR, Ruben PC, Scott F, Cohen CJ, Empfield J, Johnson JP. Molecular Pharmacology of Selective Na V1.6 and Dual Na V1.6/Na V1.2 Channel Inhibitors that Suppress Excitatory Neuronal Activity Ex Vivo. ACS Chem Neurosci 2024; 15:1169-1184. [PMID: 38359277 PMCID: PMC10958515 DOI: 10.1021/acschemneuro.3c00757] [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: 11/21/2023] [Revised: 01/25/2024] [Accepted: 01/30/2024] [Indexed: 02/17/2024] Open
Abstract
Voltage-gated sodium channel (NaV) inhibitors are used to treat neurological disorders of hyperexcitability such as epilepsy. These drugs act by attenuating neuronal action potential firing to reduce excitability in the brain. However, all currently available NaV-targeting antiseizure medications nonselectively inhibit the brain channels NaV1.1, NaV1.2, and NaV1.6, which potentially limits the efficacy and therapeutic safety margins of these drugs. Here, we report on XPC-7724 and XPC-5462, which represent a new class of small molecule NaV-targeting compounds. These compounds specifically target inhibition of the NaV1.6 and NaV1.2 channels, which are abundantly expressed in excitatory pyramidal neurons. They have a > 100-fold molecular selectivity against NaV1.1 channels, which are predominantly expressed in inhibitory neurons. Sparing NaV1.1 preserves the inhibitory activity in the brain. These compounds bind to and stabilize the inactivated state of the channels thereby reducing the activity of excitatory neurons. They have higher potency, with longer residency times and slower off-rates, than the clinically used antiseizure medications carbamazepine and phenytoin. The neuronal selectivity of these compounds is demonstrated in brain slices by inhibition of firing in cortical excitatory pyramidal neurons, without impacting fast spiking inhibitory interneurons. XPC-5462 also suppresses epileptiform activity in an ex vivo brain slice seizure model, whereas XPC-7224 does not, suggesting a possible requirement of Nav1.2 inhibition in 0-Mg2+- or 4-AP-induced brain slice seizure models. The profiles of these compounds will facilitate pharmacological dissection of the physiological roles of NaV1.2 and NaV1.6 in neurons and help define the role of specific channels in disease states. This unique selectivity profile provides a new approach to potentially treat disorders of neuronal hyperexcitability by selectively downregulating excitatory circuits.
Collapse
Affiliation(s)
- Samuel J. Goodchild
- Department
of Cellular and Molecular Biology, Xenon
Pharmaceuticals, Burnaby, BC V5G 4W8, Canada
| | - Noah Gregory Shuart
- Department
of Cellular and Molecular Biology, Xenon
Pharmaceuticals, Burnaby, BC V5G 4W8, Canada
| | - Aaron D. Williams
- Department
of Cellular and Molecular Biology, Xenon
Pharmaceuticals, Burnaby, BC V5G 4W8, Canada
| | - Wenlei Ye
- Neurocrine
Biosciences, San Diego, California 92130, United States
| | - R. Ryley Parrish
- Department
of Cellular and Molecular Biology, Xenon
Pharmaceuticals, Burnaby, BC V5G 4W8, Canada
| | - Maegan Soriano
- Department
of Cellular and Molecular Biology, Xenon
Pharmaceuticals, Burnaby, BC V5G 4W8, Canada
| | - Samrat Thouta
- Department
of Cellular and Molecular Biology, Xenon
Pharmaceuticals, Burnaby, BC V5G 4W8, Canada
| | - Janette Mezeyova
- Department
of Cellular and Molecular Biology, Xenon
Pharmaceuticals, Burnaby, BC V5G 4W8, Canada
| | - Matthew Waldbrook
- Department
of Cellular and Molecular Biology, Xenon
Pharmaceuticals, Burnaby, BC V5G 4W8, Canada
| | - Richard Dean
- Department
of Cellular and Molecular Biology, Xenon
Pharmaceuticals, Burnaby, BC V5G 4W8, Canada
| | - Thilo Focken
- Department
of Cellular and Molecular Biology, Xenon
Pharmaceuticals, Burnaby, BC V5G 4W8, Canada
| | - Mohammad-Reza Ghovanloo
- Department
of Cellular and Molecular Biology, Xenon
Pharmaceuticals, Burnaby, BC V5G 4W8, Canada
- Department
of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
- Department
of Neurology, Yale University, New Haven, Connecticut 06519, United States
| | - Peter C. Ruben
- Department
of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
| | - Fiona Scott
- Neurocrine
Biosciences, San Diego, California 92130, United States
| | - Charles J. Cohen
- Department
of Cellular and Molecular Biology, Xenon
Pharmaceuticals, Burnaby, BC V5G 4W8, Canada
| | - James Empfield
- Department
of Cellular and Molecular Biology, Xenon
Pharmaceuticals, Burnaby, BC V5G 4W8, Canada
| | - JP Johnson
- Department
of Cellular and Molecular Biology, Xenon
Pharmaceuticals, Burnaby, BC V5G 4W8, Canada
| |
Collapse
|
43
|
Mateus JC, Sousa MM, Burrone J, Aguiar P. Beyond a Transmission Cable-New Technologies to Reveal the Richness in Axonal Electrophysiology. J Neurosci 2024; 44:e1446232023. [PMID: 38479812 PMCID: PMC10941245 DOI: 10.1523/jneurosci.1446-23.2023] [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: 08/07/2023] [Revised: 12/13/2023] [Accepted: 12/14/2023] [Indexed: 03/17/2024] Open
Abstract
The axon is a neuronal structure capable of processing, encoding, and transmitting information. This assessment contrasts with a limiting, but deeply rooted, perspective where the axon functions solely as a transmission cable of somatodendritic activity, sending signals in the form of stereotypical action potentials. This perspective arose, at least partially, because of the technical difficulties in probing axons: their extreme length-to-diameter ratio and intricate growth paths preclude the study of their dynamics through traditional techniques. Recent findings are challenging this view and revealing a much larger repertoire of axonal computations. Axons display complex signaling processes and structure-function relationships, which can be modulated via diverse activity-dependent mechanisms. Additionally, axons can exhibit patterns of activity that are dramatically different from those of their corresponding soma. Not surprisingly, many of these recent discoveries have been driven by novel technology developments, which allow for in vitro axon electrophysiology with unprecedented spatiotemporal resolution and signal-to-noise ratio. In this review, we outline the state-of-the-art in vitro toolset for axonal electrophysiology and summarize the recent discoveries in axon function it has enabled. We also review the increasing repertoire of microtechnologies for controlling axon guidance which, in combination with the available cutting-edge electrophysiology and imaging approaches, have the potential for more controlled and high-throughput in vitro studies. We anticipate that a larger adoption of these new technologies by the neuroscience community will drive a new era of experimental opportunities in the study of axon physiology and consequently, neuronal function.
Collapse
Affiliation(s)
- J C Mateus
- i3S- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
| | - M M Sousa
- i3S- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
| | - J Burrone
- MRC Centre for Neurodevelopmental Disorders, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, United Kingdom
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, United Kingdom
| | - P Aguiar
- i3S- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
| |
Collapse
|
44
|
Barlow BSM, Longtin A, Joós B. Impact on backpropagation of the spatial heterogeneity of sodium channel kinetics in the axon initial segment. PLoS Comput Biol 2024; 20:e1011846. [PMID: 38489374 PMCID: PMC10942053 DOI: 10.1371/journal.pcbi.1011846] [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] [Accepted: 01/21/2024] [Indexed: 03/17/2024] Open
Abstract
In a variety of neurons, action potentials (APs) initiate at the proximal axon, within a region called the axon initial segment (AIS), which has a high density of voltage-gated sodium channels (NaVs) on its membrane. In pyramidal neurons, the proximal AIS has been reported to exhibit a higher proportion of NaVs with gating properties that are "right-shifted" to more depolarized voltages, compared to the distal AIS. Further, recent experiments have revealed that as neurons develop, the spatial distribution of NaV subtypes along the AIS can change substantially, suggesting that neurons tune their excitability by modifying said distribution. When neurons are stimulated axonally, computational modelling has shown that this spatial separation of gating properties in the AIS enhances the backpropagation of APs into the dendrites. In contrast, in the more natural scenario of somatic stimulation, our simulations show that the same distribution can impede backpropagation, suggesting that the choice of orthodromic versus antidromic stimulation can bias or even invert experimental findings regarding the role of NaV subtypes in the AIS. We implemented a range of hypothetical NaV distributions in the AIS of three multicompartmental pyramidal cell models and investigated the precise kinetic mechanisms underlying such effects, as the spatial distribution of NaV subtypes is varied. With axonal stimulation, proximal NaV availability dominates, such that concentrating right-shifted NaVs in the proximal AIS promotes backpropagation. However, with somatic stimulation, the models are insensitive to availability kinetics. Instead, the higher activation threshold of right-shifted NaVs in the AIS impedes backpropagation. Therefore, recently observed developmental changes to the spatial separation and relative proportions of NaV1.2 and NaV1.6 in the AIS differentially impact activation and availability. The observed effects on backpropagation, and potentially learning via its putative role in synaptic plasticity (e.g. through spike-timing-dependent plasticity), are opposite for orthodromic versus antidromic stimulation, which should inform hypotheses about the impact of the developmentally regulated subcellular localization of these NaV subtypes.
Collapse
Affiliation(s)
- Benjamin S. M. Barlow
- Department of Physics, University of Ottawa, STEM Complex, 150 Louis-Pasteur Pvt, Ottawa, Ontario, Canada
| | - André Longtin
- Department of Physics, University of Ottawa, STEM Complex, 150 Louis-Pasteur Pvt, Ottawa, Ontario, Canada
- Center for Neural Dynamics and AI, University of Ottawa, Ottawa, Ontario, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Béla Joós
- Department of Physics, University of Ottawa, STEM Complex, 150 Louis-Pasteur Pvt, Ottawa, Ontario, Canada
- Center for Neural Dynamics and AI, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
45
|
Bingham CS, McIntyre CC. Coupled Activation of the Hyperdirect and Cerebellothalamic Pathways with Zona Incerta Deep Brain Stimulation. Mov Disord 2024; 39:539-545. [PMID: 38321526 PMCID: PMC10963140 DOI: 10.1002/mds.29717] [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: 03/28/2023] [Revised: 10/18/2023] [Accepted: 01/02/2024] [Indexed: 02/08/2024] Open
Abstract
BACKGROUND Deep brain stimulation (DBS) of the subthalamic nucleus (STN) or ventral intermediate nucleus (VIM) are established targets for the treatment of Parkinson's disease (PD) or essential tremor (ET), respectively. However, DBS of the zona incerta (ZI) can be effective for both disorders. VIM DBS is assumed to achieve its therapeutic effect via activation of the cerebellothalamic (CBT) pathway, whereas the activation of the hyperdirect (HD) pathway likely plays a role in the mechanisms of STN DBS. Interestingly, HD pathway axons also emit collaterals to the ZI and red nucleus (RN) and the CBT pathway courses nearby to the ZI. OBJECTIVE The aim was to examine the ability of ZI DBS to mutually activate the HD and CBT pathways in a detailed computational model of human DBS. METHODS We extended a previous model of the human HD pathway to incorporate axon collaterals to the ZI and RN. The anatomical framework of the model system also included representations of the CBT pathway and internal capsule (IC) fibers of passage. We then performed detailed biophysical simulations to quantify DBS activation of the HD, CBT, and IC pathways with electrodes located in either the STN or ZI. RESULTS STN DBS and ZI DBS both robustly activated the HD pathway. However, STN DBS was limited by IC activation at higher stimulus amplitudes. Alternatively, ZI DBS avoided IC activation while simultaneously activating the HD and CBT pathways. CONCLUSIONS From both neuroanatomical and biophysical perspectives, ZI DBS represents an advantageous target for coupled activation of the HD and CBT pathways. © 2024 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Clayton S. Bingham
- Department of Biomedical Engineering, Duke University, Durham, N.C. 27708
| | - Cameron C. McIntyre
- Department of Biomedical Engineering, Duke University, Durham, N.C. 27708
- Department of Neurosurgery, Duke University, Durham, N.C. 27708
| |
Collapse
|
46
|
Abbas F, Blömer LA, Millet H, Montnach J, De Waard M, Canepari M. Analysis of the effect of the scorpion toxin AaH-II on action potential generation in the axon initial segment. Sci Rep 2024; 14:4967. [PMID: 38424206 PMCID: PMC10904771 DOI: 10.1038/s41598-024-55315-y] [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: 10/11/2023] [Accepted: 02/22/2024] [Indexed: 03/02/2024] Open
Abstract
The toxin AaH-II, from the scorpion Androctonus australis Hector venom, is a 64 amino acid peptide that targets voltage-gated Na+ channels (VGNCs) and slows their inactivation. While at macroscopic cellular level AaH-II prolongs the action potential (AP), a functional analysis of the effect of the toxin in the axon initial segment (AIS), where VGNCs are highly expressed, was never performed so far. Here, we report an original analysis of the effect of AaH-II on the AP generation in the AIS of neocortical layer-5 pyramidal neurons from mouse brain slices. After determining that AaH-II does not discriminate between Nav1.2 and Nav1.6, i.e. between the two VGNC isoforms expressed in this neuron, we established that 7 nM was the smallest toxin concentration producing a minimal detectable deformation of the somatic AP after local delivery of the toxin. Using membrane potential imaging, we found that, at this minimal concentration, AaH-II substantially widened the AP in the AIS. Using ultrafast Na+ imaging, we found that local application of 7 nM AaH-II caused a large increase in the slower component of the Na+ influx in the AIS. Finally, using ultrafast Ca2+ imaging, we observed that 7 nM AaH-II produces a spurious slow Ca2+ influx via Ca2+-permeable VGNCs. Molecules targeting VGNCs, including peptides, are proposed as potential therapeutic tools. Thus, the present analysis in the AIS can be considered a general proof-of-principle on how high-resolution imaging techniques can disclose drug effects that cannot be observed when tested at the macroscopic level.
Collapse
Affiliation(s)
- Fatima Abbas
- Univ. Grenoble Alpes, CNRS, LIPhy, 38000, Grenoble, France
- Laboratories of Excellence, Ion Channel Science and Therapeutics, 06560, Valbonne, France
| | - Laila Ananda Blömer
- Univ. Grenoble Alpes, CNRS, LIPhy, 38000, Grenoble, France
- Laboratories of Excellence, Ion Channel Science and Therapeutics, 06560, Valbonne, France
| | - Hugo Millet
- Laboratories of Excellence, Ion Channel Science and Therapeutics, 06560, Valbonne, France
- Nantes Université, CNRS, INSERM, l'institut du Thorax, 44000, Nantes, France
| | - Jérôme Montnach
- Laboratories of Excellence, Ion Channel Science and Therapeutics, 06560, Valbonne, France
- Nantes Université, CNRS, INSERM, l'institut du Thorax, 44000, Nantes, France
| | - Michel De Waard
- Laboratories of Excellence, Ion Channel Science and Therapeutics, 06560, Valbonne, France
- Nantes Université, CNRS, INSERM, l'institut du Thorax, 44000, Nantes, France
| | - Marco Canepari
- Univ. Grenoble Alpes, CNRS, LIPhy, 38000, Grenoble, France.
- Laboratories of Excellence, Ion Channel Science and Therapeutics, 06560, Valbonne, France.
- Institut National de la Santé et Recherche Médicale, Paris, France.
- Laboratoire Interdisciplinaire de Physique (UMR 5588), Bat. E45, 140 Avenue de la Physique, Domaine Univ., 38402, St Martin d'Hères Cedex, France.
| |
Collapse
|
47
|
Yuan T, Wang Y, Jin Y, Yang H, Xu S, Zhang H, Chen Q, Li N, Ma X, Song H, Peng C, Geng Z, Dong J, Duan G, Sun Q, Yang Y, Yang F, Huang Z. Coupling of Slack and Na V1.6 sensitizes Slack to quinidine blockade and guides anti-seizure strategy development. eLife 2024; 12:RP87559. [PMID: 38289338 PMCID: PMC10942592 DOI: 10.7554/elife.87559] [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] [Indexed: 02/01/2024] Open
Abstract
Quinidine has been used as an anticonvulsant to treat patients with KCNT1-related epilepsy by targeting gain-of-function KCNT1 pathogenic mutant variants. However, the detailed mechanism underlying quinidine's blockade against KCNT1 (Slack) remains elusive. Here, we report a functional and physical coupling of the voltage-gated sodium channel NaV1.6 and Slack. NaV1.6 binds to and highly sensitizes Slack to quinidine blockade. Homozygous knockout of NaV1.6 reduces the sensitivity of native sodium-activated potassium currents to quinidine blockade. NaV1.6-mediated sensitization requires the involvement of NaV1.6's N- and C-termini binding to Slack's C-terminus and is enhanced by transient sodium influx through NaV1.6. Moreover, disrupting the Slack-NaV1.6 interaction by viral expression of Slack's C-terminus can protect against SlackG269S-induced seizures in mice. These insights about a Slack-NaV1.6 complex challenge the traditional view of 'Slack as an isolated target' for anti-epileptic drug discovery efforts and can guide the development of innovative therapeutic strategies for KCNT1-related epilepsy.
Collapse
Affiliation(s)
- Tian Yuan
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science CenterBeijingChina
| | - Yifan Wang
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science CenterBeijingChina
| | - Yuchen Jin
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science CenterBeijingChina
| | - Hui Yang
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science CenterBeijingChina
| | - Shuai Xu
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science CenterBeijingChina
| | - Heng Zhang
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang UniversityZhejiangChina
| | - Qian Chen
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science CenterBeijingChina
| | - Na Li
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science CenterBeijingChina
| | - Xinyue Ma
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science CenterBeijingChina
| | - Huifang Song
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science CenterBeijingChina
| | - Chao Peng
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science CenterBeijingChina
| | - Ze Geng
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science CenterBeijingChina
| | - Jie Dong
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science CenterBeijingChina
| | - Guifang Duan
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science CenterBeijingChina
| | - Qi Sun
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science CenterBeijingChina
| | - Yang Yang
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue UniversityWest LafayetteUnited States
| | - Fan Yang
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang UniversityZhejiangChina
- Department of Biophysics, Kidney Disease Center of the First Affiliated Hospital, Zhejiang University School of Medicine, HangzhouZhejiangChina
| | - Zhuo Huang
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science CenterBeijingChina
- IDG/McGovern Institute for Brain Research, Peking UniversityBeijingChina
| |
Collapse
|
48
|
Brown CO, Uy JA, Murtaza N, Rosa E, Alfonso A, Dave BM, Kilpatrick S, Cheng AA, White SH, Scherer SW, Singh KK. Disruption of the autism-associated gene SCN2A alters synaptic development and neuronal signaling in patient iPSC-glutamatergic neurons. Front Cell Neurosci 2024; 17:1239069. [PMID: 38293651 PMCID: PMC10824931 DOI: 10.3389/fncel.2023.1239069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 12/14/2023] [Indexed: 02/01/2024] Open
Abstract
SCN2A is an autism spectrum disorder (ASD) risk gene and encodes a voltage-gated sodium channel. However, the impact of ASD-associated SCN2A de novo variants on human neuron development is unknown. We studied SCN2A using isogenic SCN2A-/- induced pluripotent stem cells (iPSCs), and patient-derived iPSCs harboring a de novo R607* truncating variant. We used Neurogenin2 to generate excitatory (glutamatergic) neurons and found that SCN2A+/R607* and SCN2A-/- neurons displayed a reduction in synapse formation and excitatory synaptic activity. We found differential impact on actional potential dynamics and neuronal excitability that reveals a loss-of-function effect of the R607* variant. Our study reveals that a de novo truncating SCN2A variant impairs the development of human neuronal function.
Collapse
Affiliation(s)
- Chad O. Brown
- Department of Biochemistry and Biomedical Sciences, Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
- Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Jarryll A. Uy
- Krembil Research Institute, University Health Network, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Nadeem Murtaza
- Department of Biochemistry and Biomedical Sciences, Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
- Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Elyse Rosa
- Department of Biochemistry and Biomedical Sciences, Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
| | - Alexandria Alfonso
- Department of Biochemistry and Biomedical Sciences, Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
| | - Biren M. Dave
- Department of Molecular Genetics, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- SickKids Research Institute, The Hospital for Sick Children, Toronto, ON, Canada
| | - Savannah Kilpatrick
- Department of Biochemistry and Biomedical Sciences, Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
- Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Annie A. Cheng
- Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Sean H. White
- Department of Biochemistry and Biomedical Sciences, Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
| | - Stephen W. Scherer
- Department of Molecular Genetics, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- SickKids Research Institute, The Hospital for Sick Children, Toronto, ON, Canada
| | - Karun K. Singh
- Department of Biochemistry and Biomedical Sciences, Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
- Krembil Research Institute, University Health Network, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
49
|
Thompson AC, Aizenman CD. Characterization of Na + currents regulating intrinsic excitability of optic tectal neurons. Life Sci Alliance 2024; 7:e202302232. [PMID: 37918964 PMCID: PMC10622587 DOI: 10.26508/lsa.202302232] [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: 06/22/2023] [Revised: 10/20/2023] [Accepted: 10/23/2023] [Indexed: 11/04/2023] Open
Abstract
Developing neurons adapt their intrinsic excitability to maintain stable output despite changing synaptic input. The mechanisms behind this process remain unclear. In this study, we examined Xenopus optic tectal neurons and found that the expressions of Nav1.1 and Nav1.6 voltage-gated Na+ channels are regulated during changes in intrinsic excitability, both during development and becsuse of changes in visual experience. Using whole-cell electrophysiology, we demonstrate the existence of distinct, fast, persistent, and resurgent Na+ currents in the tectum, and show that these Na+ currents are co-regulated with changes in Nav channel expression. Using antisense RNA to suppress the expression of specific Nav subunits, we found that up-regulation of Nav1.6 expression, but not Nav1.1, was necessary for experience-dependent increases in Na+ currents and intrinsic excitability. Furthermore, this regulation was also necessary for normal development of sensory guided behaviors. These data suggest that the regulation of Na+ currents through the modulation of Nav1.6 expression, and to a lesser extent Nav1.1, plays a crucial role in controlling the intrinsic excitability of tectal neurons and guiding normal development of the tectal circuitry.
Collapse
|
50
|
Harris AC, Sun J, Jacobs KM. Concussive Head Trauma Deranges Axon Initial Segment Function in Axotomized and Intact Layer 5 Pyramidal Neurons. J Neurotrauma 2024; 41:244-270. [PMID: 37650832 PMCID: PMC11074420 DOI: 10.1089/neu.2022.0469] [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] [Indexed: 09/01/2023] Open
Abstract
The axon initial segment (AIS) is a critical locus of control of action potential (AP) generation and neuronal information synthesis. Concussive traumatic brain injury gives rise to diffuse axotomy, and the majority of neocortical axonal injury arises at the AIS. Consequently, concussive traumatic brain injury might profoundly disrupt the functional specialization of this region. To investigate this hypothesis, one and two days after mild central fluid percussion injury in Thy1-YFP-H mice, we recorded high-resolution APs from axotomized and adjacent intact layer 5 pyramidal neurons and applied a second derivative (2o) analysis to measure the AIS- and soma-regional contributions to the AP upstroke. All layer 5 pyramidal neurons recorded from sham animals manifested two stark 2o peaks separated by a negative intervening slope. In contrast, within injured mice, we discovered a subset of axotomized layer 5 pyramidal neurons in which the AIS-regional 2o peak was abolished, a functional perturbation associated with diminished excitability, axonal sprouting and distention of the AIS as assessed by staining for ankyrin-G. Our analysis revealed an additional subpopulation of both axotomized and intact layer 5 pyramidal neurons that manifested a melding together of the AIS- and soma-regional 2o peaks, suggesting a more subtle aberration of sodium channel function and/or translocation of the AIS initiation zone closer to the soma. When these experiments were repeated in animals in which cyclophilin-D was knocked out, these effects were ameliorated, suggesting that trauma-induced AIS functional perturbation is associated with mitochondrial calcium dysregulation.
Collapse
Affiliation(s)
- Alan C. Harris
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Jianli Sun
- Delaware Center for Neuroscience Research, Delaware State University, Dover, Delaware, USA
| | - Kimberle M. Jacobs
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, Virginia, USA
| |
Collapse
|