1
|
Wood JN, Yan N, Huang J, Zhao J, Akopian A, Cox JJ, Woods CG, Nassar MA. Sensory neuron sodium channels as pain targets; from cocaine to Journavx (VX-548, suzetrigine). J Gen Physiol 2025; 157:e202513778. [PMID: 40294084 PMCID: PMC12036950 DOI: 10.1085/jgp.202513778] [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: 02/07/2025] [Revised: 03/20/2025] [Accepted: 03/31/2025] [Indexed: 04/30/2025] Open
Abstract
Voltage-gated sodium channels underpin electrical signaling in sensory neurons. Their activity is an essential element in the vast majority of pain conditions, making them significant drug targets. Sensory neuron sodium channels play roles not only in afferent signaling but also in a range of efferent regulatory mechanisms. Side effects through actions on other cell types and efferent signaling are thus important issues to address during analgesic drug development. As an example, the human genetic evidence for NaV1.7 as an ideal pain target contrasts with the side effects of NaV1.7 antagonists. In this review, we describe the history and progress toward the development of useful analgesic drugs and the renewed focus on NaV1.8 as a key target in pain treatment. NaV1.8 antagonists alone or in combination with other analgesics are likely to provide new opportunities for pain relief for the vast number of people (about 33% of the population) impacted by chronic pain, particularly present in aging populations.
Collapse
Affiliation(s)
- John N. Wood
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, UCL, London, UK
| | - Nieng Yan
- Beijing Frontier Research Center for Biological Structures, State Key Laboratory of Membrane Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
- Institute of Bio-Architecture and Bio-Interactions (IBABI), Shenzhen Medical Academy of Research and Translation, Shenzhen, China
| | - Jian Huang
- Institute of Bio-Architecture and Bio-Interactions (IBABI), Shenzhen Medical Academy of Research and Translation, Shenzhen, China
| | - Jing Zhao
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, UCL, London, UK
| | - Armen Akopian
- Department of Endodontics, The School of the Dentistry, UTHSCSA, San Antonio, TX, USA
| | - James J. Cox
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, UCL, London, UK
| | | | - Mohammed A. Nassar
- School of Biosciences, Firth Court, University of Sheffield, Sheffield, UK
| |
Collapse
|
2
|
Theys M, De Waele J, Garud S, Willegems K, Van Petegem F, Bosmans F. A robust expression system reveals distinct gating mechanisms and calmodulin regulation of Na V1.9 channels. SCIENCE ADVANCES 2025; 11:eadt9799. [PMID: 40435262 PMCID: PMC12118636 DOI: 10.1126/sciadv.adt9799] [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: 10/18/2024] [Accepted: 04/22/2025] [Indexed: 06/01/2025]
Abstract
NaV1.9 is a voltage-gated Na+ channel subtype with unique gating properties that are poorly understood, partly due to the lack of reliable heterologous expression systems. Here, we present a transient expression protocol that produces robust mouse NaV1.9 currents, enabling direct electrophysiological comparisons with native dorsal root ganglion neurons. To further understand the low current density observed in human NaV1.9, we created chimeras with NaV1.5 and identified a role for the C-tail-specifically the IQ motif and EF-hand-in regulating current densities, likely due to a weak affinity for calmodulin. Isothermal titration calorimetry experiments indicated that, unlike other NaV channel subtypes, calmodulin binding to the C-tail is likely too weak to occur under physiological conditions. Markedly, the pre-IQ region did not influence channel expression but was responsible for conferring the characteristic depolarized voltage dependency of inactivation of NaV1.9. Our findings provide insights into the unique gating mechanisms of NaV1.9 and demonstrate the robustness of this platform for structure-function studies.
Collapse
Affiliation(s)
- Margaux Theys
- Molecular Physiology and Neurophysics group, Department of Basic and Applied Medical Sciences, Faculty of Medicine and Health Sciences, University of Ghent, Ghent 9000, Belgium
| | - Jolien De Waele
- Molecular Physiology and Neurophysics group, Department of Basic and Applied Medical Sciences, Faculty of Medicine and Health Sciences, University of Ghent, Ghent 9000, Belgium
| | - Sharang Garud
- Department of Biochemistry and Molecular Biology, Life Sciences Centre, University of British Columbia, Vancouver, BC, Canada
| | - Katrien Willegems
- Department of Biochemistry and Molecular Biology, Life Sciences Centre, University of British Columbia, Vancouver, BC, Canada
| | - Filip Van Petegem
- Department of Biochemistry and Molecular Biology, Life Sciences Centre, University of British Columbia, Vancouver, BC, Canada
| | - Frank Bosmans
- Molecular Physiology and Neurophysics group, Department of Basic and Applied Medical Sciences, Faculty of Medicine and Health Sciences, University of Ghent, Ghent 9000, Belgium
- Experimental Pharmacology group (EFAR), Department of Pharmaceutical Sciences, Faculty of Medicine and Pharmaceutical Sciences, Vrije Universiteit Brussel, Brussels 1050, Belgium
| |
Collapse
|
3
|
Zheng Y, Chen T, Vaissier Welborn V. Preorganized Electric Fields in Voltage-Gated Sodium Channels. Chembiochem 2025; 26:e202500314. [PMID: 40327008 PMCID: PMC12117427 DOI: 10.1002/cbic.202500314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2025] [Revised: 04/30/2025] [Accepted: 05/06/2025] [Indexed: 05/07/2025]
Abstract
Enzymes are reported to catalyze reactions by generating electric fields that promote the evolution of the reaction in the active site. Although seldom used outside enzymatic catalysis, electrostatic preorganization theory and language of electric fields can be generalized to other biological macromolecules. Herein, we performed molecular dynamics simulations of human Nav1.5, Nav1.6, and Nav1.7 with the atomic multipole optmimized energetics for biomolecular applications polarizable force field. We show that in the absence of an external potential, charged and uncharged residues generate strong electric fields that assist in Na+ motion in the pore. This work emphasizes the importance of charge-dipole interactions in modulating Na+ dynamics, in addition to charge-charge interactions, the focus of a majority of previous studies. Finally, we find that residues share a high level of mutual information through electric fields that can enable the optimization of allosteric pathways.
Collapse
Affiliation(s)
- Yi Zheng
- Department of ChemistryVirginia TechBlacksburgVA24061USA
- Macromolecules Innovation InstituteVirginia TechBlacksburgVA24061USA
| | - Taoyi Chen
- Department of ChemistryVirginia TechBlacksburgVA24061USA
- Macromolecules Innovation InstituteVirginia TechBlacksburgVA24061USA
| | - Valerie Vaissier Welborn
- Department of ChemistryVirginia TechBlacksburgVA24061USA
- Macromolecules Innovation InstituteVirginia TechBlacksburgVA24061USA
| |
Collapse
|
4
|
Ruiz-Castelan JE, Villa-Díaz F, Castro ME, Melendez FJ, Scior T. The α/β3 complex of human voltage-gated sodium channel hNa v1.7 to study mechanistic differences in presence and absence of auxiliary subunit β3. J Mol Model 2025; 31:168. [PMID: 40397258 PMCID: PMC12095431 DOI: 10.1007/s00894-025-06378-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 04/25/2025] [Indexed: 05/22/2025]
Abstract
CONTEXT In the context of structural interactomics, we generated a 3D model between α and β3 subunits for the hitherto unknown human voltage-gated sodium channel complex (hNa 1.7α/β3). We embedded our 3D model in a membrane lipid bilayer for molecular dynamics (MD) simulations of the sodium cation passage from the outer vestibule through the inner pore segment of our hNa 1.7 complex in presence and absence of auxiliary subunit β3 with remarkable changes close to electrophysiological study results. A complete passage could not be expected due to because the inactivated state of the underlying 3D template. A complete sodium ion passage would require an open state of the channel. The computed observations concerning side chain rearrangements for favorable cooperativity under evolutionary neighborhood conditions, favorable and unfavorable amino acid interactions, proline kink, loop, and helix displacements were all found in excellent keeping with the extant literature without any exception nor contradiction. Complex-stabilizing pairs of interacting amino acids with evolutionary neighborhood complementary were identified. METHODS The following tools were used: sequence search and alignment by FASTA and Clustal Omega; 3D model visualization and homology modeling by Vega ZZ, SPDBV, Chimera and Modeller, respectively; missing sections (loops) by Alphafold; geometry optimization prior to MD runs by GROMACS 2021.4 under the CHARMM 36 force field; local healing of bad contacts by SPDBV based on its Ramachandran plots; protein-protein docking by HDOCK 2.4; membrane insertion assisted by OPM; Berendsen V-rescaling for NVT; Parrinello-Rahman and Nose-Hoover for MPT; MD analyses by VMD and XMGRACE.
Collapse
Grants
- 100256733-VIEP2024 Vicerrectoría de Investigación y Estudios de Posgrado (VIEP-BUAP, Mexico)
- 100256733-VIEP2024 Vicerrectoría de Investigación y Estudios de Posgrado (VIEP-BUAP, Mexico)
- 100256733-VIEP2024 Vicerrectoría de Investigación y Estudios de Posgrado (VIEP-BUAP, Mexico)
- 100256733-VIEP2024 Vicerrectoría de Investigación y Estudios de Posgrado (VIEP-BUAP, Mexico)
- 100256733-VIEP2024 Vicerrectoría de Investigación y Estudios de Posgrado (VIEP-BUAP, Mexico)
- BUAP-CA-263 PRODEP Academic Group (SEP, Mexico)
- BUAP-CA-263 PRODEP Academic Group (SEP, Mexico)
- BUAP-CA-263 PRODEP Academic Group (SEP, Mexico)
- BUAP-CA-263 PRODEP Academic Group (SEP, Mexico)
- BUAP-CA-263 PRODEP Academic Group (SEP, Mexico)
Collapse
Affiliation(s)
| | - Fernando Villa-Díaz
- Laboratory of Basical Science, Tecnologico Nacional de Mexico, Campus Guaymas, C.P. 85480, Sonora, Mexico
| | | | - Francisco J Melendez
- Laboratory of Theoretical Chemistry, Faculty of Chemical Sciences, BUAP, C.P. 72570, Puebla, Mexico.
| | - Thomas Scior
- Laboratory of Computational Molecular Simulations, Faculty of Chemical Sciences, BUAP, C.P. 72570, Puebla, Mexico.
| |
Collapse
|
5
|
Biswas R, López-Serrano AL, Purohit A, Ramirez-Navarro A, Huang HL, Grandinetti G, Cheng X, Heissler SM, Deschênes I, Chinthalapudi K. Structural basis of human Na v1.5 gating mechanisms. Proc Natl Acad Sci U S A 2025; 122:e2416181122. [PMID: 40366698 PMCID: PMC12107192 DOI: 10.1073/pnas.2416181122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 04/14/2025] [Indexed: 05/15/2025] Open
Abstract
Voltage-gated Nav1.5 channels are central to the generation and propagation of cardiac action potentials. Aberrations in their function are associated with a wide spectrum of cardiac diseases including arrhythmias and heart failure. Despite decades of progress in Nav1.5 biology, the lack of structural insights into intracellular regions has hampered our understanding of its gating mechanisms. Here, we present two cryo-EM structures of human Nav1.5 in open states, revealing sequential conformational changes in gating charges of the voltage-sensing domains (VSDs) and several intracellular regions. Despite the channel being in the open state, these structures show repositioning, but no dislodging of the IFM motif in the receptor site. Molecular dynamics analyses show our structures with CTD conduct Na+ ions. Notably, our structural findings highlight a dynamic C-terminal domain (CTD) and III-IV linker interaction, which regulates the conformation of VSDs and pore opening. Electrophysiological studies confirm that disrupting this interaction alters fast inactivation of Nav1.5. Together, our structure-function studies establish a foundation for understanding the gating mechanisms of Nav1.5 and the mechanisms underlying CTD-related channelopathies.
Collapse
Affiliation(s)
- Rupam Biswas
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH43210
| | - Ana Laura López-Serrano
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH43210
- Frick Center for Heart Failure and Arrhythmia Research, The Ohio State University Wexner Medical Center, Columbus, OH43210
| | - Apoorva Purohit
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH43210
| | - Angelina Ramirez-Navarro
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH43210
- Frick Center for Heart Failure and Arrhythmia Research, The Ohio State University Wexner Medical Center, Columbus, OH43210
| | - Hsiang-Ling Huang
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH43210
| | - Giovanna Grandinetti
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH43210
- Center for Electron Microscopy and Analysis, College of Engineering, The Ohio State University, Columbus, OH43210
| | - Xiaolin Cheng
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH43210
- Translational Data Analytics Institute at The Ohio State University, Columbus, OH43210
| | - Sarah M. Heissler
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH43210
| | - Isabelle Deschênes
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH43210
- Frick Center for Heart Failure and Arrhythmia Research, The Ohio State University Wexner Medical Center, Columbus, OH43210
| | - Krishna Chinthalapudi
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH43210
| |
Collapse
|
6
|
Al-Kuraishy HM, Jabir MS, Al-Gareeb AI, Albuhadily AK, Klionsky DJ, Rafeeq MF. Epilepsy and autophagy modulators: a therapeutic split. Autophagy 2025. [PMID: 40375490 DOI: 10.1080/15548627.2025.2506292] [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: 06/27/2024] [Revised: 05/04/2025] [Accepted: 05/06/2025] [Indexed: 05/18/2025] Open
Abstract
Epilepsy is a neurological disease characterized by repeated unprovoked seizure. Epilepsy is controlled by anti-epileptic drugs (AEDs); however, one third of epileptic patients have symptoms that are not controlled by AEDs in a condition called refractory epilepsy. Dysregulation of macroautophagy/autophagy is involved in the pathogenesis of epilepsy. Autophagy prevents the development and progression of epilepsy through regulating the balance between inhibitory and excitatory neurotransmitters. Induction of autophagy and autophagy-related proteins could be a novel therapeutic strategy in the management of epilepsy. Despite the protective role of autophagy against epileptogenesis and epilepsy, its role in status epilepticus is perplexing and might reflect its nature as a double-edged sword. Autophagy inducers play a critical role in reducing seizure frequency and severity, and could be an adjuvant treatment in the management of epilepsy. However, autophagy inhibitors also have an anticonvulsant effect. Therefore, the aim of the present mini-review is to discuss the potential role of autophagy in the pathogenesis of epileptogenesis and epilepsy, and how autophagy modulators affect epileptogenesis and epilepsy.
Collapse
Affiliation(s)
- Hayder M Al-Kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, Mustansiriyah University, Baghdad, Iraq
| | - Majid S Jabir
- Department of Applied Science, University of Technology-Iraq, Baghdad, Iraq
| | | | - Ali K Albuhadily
- Department of Clinical Pharmacology and Medicine, College of Medicine, Mustansiriyah University, Baghdad, Iraq
| | | | - Mayyadah F Rafeeq
- Department of Applied Science, University of Technology-Iraq, Baghdad, Iraq
| |
Collapse
|
7
|
Ruiz-Alías G, Soldevila S, Altafaj X, Cordomí A, Olivella M. Missense variants pathogenicity annotation from homologous proteins. Bioinformatics 2025; 41:btaf305. [PMID: 40366734 PMCID: PMC12122210 DOI: 10.1093/bioinformatics/btaf305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 04/09/2025] [Accepted: 05/13/2025] [Indexed: 05/15/2025] Open
Abstract
MOTIVATION High-throughput DNA sequencing has revealed millions of single nucleotide variants (SNVs) in the human genome, with a small fraction linked to disease. The effect of missense variants, which alter the protein sequence, is particularly challenging to interpret due to the scarcity of clinical annotations and experimental information. While using conservation and structural information, current prediction tools still struggle to predict variant pathogenicity. In this study, we explored the pathogenicity of homologous missense variants-variants in equivalent positions across homologous proteins-focusing on proteins involved in autosomal dominant diseases. RESULTS Our analysis of 2976 pathogenic and 17 555 non-pathogenic homologous variants demonstrated that pathogenicity can be extrapolated with 95% accuracy within a family, or up to 98% for closer homologs. Remarkably, the evaluation of 27 commonly used mutation predictor methods revealed that they were not fully capturing this biological feature. To facilitate the exploration of homologous variants, we created HomolVar, a web server that computationally predicts the pathogenesis of missense variants using annotations from homologous variants, freely available at https://rarevariants.org/HomolVar. Overall, these findings and the accompanying tool offer a robust method for predicting the pathogenicity of unannotated variants, enhancing genotype-phenotype correlations, and contributing to diagnosing rare genetic disorders. AVAILABILITY AND IMPLEMENTATION HomolVar is freely available at https://rarevariants.org/HomolVar.
Collapse
Affiliation(s)
- Gabriel Ruiz-Alías
- Department of Biosciences, Faculty of Sciences and Technology, University of Vic-Central University of Catalonia, Vic, Barcelona 08500, Spain
- Institute for Research and Innovation in Life and Health Sciences (IRIS-CC), University of Vic-Central University of Catalonia, Vic, Barcelona 08500, Spain
| | - Sergi Soldevila
- Department of Biosciences, Faculty of Sciences and Technology, University of Vic-Central University of Catalonia, Vic, Barcelona 08500, Spain
- Institute for Research and Innovation in Life and Health Sciences (IRIS-CC), University of Vic-Central University of Catalonia, Vic, Barcelona 08500, Spain
| | - Xavier Altafaj
- Department of Biomedicine, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, Barcelona 08036, Spain
- Agustí Pi i Sunyer Biomedical Research Institute (IDIBAPS), University of Barcelona, Barcelona 08036, Spain
| | - Arnau Cordomí
- Department of Biochemistry and Molecular Biology, Faculty of Biosciences, Universitat Autònoma de Barcelona (UAB), Barcelona 08193, Spain
| | - Mireia Olivella
- Department of Biosciences, Faculty of Sciences and Technology, University of Vic-Central University of Catalonia, Vic, Barcelona 08500, Spain
- Institute for Research and Innovation in Life and Health Sciences (IRIS-CC), University of Vic-Central University of Catalonia, Vic, Barcelona 08500, Spain
| |
Collapse
|
8
|
Neureiter EG, Erickson-Oberg MQ, Nigam A, Johnson JW. Inhibition of NMDA receptors and other ion channel types by membrane-associated drugs. Front Pharmacol 2025; 16:1561956. [PMID: 40371334 PMCID: PMC12075551 DOI: 10.3389/fphar.2025.1561956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Accepted: 04/15/2025] [Indexed: 05/16/2025] Open
Abstract
N-methyl-D-aspartate receptors (NMDARs) are ligand-gated ion channels present at most excitatory synapses in the brain that play essential roles in cognitive functions including learning and memory consolidation. However, NMDAR dysregulation is implicated in many nervous system disorders. Diseases that involve pathological hyperactivity of NMDARs can be treated clinically through inhibition by channel blocking drugs. NMDAR channel block can occur via two known mechanisms. First, in traditional block, charged drug molecules can enter the channel directly from the extracellular solution after NMDAR activation and channel opening. Second, uncharged molecules of channel blocking drug can enter the hydrophobic plasma membrane, and upon NMDAR activation the membrane-associated drug can transit into the channel through a fenestration within the NMDAR. This membrane-associated mechanism of action is called membrane to channel inhibition (MCI) and is not well understood despite the clinical importance of NMDAR channel blocking drugs. Intriguingly, a hydrophobic route of access for drugs is not unique to NMDARs. Our review will address inhibition of NMDARs and other ion channels by membrane-associated drugs and consider how the path of access may affect a drug's therapeutic potential.
Collapse
Affiliation(s)
| | | | | | - Jon W. Johnson
- Department of Neuroscience and Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
9
|
Fukuoka R, Yano Y, Hara N, Sadamoto C, Maturana AD, Kita M. Hyperpolarization Modulation of the T-Type hCa v3.2 Channel by Human Synenkephalin [1-53], a Shrew Neurotoxin Analogue without Paralytic Effects. Angew Chem Int Ed Engl 2025:e202503891. [PMID: 40274533 DOI: 10.1002/anie.202503891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 04/24/2025] [Accepted: 04/24/2025] [Indexed: 04/26/2025]
Abstract
Mammalian secreted venoms mainly consist of peptides and proteases used for defense or predation. Blarina paralytic peptides (BPPs), mealworm-targeting neurotoxins from shrew, are very similar to human synenkephalin. This peptide is released from proenkephalin in the brain along with opioid peptides that mediate analgesic and antidepressant effects, though its physiological function is unclear. Here, we synthesized and characterized human synenkephalin [1-53] (hSYN) and reveal its disulfide bond connectivity. Similar to BPP2, hSYN caused a hyperpolarizing shift in the human T-type voltage-gated calcium channel (hCav3.2) at 0.74 µM, but did not paralyze mealworms. Molecular docking and molecular dynamics simulations showed that hSYN and BPP2 interact with hCav3.2 channel differently, due to differences in polar residues. Since Cav3.2 channel regulates neuronal excitability and is implicated in conditions like autism and epilepsy, our findings on hSYN could provide insight into the channel gating and agonistic mechanisms, along with potential pathways for developing treatments for neurological disorders.
Collapse
Affiliation(s)
- Ryo Fukuoka
- Department of Applied Biosciences, Graduate School of Bioagricultural Sciences, Nagoya University, Furo-cho, Chikusa, Nagoya, 464-8601, Japan
| | - Yusuke Yano
- Department of Applied Biosciences, Graduate School of Bioagricultural Sciences, Nagoya University, Furo-cho, Chikusa, Nagoya, 464-8601, Japan
| | - Nozomi Hara
- Department of Applied Biosciences, Graduate School of Bioagricultural Sciences, Nagoya University, Furo-cho, Chikusa, Nagoya, 464-8601, Japan
| | - Chihiro Sadamoto
- Department of Applied Biosciences, Graduate School of Bioagricultural Sciences, Nagoya University, Furo-cho, Chikusa, Nagoya, 464-8601, Japan
| | - Andres D Maturana
- Department of Applied Biosciences, Graduate School of Bioagricultural Sciences, Nagoya University, Furo-cho, Chikusa, Nagoya, 464-8601, Japan
| | - Masaki Kita
- Department of Applied Biosciences, Graduate School of Bioagricultural Sciences, Nagoya University, Furo-cho, Chikusa, Nagoya, 464-8601, Japan
- Promotion Office for Open Innovation, Institutes of Innovation for Future Society, Nagoya University, Furo-cho, Chikusa, Nagoya, 464-8601, Japan
| |
Collapse
|
10
|
Zhou X, Chen H, Peng S, Si Y, Wang G, Yang L, Zhou Q, Lu M, Xie Q, He X, Wu M, Xiao X, Luo X, Feng X, Wang W, Luo S, Li Y, Qin J, Chen M, Zhang Q, Hu W, Liang S, Hou T, Liu Z. Inherent fast inactivation particle of Nav channels as a new binding site for a neurotoxin. EMBO J 2025:10.1038/s44318-025-00438-9. [PMID: 40263599 DOI: 10.1038/s44318-025-00438-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 03/01/2025] [Accepted: 03/04/2025] [Indexed: 04/24/2025] Open
Abstract
Neurotoxins derived from animal venoms are indispensable tools for probing the structure and function of voltage-gated sodium (Nav) channels. Utilizing a novel centipede peptide toxin called rpTx1, we show that the "inherent inactivation particle" of Nav channels represents a binding site for a neurotoxin. The toxin comprises two functional domains: one for cell penetration and one for modulating Nav channel activity. After crossing the cell membrane, rpTx1 preferentially binds to and stabilizes the IFMT motif (the conserved core region of the fast inactivation particle in mammalian Nav channels) in the unbound state, preventing this motif from associating with its receptor site and thereby inhibiting the fast inactivation of Nav channels. This competition between rpTx1 and the receptor site for interacting with the IFMT motif may account for the higher activity of rpTx1 on Nav1.8 than on other Nav subtypes, given the weaker relative affinity between the receptor site and the IFMT motif of Nav1.8. Overall, this study should promote the investigation of the intracellular modulation of Nav channels by neurotoxins.
Collapse
Affiliation(s)
- Xi Zhou
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, China.
- Peptide and Small Molecule Drug R&D Platform, Furong Laboratory, Hunan Normal University, Changsha, 410081, Hunan, China.
- Institute of Interdisciplinary Studies, Hunan Normal University, Changsha, 410081, China.
| | - Haiyi Chen
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou, Zhejiang, 310058, China
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
| | - Shuijiao Peng
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, China
- Peptide and Small Molecule Drug R&D Platform, Furong Laboratory, Hunan Normal University, Changsha, 410081, Hunan, China
- Institute of Interdisciplinary Studies, Hunan Normal University, Changsha, 410081, China
- Hunan Provincial Key Laboratory of Microbial Molecular Biology, Hunan Provincial Center for Disease Control and Prevention, Changsha 410000, China
| | - Yuxin Si
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, China
- Peptide and Small Molecule Drug R&D Platform, Furong Laboratory, Hunan Normal University, Changsha, 410081, Hunan, China
- Institute of Interdisciplinary Studies, Hunan Normal University, Changsha, 410081, China
| | - Gaoang Wang
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Li Yang
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, China
- Peptide and Small Molecule Drug R&D Platform, Furong Laboratory, Hunan Normal University, Changsha, 410081, Hunan, China
- Institute of Interdisciplinary Studies, Hunan Normal University, Changsha, 410081, China
| | - Qing Zhou
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, China
- Peptide and Small Molecule Drug R&D Platform, Furong Laboratory, Hunan Normal University, Changsha, 410081, Hunan, China
- Institute of Interdisciplinary Studies, Hunan Normal University, Changsha, 410081, China
| | - Minjuan Lu
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, China
- Peptide and Small Molecule Drug R&D Platform, Furong Laboratory, Hunan Normal University, Changsha, 410081, Hunan, China
- Institute of Interdisciplinary Studies, Hunan Normal University, Changsha, 410081, China
| | - Qiaoling Xie
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, China
- Peptide and Small Molecule Drug R&D Platform, Furong Laboratory, Hunan Normal University, Changsha, 410081, Hunan, China
- Institute of Interdisciplinary Studies, Hunan Normal University, Changsha, 410081, China
| | - Xi He
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, China
- Peptide and Small Molecule Drug R&D Platform, Furong Laboratory, Hunan Normal University, Changsha, 410081, Hunan, China
- Institute of Interdisciplinary Studies, Hunan Normal University, Changsha, 410081, China
| | - Meijing Wu
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, China
- Peptide and Small Molecule Drug R&D Platform, Furong Laboratory, Hunan Normal University, Changsha, 410081, Hunan, China
- Institute of Interdisciplinary Studies, Hunan Normal University, Changsha, 410081, China
| | - Xin Xiao
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, China
- Peptide and Small Molecule Drug R&D Platform, Furong Laboratory, Hunan Normal University, Changsha, 410081, Hunan, China
- Institute of Interdisciplinary Studies, Hunan Normal University, Changsha, 410081, China
| | - Xiaoqing Luo
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, China
- Peptide and Small Molecule Drug R&D Platform, Furong Laboratory, Hunan Normal University, Changsha, 410081, Hunan, China
- Institute of Interdisciplinary Studies, Hunan Normal University, Changsha, 410081, China
| | - Xujun Feng
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, China
- Peptide and Small Molecule Drug R&D Platform, Furong Laboratory, Hunan Normal University, Changsha, 410081, Hunan, China
- Institute of Interdisciplinary Studies, Hunan Normal University, Changsha, 410081, China
| | - Wenxing Wang
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, China
- Peptide and Small Molecule Drug R&D Platform, Furong Laboratory, Hunan Normal University, Changsha, 410081, Hunan, China
- Institute of Interdisciplinary Studies, Hunan Normal University, Changsha, 410081, China
| | - Sen Luo
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, China
- Peptide and Small Molecule Drug R&D Platform, Furong Laboratory, Hunan Normal University, Changsha, 410081, Hunan, China
- Institute of Interdisciplinary Studies, Hunan Normal University, Changsha, 410081, China
| | - Yaqi Li
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, China
- Peptide and Small Molecule Drug R&D Platform, Furong Laboratory, Hunan Normal University, Changsha, 410081, Hunan, China
- Institute of Interdisciplinary Studies, Hunan Normal University, Changsha, 410081, China
| | - Jiaxin Qin
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, China
- Peptide and Small Molecule Drug R&D Platform, Furong Laboratory, Hunan Normal University, Changsha, 410081, Hunan, China
- Institute of Interdisciplinary Studies, Hunan Normal University, Changsha, 410081, China
| | - Minzhi Chen
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, China
- Peptide and Small Molecule Drug R&D Platform, Furong Laboratory, Hunan Normal University, Changsha, 410081, Hunan, China
- Institute of Interdisciplinary Studies, Hunan Normal University, Changsha, 410081, China
| | - Qianqian Zhang
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, China
- Peptide and Small Molecule Drug R&D Platform, Furong Laboratory, Hunan Normal University, Changsha, 410081, Hunan, China
- Institute of Interdisciplinary Studies, Hunan Normal University, Changsha, 410081, China
| | - Weijun Hu
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, China
- Peptide and Small Molecule Drug R&D Platform, Furong Laboratory, Hunan Normal University, Changsha, 410081, Hunan, China
- Institute of Interdisciplinary Studies, Hunan Normal University, Changsha, 410081, China
| | - Songping Liang
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, China
- Peptide and Small Molecule Drug R&D Platform, Furong Laboratory, Hunan Normal University, Changsha, 410081, Hunan, China
- Institute of Interdisciplinary Studies, Hunan Normal University, Changsha, 410081, China
| | - Tingjun Hou
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou, Zhejiang, 310058, China.
| | - Zhonghua Liu
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, China.
- Peptide and Small Molecule Drug R&D Platform, Furong Laboratory, Hunan Normal University, Changsha, 410081, Hunan, China.
- Institute of Interdisciplinary Studies, Hunan Normal University, Changsha, 410081, China.
| |
Collapse
|
11
|
Dong Z, Xiong B, Sun T, Jiang R, Feng F, Sun H. Brief analysis of Na v1.7 inhibitors: Mechanism of action and new research trends. Bioorg Med Chem 2025; 128:118180. [PMID: 40403415 DOI: 10.1016/j.bmc.2025.118180] [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: 02/01/2025] [Revised: 03/12/2025] [Accepted: 04/01/2025] [Indexed: 05/24/2025]
Abstract
Nav1.7 has been the most studied ion channel among the 9 subtypes of sodium ion, and it is also one of the popular analgesic targets in recent years. Compared with opioid receptors, because of its advantages of targeting a variety of pain types and being unrelated to addiction, many related inhibitors have been excavated for it, including old drugs and new uses, peptides, and new skeleton small molecules. Some of these inhibitors have reached the second phase of clinical research, and some are still in the laboratory research stage. So far, no exclusive Nav1.7 inhibitor has successfully passed the third phase of clinical research and entered the field of vision of patients. This article reviews the action sites and mechanisms of different Nav1.7 inhibitors in terms of historical background and related analgesic activities, and also summarizes the related inhibitors that are currently under active development, hoping to provide useful information for the research of new Nav1.7 inhibitors.
Collapse
Affiliation(s)
- Zhoujun Dong
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, China
| | - Baichen Xiong
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Tianyu Sun
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Ruijia Jiang
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Feng Feng
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, China.
| | - Haopeng Sun
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China.
| |
Collapse
|
12
|
Akiyama C, Sakata S, Ono F. Normal locomotion in zebrafish lacking the sodium channel NaV1.4 suggests that the need for muscle action potentials is not universal. PLoS Biol 2025; 23:e3003137. [PMID: 40273189 PMCID: PMC12021243 DOI: 10.1371/journal.pbio.3003137] [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: 02/05/2025] [Accepted: 04/01/2025] [Indexed: 04/26/2025] Open
Abstract
Extensive studies over decades have firmly established the concept that action potentials (APs) in muscles are indispensable for muscle contraction. To re-examine the significance of APs, we generated zebrafish lacking APs by editing the scn4aa and scn4ab genes, which together encode NaV1.4 (NaVDKO), using the CRISPR-Cas9 system. Surprisingly, the escape response of NaVDKOs to tactile stimuli, both in the embryonic and adult stages, was indistinguishable from that of wild-type (WT) fish. Ca2+ imaging using the calcium indicator protein GCaMP revealed that myofibers isolated from WT fish could be excited by the application of acetylcholine (ACh), even in the presence of tetrodotoxin (TTX) indicating that NaVs are dispensable for skeletal muscle contraction in zebrafish. Mathematical simulations showed that the end-plate potential was able to elicit a change in membrane potential large enough to activate the dihydropyridine receptors of the entire muscle fiber owing to the small fiber size and the disseminated distribution of neuromuscular synapses in both adults and embryos. Our data demonstrate that NaVs are not essential for muscle contraction in zebrafish and that the physiological significance of NaV1.4 in muscle is not uniform across vertebrates.
Collapse
Affiliation(s)
- Chifumi Akiyama
- Department of Physiology, Division of Life Sciences, Faculty of Medicine, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Souhei Sakata
- Department of Physiology, Division of Life Sciences, Faculty of Medicine, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Fumihito Ono
- Department of Physiology, Division of Life Sciences, Faculty of Medicine, Osaka Medical and Pharmaceutical University, Osaka, Japan
| |
Collapse
|
13
|
Elhanafy E, Akbari Ahangar A, Roth R, Gamal El-Din TM, Bankston JR, Li J. The differential impacts of equivalent gating-charge mutations in voltage-gated sodium channels. J Gen Physiol 2025; 157:e202413669. [PMID: 39820972 PMCID: PMC11740781 DOI: 10.1085/jgp.202413669] [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: 09/08/2024] [Revised: 11/27/2024] [Accepted: 12/25/2024] [Indexed: 01/19/2025] Open
Abstract
Voltage-gated sodium (Nav) channels are pivotal for cellular signaling, and mutations in Nav channels can lead to excitability disorders in cardiac, muscular, and neural tissues. A major cluster of pathological mutations localizes in the voltage-sensing domains (VSDs), resulting in either gain-of-function, loss-of-function effects, or both. However, the mechanism behind this functional diversity of mutations at equivalent positions remains elusive. Through hotspot analysis, we identified three gating charges (R1, R2, and R3) as major mutational hotspots in VSDs. The same amino acid substitutions at equivalent gating-charge positions in VSDI and VSDII of the cardiac sodium channel Nav1.5 show differential gating property impacts in electrophysiology measurements. We conducted molecular dynamics (MD) simulations on wild-type channels and six mutants to elucidate the structural basis of their differential impacts. Our 120-µs MD simulations with applied external electric fields captured VSD state transitions and revealed the differential structural dynamics between equivalent R-to-Q mutants. Notably, we observed transient leaky conformations in some mutants during structural transitions, offering a detailed structural explanation for gating-pore currents. Our salt-bridge network analysis uncovered VSD-specific and state-dependent interactions among gating charges, countercharges, and lipids. This detailed analysis revealed how mutations disrupt critical electrostatic interactions, thereby altering VSD permeability and modulating gating properties. By demonstrating the crucial importance of considering the specific structural context of each mutation, our study advances our understanding of structure-function relationships in Nav channels. Our work establishes a robust framework for future investigations into the molecular basis of ion channel-related disorders.
Collapse
Affiliation(s)
- Eslam Elhanafy
- Department of Biomolecular Sciences, School of Pharmacy, University of Mississippi, Oxford, MS, USA
| | - Amin Akbari Ahangar
- Department of Biomolecular Sciences, School of Pharmacy, University of Mississippi, Oxford, MS, USA
| | - Rebecca Roth
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | | | - John R. Bankston
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Jing Li
- Department of Biomolecular Sciences, School of Pharmacy, University of Mississippi, Oxford, MS, USA
| |
Collapse
|
14
|
Jorgensen C. Understanding the role of mutations in voltage-gated sodium ion channels for cardiovascular disorders. J Gen Physiol 2025; 157:e202413744. [PMID: 39846865 PMCID: PMC11756374 DOI: 10.1085/jgp.202413744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2025] Open
Abstract
Elhanafy et al. used Molecular Dynamics simulations and electrophysiology to show how identical mutations in the volgage sending domain of sodium channels can yield differential functional effects.
Collapse
Affiliation(s)
- Christian Jorgensen
- School of Medicine, Pharmacy and Biomedical Sciences, Faculty of Science & Health, University of Portsmouth, Portsmouth, UK
| |
Collapse
|
15
|
Peng S, Chen M, Wu M, Liu Z, Tang D, Zhou X. Elucidating the roles of voltage sensors in Na V1.9 activation and inactivation through a spider toxin. Biochim Biophys Acta Gen Subj 2025; 1869:130762. [PMID: 39800272 DOI: 10.1016/j.bbagen.2025.130762] [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: 08/23/2024] [Revised: 12/18/2024] [Accepted: 01/06/2025] [Indexed: 01/15/2025]
Abstract
The gating process of voltage-gated sodium (NaV) channels is extraordinary intrinsic and involves numerous factors, such as voltage-sensing domain (VSD), the N-terminus and C-terminus, and the auxiliary subunits. To date, the gating mechanism of NaV channel has not been clearly elucidated. NaV1.9 has garnered significant attention due to its slow gating kinetics. Due to the challenges of NaV1.9 heterologous expression, research on its gating mechanism is relatively limited. Whether there are any differences in the functions of the four VSDs in NaV1.9 compared to those in other subtypes remains an open question. Here, we employed the established chimera method to transplant the S3b-S4 motif from the VSDIV of the toxin-sensitive donor channel (NaV1.9) into the receptor channel (NaV1.9/1.8 DIV S3b-S4 chimera). This modification imparted animal toxin sensitivity to the other three VSDs. Our results demonstrate that all four VSDs of NaV1.9 are involved in channel opening, VSDIII and VSDIV are primarily involved in regulating fast inactivation, and VSDII also regulates the steady-state inactivation of channels. These findings provide a new insight into the gating mechanism of NaV1.9.
Collapse
Affiliation(s)
- Shuijiao Peng
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China; Hunan Provincial Center for Disease Control and Prevention, Hunan Provincial Key Laboratory of Microbial Molecular Biology, Changsha 410000, Hunan, China
| | - Minzhi Chen
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China; Peptide and small molecule drug R&D platform, Furong Laboratory, Hunan Normal University, Changsha 410081, Hunan, China; Institute of Interdisciplinary Studies, Hunan Normal University, Changsha 410081, Hunan, China
| | - Meijing Wu
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China; Peptide and small molecule drug R&D platform, Furong Laboratory, Hunan Normal University, Changsha 410081, Hunan, China; Institute of Interdisciplinary Studies, Hunan Normal University, Changsha 410081, Hunan, China
| | - Zhonghua Liu
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China; Peptide and small molecule drug R&D platform, Furong Laboratory, Hunan Normal University, Changsha 410081, Hunan, China; Institute of Interdisciplinary Studies, Hunan Normal University, Changsha 410081, Hunan, China
| | - Dongfang Tang
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China; College of Chemistry and Bioengineering, Hunan University of Science and Engineering, Yongzhou, 425199, Hunan, China.
| | - Xi Zhou
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China; Peptide and small molecule drug R&D platform, Furong Laboratory, Hunan Normal University, Changsha 410081, Hunan, China; Institute of Interdisciplinary Studies, Hunan Normal University, Changsha 410081, Hunan, China.
| |
Collapse
|
16
|
Neumann B, McCarthy S, Gonen S. Structural basis of inhibition of human Na V1.8 by the tarantula venom peptide Protoxin-I. Nat Commun 2025; 16:1459. [PMID: 39920100 PMCID: PMC11805909 DOI: 10.1038/s41467-024-55764-z] [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: 08/28/2024] [Accepted: 12/24/2024] [Indexed: 02/09/2025] Open
Abstract
Voltage-gated sodium channels (NaVs) selectively permit diffusion of sodium ions across the cell membrane and, in excitable cells, are responsible for propagating action potentials. One of the nine human NaV isoforms, NaV1.8, is a promising target for analgesics, and selective inhibitors are of interest as therapeutics. One such inhibitor, the gating-modifier peptide Protoxin-I derived from tarantula venom, blocks channel opening by shifting the activation voltage threshold to more depolarized potentials, but the structural basis for this inhibition has not previously been determined. Using monolayer graphene grids, we report the cryogenic electron microscopy structures of full-length human apo-NaV1.8 and the Protoxin-I-bound complex at 3.1 Å and 2.8 Å resolution, respectively. The apo structure shows an unexpected movement of the Domain I S4-S5 helix, and VSDI was unresolvable. We find that Protoxin-I binds to and displaces the VSDII S3-S4 linker, hindering translocation of the S4II helix during activation.
Collapse
Affiliation(s)
- Bryan Neumann
- Department of Molecular Biology and Biochemistry, University of California Irvine, Irvine, CA, USA
| | - Stephen McCarthy
- Department of Molecular Biology and Biochemistry, University of California Irvine, Irvine, CA, USA
| | - Shane Gonen
- Department of Molecular Biology and Biochemistry, University of California Irvine, Irvine, CA, USA.
| |
Collapse
|
17
|
Jan L. Voltage sensors. Mol Pharmacol 2025; 107:100011. [PMID: 40023511 DOI: 10.1016/j.molpha.2024.100011] [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: 08/01/2024] [Accepted: 11/06/2024] [Indexed: 03/04/2025] Open
Abstract
Widely distributed in all kingdoms of life, voltage sensors in the membrane serve important functions via their movements driven by changes in voltage across the membrane (membrane potential). A voltage sensor domain contains 4 transmembrane segments (S1-S4). The S1-S3 helices form a hydrophobic constriction site (HCS, also known as the gating charge transfer center) that spans roughly one-third of the membrane thickness. Flanked by aqueous vestibules connected to the extracellular solution above the HCS or cytoplasmic solution below the HCS, the HCS forms a gating pore for the S4 segment bearing multiple basic residues. Membrane potential changes cause S4 to move through the HCS in a 310 helical conformation. This S4 translocation generates a gating current as the positively charged S4 basic residues traverse the membrane electric field, transferring these gating charges from one aqueous vestibule to the other. For voltage-gated ion channels with their voltage sensor domains connected to pore domains, the HCS in the voltage sensor domain allows S4 but not ions to go through, while the channel pore formed by the pore domains mediates ion permeation. Voltage sensor mutations could result in ω currents that are conducted through the gating pore of mutant voltage-gated ion channels. These ω currents may cause pathological consequences in patients with periodic paralysis. Besides voltage-gated ion channels, the sperm-specific Na+/H+ exchanger and voltage-sensing phosphatases contain voltage sensors for membrane potential regulation. Notably, voltage-gated proton channels that are important for pH homeostasis are formed solely by the voltage sensor domain, which mediates proton permeation. SIGNIFICANCE STATEMENT: Voltage sensors mediate voltage regulation of ion channels, transporters, and phosphatases. The voltage sensor domain composed of 4 transmembrane segments (S1-S4) focuses the membrane electric field to the hydrophobic constriction site. To mediate voltage regulation, S4 basic residues within a 310 helix move across the hydrophobic constriction site without concurrent ion flow through this gating pore. As a counterexample, voltage-gated proton channels are formed by the voltage sensor to mediate proton permeation. These ingeniously engineered voltage sensors are conserved throughout evolution.
Collapse
Affiliation(s)
- Lily Jan
- Departments of Physiology, Biochemistry and Biophysics, Howard Hughes Medical Institute, University of California, San Francisco, California.
| |
Collapse
|
18
|
Lei D, Wang Y, Zhang Q, Wang S, Jiang L, Zhang Z. High-performance solid-state proton gating membranes based on two-dimensional hydrogen-bonded organic framework composites. Nat Commun 2025; 16:754. [PMID: 39819979 PMCID: PMC11739393 DOI: 10.1038/s41467-025-56228-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 01/13/2025] [Indexed: 01/19/2025] Open
Abstract
Biological ion channels exhibit strong gating effects due to their zero-current closed states. However, the gating capabilities of artificial nanochannels have typically fallen short of biological channels, primarily owing to the larger nanopores that fail to completely block ion transport in the off-states. Here, we demonstrate solid-state hydrogen-bonded organic frameworks-based membranes to achieve high-performance ambient humidity-controlled proton gating, accomplished by switching the proton transport pathway instead of relying on conventional ion blockage/activation effects. Density functional theory calculations reveal that the reversible formation and disruption of humidity-induced water bridges within the frameworks facilitates the switching of proton transport mode from the adsorption site hopping to the Grotthuss mechanism. This transition, coupled with the introduction of bacterial cellulose to enhance desorption/adsorption of water clusters, enables us to achieve a superior proton gating ratio of up to 5740, surpassing state-of-the-art solid-state gating devices. Moreover, the developed membrane operates entirely on solid-state principles, rendering it highly versatile for a myriad of applications from environmental detection to human health monitoring. This study offers perspectives for the design of efficient proton gating systems.
Collapse
Affiliation(s)
- Dandan Lei
- Key Laboratory of Precision and Intelligent Chemistry, Department of Applied Chemistry, School of Chemistry and Materials Science, University of Science and Technology of China, Hefei, Anhui, China
- Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou, Jiangsu, China
| | - Yixiang Wang
- Key Laboratory of Precision and Intelligent Chemistry, Department of Applied Chemistry, School of Chemistry and Materials Science, University of Science and Technology of China, Hefei, Anhui, China
- Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou, Jiangsu, China
| | - Qixiang Zhang
- Key Laboratory of Precision and Intelligent Chemistry, Department of Applied Chemistry, School of Chemistry and Materials Science, University of Science and Technology of China, Hefei, Anhui, China
- Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou, Jiangsu, China
- School of Physics and Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shuqi Wang
- Key Laboratory of Precision and Intelligent Chemistry, Department of Applied Chemistry, School of Chemistry and Materials Science, University of Science and Technology of China, Hefei, Anhui, China
- Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou, Jiangsu, China
| | - Lei Jiang
- Key Laboratory of Precision and Intelligent Chemistry, Department of Applied Chemistry, School of Chemistry and Materials Science, University of Science and Technology of China, Hefei, Anhui, China
- Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou, Jiangsu, China
- CAS Key Laboratory of Bio-inspired Materials and Interfacial Science, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, China
| | - Zhen Zhang
- Key Laboratory of Precision and Intelligent Chemistry, Department of Applied Chemistry, School of Chemistry and Materials Science, University of Science and Technology of China, Hefei, Anhui, China.
- Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou, Jiangsu, China.
| |
Collapse
|
19
|
Berecki G, Tao E, Howell KB, Coorg RK, Andersen E, Kahlig K, Wolff M, Corry B, Petrou S. Nav1.2 channel mutations preventing fast inactivation lead to SCN2A encephalopathy. Brain 2025; 148:212-226. [PMID: 38939966 PMCID: PMC11706276 DOI: 10.1093/brain/awae213] [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: 03/06/2024] [Revised: 05/29/2024] [Accepted: 06/11/2024] [Indexed: 06/29/2024] Open
Abstract
SCN2A gene-related early-infantile developmental and epileptic encephalopathy (EI-DEE) is a rare and severe disorder that manifests in early infancy. SCN2A mutations affecting the fast inactivation gating mechanism can result in altered voltage dependence and incomplete inactivation of the encoded neuronal Nav1.2 channel and lead to abnormal neuronal excitability. In this study, we evaluated clinical data of seven missense Nav1.2 variants associated with DEE and performed molecular dynamics simulations, patch-clamp electrophysiology and dynamic clamp real-time neuronal modelling to elucidate the molecular and neuron-scale phenotypic consequences of the mutations. The N1662D mutation almost completely prevented fast inactivation without affecting activation. The comparison of wild-type and N1662D channel structures suggested that the ambifunctional hydrogen bond formation between residues N1662 and Q1494 is essential for fast inactivation. Fast inactivation could also be prevented with engineered Q1494A or Q1494L Nav1.2 channel variants, whereas Q1494E or Q149K variants resulted in incomplete inactivation and persistent current. Molecular dynamics simulations revealed a reduced affinity of the hydrophobic IFM-motif to its receptor site with N1662D and Q1494L variants relative to wild-type. These results demonstrate that the interactions between N1662 and Q1494 underpin the stability and the orientation of the inactivation gate and are essential for the development of fast inactivation. Six DEE-associated Nav1.2 variants, with mutations mapped to channel segments known to be implicated in fast inactivation were also evaluated. Remarkably, the L1657P variant also prevented fast inactivation and produced biophysical characteristics that were similar to those of N1662D, whereas the M1501V, M1501T, F1651C, P1658S and A1659V variants resulted in biophysical properties that were consistent with gain-of-function and enhanced action potential firing of hybrid neurons in dynamic action potential clamp experiments. Paradoxically, low densities of N1662D or L1657P currents potentiated action potential firing, whereas increased densities resulted in sustained depolarization. Our results provide novel structural insights into the molecular mechanism of Nav1.2 channel fast inactivation and inform treatment strategies for SCN2A-related EI-DEE. The contribution of non-inactivating Nav1.2 channels to neuronal excitability may constitute a distinct cellular mechanism in the pathogenesis of SCN2A-related DEE.
Collapse
Affiliation(s)
- Géza Berecki
- Ion Channels and Human Disease Group, The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC 3052, Australia
- Department of the Florey Institute, University of Melbourne, Parkville, VIC 3050, Australia
| | - Elaine Tao
- Division of Biomedical Science and Biochemistry, Research School of Biology, Australian National University, Canberra, ACT 2601, Australia
| | - Katherine B Howell
- Department of Neurology, Royal Children’s Hospital, Parkville, VIC 3052, Australia
- Neuroscience, Murdoch Children’s Research Institute, Parkville, VIC 3052, Australia
| | - Rohini K Coorg
- Division of Neurology and Developmental Neuroscience, Department of Pediatrics, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030, USA
| | - Erik Andersen
- Department of Paediatrics and Child Health, University of Otago, Wellington 6242, New Zealand
| | - Kris Kahlig
- Praxis Precision Medicines, Inc., Cambridge, MA 02142, USA
| | - Markus Wolff
- Swiss Epilepsy Center, Klinik Lengg, Zürich 8001, Switzerland
| | - Ben Corry
- Division of Biomedical Science and Biochemistry, Research School of Biology, Australian National University, Canberra, ACT 2601, Australia
| | - Steven Petrou
- Ion Channels and Human Disease Group, The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC 3052, Australia
- Department of the Florey Institute, University of Melbourne, Parkville, VIC 3050, Australia
- Praxis Precision Medicines, Inc., Cambridge, MA 02142, USA
| |
Collapse
|
20
|
Short B. A misstep in the multistep process of fast inactivation. J Gen Physiol 2025; 157:e202413735. [PMID: 39688837 DOI: 10.1085/jgp.202413735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2024] Open
Abstract
JGP study (Liu and Bezanilla. https://doi.org/10.1085/jgp.202413667) reveals that a sodium channel mutant blocks fast inactivation downstream of inactivation particle binding, diverting the channel into an alternative open state.
Collapse
Affiliation(s)
- Ben Short
- Science Writer, Rockefeller University Press, New York, NY, USA
| |
Collapse
|
21
|
Beaudoin CA, Kohli M, Salvage SC, Liu H, Arundel SJ, Hamaia SW, Lei M, Huang CLH, Jackson AP. Isoform-specific N-linked glycosylation of NaV channel α-subunits alters β-subunit binding sites. J Gen Physiol 2025; 157:e202413609. [PMID: 39680039 PMCID: PMC11666101 DOI: 10.1085/jgp.202413609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 09/19/2024] [Accepted: 11/25/2024] [Indexed: 12/17/2024] Open
Abstract
Voltage-gated sodium channel α-subunits (NaV1.1-1.9) initiate and propagate action potentials in neurons and myocytes. The NaV β-subunits (β1-4) have been shown to modulate α-subunit properties. Homo-oligomerization of β-subunits on neighboring or opposing plasma membranes has been suggested to facilitate cis or trans interactions, respectively. The interactions between several NaV channel isoforms and β-subunits have been determined using cryogenic electron microscopy (cryo-EM). Interestingly, the NaV cryo-EM structures reveal the presence of N-linked glycosylation sites. However, only the first glycan moieties are typically resolved at each site due to the flexibility of mature glycan trees. Thus, existing cryo-EM structures may risk de-emphasizing the structural implications of glycans on the NaV channels. Herein, molecular modeling and all-atom molecular dynamics simulations were applied to investigate the conformational landscape of N-linked glycans on NaV channel surfaces. The simulations revealed that negatively charged sialic acid residues of two glycan sites may interact with voltage-sensing domains. Notably, two NaV1.5 isoform-specific glycans extensively cover the α-subunit region that, in other NaV channel α-subunit isoforms, corresponds to the binding site for the β1- (and likely β3-) subunit immunoglobulin (Ig) domain. NaV1.8 contains a unique N-linked glycosylation site that likely prevents its interaction with the β2 and β4-subunit Ig-domain. These isoform-specific glycans may have evolved to facilitate specific functional interactions, for example, by redirecting β-subunit Ig-domains outward to permit cis or trans supraclustering within specialized cellular compartments such as the cardiomyocyte perinexal space. Further experimental work is necessary to validate these predictions.
Collapse
Affiliation(s)
| | - Manas Kohli
- Department of Biochemistry, Hopkins Building, University of Cambridge, Cambridge, UK
| | - Samantha C. Salvage
- Department of Biochemistry, Hopkins Building, University of Cambridge, Cambridge, UK
| | - Hengrui Liu
- Department of Biochemistry, Hopkins Building, University of Cambridge, Cambridge, UK
| | - Samuel J. Arundel
- Department of Biochemistry, Hopkins Building, University of Cambridge, Cambridge, UK
| | - Samir W. Hamaia
- Department of Biochemistry, Hopkins Building, University of Cambridge, Cambridge, UK
| | - Ming Lei
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Christopher L.-H. Huang
- Department of Biochemistry, Hopkins Building, University of Cambridge, Cambridge, UK
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Antony P. Jackson
- Department of Biochemistry, Hopkins Building, University of Cambridge, Cambridge, UK
| |
Collapse
|
22
|
Liu Y, Bezanilla F. A sodium channel mutant removes fast inactivation with the inactivation particle bound. J Gen Physiol 2025; 157:e202413667. [PMID: 39601860 PMCID: PMC11602646 DOI: 10.1085/jgp.202413667] [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: 08/28/2024] [Revised: 11/01/2024] [Accepted: 11/13/2024] [Indexed: 11/29/2024] Open
Abstract
Fast inactivation is a key feature of voltage-gated sodium channels and is pivotal for countless physiological functions. Despite the prevalence of the canonical ball-and-chain model, more recent structural results suggest that fast inactivation requires multiple conformational changes beyond the binding of the inactivation particle, the IFM motif. Combining ionic current, gating current, and fluorescent measurements, here we showed that a double mutant at the bottom of the pore domain (CW) removes fast inactivation by interrupting the communication of the IFM motif and the pore. Instead of triggering fast inactivation, the IFM motif binding in CW allows the channel to enter an alternative open state. This alternative open state severely influenced the voltage sensor movements and was not accessible to wild type or other fast inactivation-deficient channels. Our results highlight the multistep nature of the fast inactivation process in mammalian voltage-gated sodium channels and demonstrate that CW modifies the channel behaviors more profoundly than simple removal of fast inactivation.
Collapse
Affiliation(s)
- Yichen Liu
- Department of Neurobiology, University of Chicago, Chicago, IL, USA
| | - Francisco Bezanilla
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL, USA
- Centro Interdisciplinario de Neurociencias de Valparaiso, Valparaiso, Chile
| |
Collapse
|
23
|
Şulea TA, Draga S, Mernea M, Corlan AD, Radu BM, Petrescu AJ, Amuzescu B. Differential Inhibition by Cenobamate of Canonical Human Nav1.5 Ion Channels and Several Point Mutants. Int J Mol Sci 2025; 26:358. [PMID: 39796214 PMCID: PMC11720074 DOI: 10.3390/ijms26010358] [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: 11/25/2024] [Revised: 12/21/2024] [Accepted: 12/31/2024] [Indexed: 01/30/2025] Open
Abstract
Cenobamate is a new and highly effective antiseizure compound used for the treatment of adults with focal onset seizures and particularly for epilepsy resistant to other antiepileptic drugs. It acts on multiple targets, as it is a positive allosteric activator of γ-aminobutyric acid type A (GABAA) receptors and an inhibitor of neuronal sodium channels, particularly of the late or persistent Na+ current. We recently evidenced the inhibitory effects of cenobamate on the peak and late current component of the human cardiac isoform hNav1.5. The determined apparent IC50 values of 87.6 µM (peak) and 46.5 µM (late current) are within a clinically relevant range of concentrations (the maximal plasma therapeutic effective concentration for a daily dose of 400 mg in humans is 170 µM). In this study, we built a 3D model of the canonical hNav1.5 channel (UniProt Q14524-1) in open conformation using AlphaFold2, embedded it in a DPPC lipid bilayer, corrected the residue protonation state (pH 7.2) with H++, and added 2 Na+ ions in the selectivity filter. By molecular docking, we found the cenobamate binding site in the central cavity. We identified 10-point mutant variants in the binding site region and explored them via docking and MD. Mutants N1462K/Y (rs1064795922, rs199473614) and M1765R (rs752476527) (by docking) and N932S (rs2061582195) (by MD) featured higher predicted affinity than wild-type.
Collapse
Affiliation(s)
- Teodor Asvadur Şulea
- Department of Bioinformatics and Structural Biochemistry, Institute of Biochemistry of the Romanian Academy, Splaiul Independentei 296, 060031 Bucharest, Romania; (T.A.Ş.); (A.-J.P.)
| | - Sorin Draga
- Biotehnos SA, Gorunului Str. 3-5, 075100 Otopeni, Romania;
- Non-Governmental Research Organization Biologic, 14 Schitului Str., 032044 Bucharest, Romania
| | - Maria Mernea
- Department of Anatomy, Animal Physiology and Biophysics, Faculty of Biology, University of Bucharest, Splaiul Independentei 91-95, 050095 Bucharest, Romania; (B.M.R.); (B.A.)
| | - Alexandru Dan Corlan
- Cardiology Research Unit, University and Emergency Hospital of Bucharest, Splaiul Independenței 169, 050098 Bucharest, Romania;
| | - Beatrice Mihaela Radu
- Department of Anatomy, Animal Physiology and Biophysics, Faculty of Biology, University of Bucharest, Splaiul Independentei 91-95, 050095 Bucharest, Romania; (B.M.R.); (B.A.)
| | - Andrei-Jose Petrescu
- Department of Bioinformatics and Structural Biochemistry, Institute of Biochemistry of the Romanian Academy, Splaiul Independentei 296, 060031 Bucharest, Romania; (T.A.Ş.); (A.-J.P.)
| | - Bogdan Amuzescu
- Department of Anatomy, Animal Physiology and Biophysics, Faculty of Biology, University of Bucharest, Splaiul Independentei 91-95, 050095 Bucharest, Romania; (B.M.R.); (B.A.)
| |
Collapse
|
24
|
Anghelescu GDC, Mernea M, Mihăilescu DF. Mapping O- and N-Glycosylation in Transmembrane and Interface Regions of Proteins: Insights from a Database Search Study. Int J Mol Sci 2025; 26:327. [PMID: 39796186 PMCID: PMC11720221 DOI: 10.3390/ijms26010327] [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: 12/08/2024] [Revised: 12/30/2024] [Accepted: 12/31/2024] [Indexed: 01/13/2025] Open
Abstract
Glycosylation is a critical post-translational modification that influences protein folding, stability and function. While extensively studied in extracellular and intracellular regions, glycosylation within transmembrane (TM) regions and at membrane interfaces remains poorly understood. This study aimed to map O- and N-glycosylation sites in these regions using a comprehensive database search and structural validation where possible. Extensive database searches revealed glycosylation sites in a range of membrane proteins. Only the sites falling in the TM regions and at the membrane interface (according to Uniprot annotations) were retained. The location of these sites was confirmed based on available 3D structures. We identified 32 O-glycosylation sites and 7 N-glycosylation sites in the TM domains of 29 proteins. O-GlcNAc sites validated as located within TM regions presented side chains either oriented toward the lipid bilayer or buried within the protein. N-glycosylation sites predicted in protein TM regions were largely confined to interface or extracellular domains. The results obtained here highlight the occurrence of glycosylation in TM regions of proteins and at membrane interfaces. This dataset provides a valuable foundation for the further exploration of structural and functional roles of glycosylation in membrane-associated regions.
Collapse
Affiliation(s)
- Giorgiana Diana Carmen Anghelescu
- Doctoral School in Biology, Faculty of Biology, University of Bucharest, 91–95 Splaiul Independenței Str., 050095 Bucharest, Romania;
| | - Maria Mernea
- Department of Anatomy, Animal Physiology and Biophysics, Faculty of Biology, University of Bucharest, 91–95 Splaiul Independenței Str., 050095 Bucharest, Romania;
| | - Dan Florin Mihăilescu
- Department of Anatomy, Animal Physiology and Biophysics, Faculty of Biology, University of Bucharest, 91–95 Splaiul Independenței Str., 050095 Bucharest, Romania;
| |
Collapse
|
25
|
Wang J, Wu Z, Cao L, Long F. Differential Regulation of Hyaluronan Synthesis by Three Isoforms of Hyaluronan Synthases in Mammalian Cells. Biomolecules 2024; 14:1567. [PMID: 39766274 PMCID: PMC11673962 DOI: 10.3390/biom14121567] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 11/21/2024] [Accepted: 11/30/2024] [Indexed: 01/11/2025] Open
Abstract
Hyaluronan (HA) is one of the crucial components of the extracellular matrix in vertebrates and is synthesized by three hyaluronan synthases (HASs), namely HAS1, HAS2, and HAS3. The low expression level of HASs in normal keratinocytes and other various types of cells presents a recognized challenge, impeding biological and pathological research on their localization. In this study, the human proteins HAS1, HAS2, and HAS3 with fused maltose-binding protein (MBP) tags were successfully expressed at high levels and purified for the first time in HEK293F cells. The enzymatic properties of the three HAS proteins were further characterized and compared. A pulse-field gel electrophoresis analysis of the size distribution of the hyaluronan generated in vitro by the membrane proteins demonstrated that the three HAS isoforms generate HA polymer chains at different molecular masses. Kinetic studies demonstrated that the three HAS proteins differed in their catalytic efficiency and apparent Km values for the two substrates, UDP-GlcA and UDP-GlcNAc. Furthermore, the cellular hyaluronan secretion by the three isoenzymes was evaluated and quantified in the HEK 293T cells transfected with GFP-tagged HAS1-GFP, HAS2-GFP, and HAS3-GFP using an ELISA assay. These findings enhance our understanding of the membrane protein HASs in mammalian cells.
Collapse
Affiliation(s)
- Jie Wang
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China; (J.W.); (Z.W.); (L.C.)
- Ministry of Education Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
| | - Zhikun Wu
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China; (J.W.); (Z.W.); (L.C.)
- Ministry of Education Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
| | - Longtao Cao
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China; (J.W.); (Z.W.); (L.C.)
- Ministry of Education Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
| | - Feng Long
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China; (J.W.); (Z.W.); (L.C.)
- Ministry of Education Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
| |
Collapse
|
26
|
Nguyen PT, Harris BJ, Mateos DL, González AH, Murray AM, Yarov-Yarovoy V. Structural modeling of ion channels using AlphaFold2, RoseTTAFold2, and ESMFold. Channels (Austin) 2024; 18:2325032. [PMID: 38445990 PMCID: PMC10936637 DOI: 10.1080/19336950.2024.2325032] [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: 09/05/2023] [Accepted: 01/14/2024] [Indexed: 03/07/2024] Open
Abstract
Ion channels play key roles in human physiology and are important targets in drug discovery. The atomic-scale structures of ion channels provide invaluable insights into a fundamental understanding of the molecular mechanisms of channel gating and modulation. Recent breakthroughs in deep learning-based computational methods, such as AlphaFold, RoseTTAFold, and ESMFold have transformed research in protein structure prediction and design. We review the application of AlphaFold, RoseTTAFold, and ESMFold to structural modeling of ion channels using representative voltage-gated ion channels, including human voltage-gated sodium (NaV) channel - NaV1.8, human voltage-gated calcium (CaV) channel - CaV1.1, and human voltage-gated potassium (KV) channel - KV1.3. We compared AlphaFold, RoseTTAFold, and ESMFold structural models of NaV1.8, CaV1.1, and KV1.3 with corresponding cryo-EM structures to assess details of their similarities and differences. Our findings shed light on the strengths and limitations of the current state-of-the-art deep learning-based computational methods for modeling ion channel structures, offering valuable insights to guide their future applications for ion channel research.
Collapse
Affiliation(s)
- Phuong Tran Nguyen
- Department of Physiology and Membrane Biology, University of California School of Medicine, Davis, CA, USA
| | - Brandon John Harris
- Department of Physiology and Membrane Biology, University of California School of Medicine, Davis, CA, USA
- Biophysics Graduate Group, University of California School of Medicine, Davis, CA, USA
| | - Diego Lopez Mateos
- Department of Physiology and Membrane Biology, University of California School of Medicine, Davis, CA, USA
- Biophysics Graduate Group, University of California School of Medicine, Davis, CA, USA
| | - Adriana Hernández González
- Department of Physiology and Membrane Biology, University of California School of Medicine, Davis, CA, USA
- Biophysics Graduate Group, University of California School of Medicine, Davis, CA, USA
| | | | - Vladimir Yarov-Yarovoy
- Department of Physiology and Membrane Biology, University of California School of Medicine, Davis, CA, USA
- Department of Anesthesiology and Pain Medicine, University of California School of Medicine, Davis, CA, USA
| |
Collapse
|
27
|
Egunjobi F, Andreazza F, Zhorov BS, Dong K. A unique mechanism of transfluthrin action revealed by mapping its binding sites in the mosquito sodium channel. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2024; 175:104214. [PMID: 39566664 PMCID: PMC11624841 DOI: 10.1016/j.ibmb.2024.104214] [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: 09/03/2024] [Revised: 11/16/2024] [Accepted: 11/17/2024] [Indexed: 11/22/2024]
Abstract
Pyrethroid insecticides exert their toxic action by prolonging the opening of insect voltage-gated sodium channels, resulting in the characteristic tail current during membrane repolarization in voltage clamp experiments. Permethrin (PMT) and deltamethrin (DMT), representative type I and type II pyrethroids, respectively, are predicted to bind to two lipid-exposed pyrethroid receptor sites, PyR1 and PyR2, at the lipid-exposed interfaces of repeats II/III and I/II, respectively. Transfluthrin (TF), a volatile type I pyrethroid and mosquito repellent, has received increased attention in the global combat of vector-borne human diseases. However, the electrophysiological and molecular bases of TF action on insect sodium channels remain unexplored. In this study we discovered that, unlike DMT and PMT, TF barely induces the characteristic tail current of the Aedes aegypti mosquito sodium channel (AaNav1-1) expressed in Xenopus oocytes. Instead, TF induces a unique persistent current. We docked TF into the AlphaFold2 model of AaNav1-1 and found that the tetrafluorophenyl ring of TF binds to alpha helices S5, P1, and S6, but not to the linker helices S4-S5 within either PyR1 or PyR2. In agreement with the model, functional examination of 15 AaNav1-1 mutants demonstrated that substitutions of DMT/PMT-sensing residues in helices S5, P1, and S6, but not in the linker-helices S4-S5, altered channel sensitivity to TF. These results revealed the unique action of TF on channel gating and suggest a distinct subtype of type I pyrethroids with a previously uncharacterized pattern of interactions with residues at the dual pyrethroid receptor sites.
Collapse
Affiliation(s)
| | | | - Boris S Zhorov
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, L8S 4K1, Canada; Sechenov Institute of Evolutionary Physiology & Biochemistry, Russian Academy of Sciences, Saint Petersburg, 194223, Russia; Almazov National Medical Research Centre, Saint Petersburg, 197341, Russia
| | - Ke Dong
- Department of Biology, Duke University, Durham, NC, 27708, USA.
| |
Collapse
|
28
|
Li Z, Wu Q, Yan N. A structural atlas of druggable sites on Na v channels. Channels (Austin) 2024; 18:2287832. [PMID: 38033122 PMCID: PMC10732651 DOI: 10.1080/19336950.2023.2287832] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 11/20/2023] [Indexed: 12/02/2023] Open
Abstract
Voltage-gated sodium (Nav) channels govern membrane excitability by initiating and propagating action potentials. Consistent with their physiological significance, dysfunction, or mutations in these channels are associated with various channelopathies. Nav channels are thereby major targets for various clinical and investigational drugs. In addition, a large number of natural toxins, both small molecules and peptides, can bind to Nav channels and modulate their functions. Technological breakthrough in cryo-electron microscopy (cryo-EM) has enabled the determination of high-resolution structures of eukaryotic and eventually human Nav channels, alone or in complex with auxiliary subunits, toxins, and drugs. These studies have not only advanced our comprehension of channel architecture and working mechanisms but also afforded unprecedented clarity to the molecular basis for the binding and mechanism of action (MOA) of prototypical drugs and toxins. In this review, we will provide an overview of the recent advances in structural pharmacology of Nav channels, encompassing the structural map for ligand binding on Nav channels. These findings have established a vital groundwork for future drug development.
Collapse
Affiliation(s)
- Zhangqiang Li
- Beijing Frontier Research Center for Biological Structures, State Key Laboratory of Membrane Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Qiurong Wu
- Beijing Frontier Research Center for Biological Structures, State Key Laboratory of Membrane Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Nieng Yan
- Beijing Frontier Research Center for Biological Structures, State Key Laboratory of Membrane Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
- Shenzhen Medical Academy of Research and Translation, Shenzhen, Guangdong Province, China
| |
Collapse
|
29
|
Huang J, Pan X, Yan N. Structural biology and molecular pharmacology of voltage-gated ion channels. Nat Rev Mol Cell Biol 2024; 25:904-925. [PMID: 39103479 DOI: 10.1038/s41580-024-00763-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/26/2024] [Indexed: 08/07/2024]
Abstract
Voltage-gated ion channels (VGICs), including those for Na+, Ca2+ and K+, selectively permeate ions across the cell membrane in response to changes in membrane potential, thus participating in physiological processes involving electrical signalling, such as neurotransmission, muscle contraction and hormone secretion. Aberrant function or dysregulation of VGICs is associated with a diversity of neurological, psychiatric, cardiovascular and muscular disorders, and approximately 10% of FDA-approved drugs directly target VGICs. Understanding the structure-function relationship of VGICs is crucial for our comprehension of their working mechanisms and role in diseases. In this Review, we discuss how advances in single-particle cryo-electron microscopy have afforded unprecedented structural insights into VGICs, especially on their interactions with clinical and investigational drugs. We present a comprehensive overview of the recent advances in the structural biology of VGICs, with a focus on how prototypical drugs and toxins modulate VGIC activities. We explore how these structures elucidate the molecular basis for drug actions, reveal novel pharmacological sites, and provide critical clues to future drug discovery.
Collapse
Affiliation(s)
- Jian Huang
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Xiaojing Pan
- Institute of Bio-Architecture and Bio-Interactions (IBABI), Shenzhen Medical Academy of Research and Translation (SMART), Shenzhen, Guangdong, China.
| | - Nieng Yan
- Institute of Bio-Architecture and Bio-Interactions (IBABI), Shenzhen Medical Academy of Research and Translation (SMART), Shenzhen, Guangdong, China.
- Beijing Frontier Research Center for Biological Structure, Tsinghua-Peking Joint Center for Life Sciences, State Key Laboratory of Membrane Biology, Tsinghua University, Beijing, China.
| |
Collapse
|
30
|
Collaço RDC, Van Petegem F, Bosmans F. ω-Grammotoxin-SIA inhibits voltage-gated Na+ channel currents. J Gen Physiol 2024; 156:e202413563. [PMID: 39042091 PMCID: PMC11270453 DOI: 10.1085/jgp.202413563] [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: 02/09/2024] [Revised: 06/14/2024] [Accepted: 07/10/2024] [Indexed: 07/24/2024] Open
Abstract
ω-Grammotoxin-SIA (GrTX-SIA) was originally isolated from the venom of the Chilean rose tarantula and demonstrated to function as a gating modifier of voltage-gated Ca2+ (CaV) channels. Later experiments revealed that GrTX-SIA could also inhibit voltage-gated K+ (KV) channel currents via a similar mechanism of action that involved binding to a conserved S3-S4 region in the voltage-sensing domains (VSDs). Since voltage-gated Na+ (NaV) channels contain homologous structural motifs, we hypothesized that GrTX-SIA could inhibit members of this ion channel family as well. Here, we show that GrTX-SIA can indeed impede the gating process of multiple NaV channel subtypes with NaV1.6 being the most susceptible target. Moreover, molecular docking of GrTX-SIA onto NaV1.6, supported by a p.E1607K mutation, revealed the voltage sensor in domain IV (VSDIV) as being a primary site of action. The biphasic manner in which current inhibition appeared to occur suggested a second, possibly lower-sensitivity binding locus, which was identified as VSDII by using KV2.1/NaV1.6 chimeric voltage-sensor constructs. Subsequently, the NaV1.6p.E782K/p.E838K (VSDII), NaV1.6p.E1607K (VSDIV), and particularly the combined VSDII/VSDIV mutant lost virtually all susceptibility to GrTX-SIA. Together with existing literature, our data suggest that GrTX-SIA recognizes modules in NaV channel VSDs that are conserved among ion channel families, thereby allowing it to act as a comprehensive ion channel gating modifier peptide.
Collapse
Affiliation(s)
- Rita de Cássia Collaço
- Department of Basic and Applied Medical Sciences, Molecular Physiology and Neurophysics Group, Ghent University, Ghent, Belgium
| | - Filip Van Petegem
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, Canada
- Life Sciences Institute, University of British Columbia, Vancouver, Canada
| | - Frank Bosmans
- Department of Basic and Applied Medical Sciences, Molecular Physiology and Neurophysics Group, Ghent University, Ghent, Belgium
- Faculty of Medicine and Pharmacy, Center for Neurosciences (C4N), Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
31
|
Phulera S, Dickson CJ, Schwalen CJ, Khoshouei M, Cassell SJ, Sun Y, Condos T, Whicher J, Weihofen WA. Scorpion α-toxin LqhαIT specifically interacts with a glycan at the pore domain of voltage-gated sodium channels. Structure 2024; 32:1611-1620.e4. [PMID: 39181123 DOI: 10.1016/j.str.2024.07.021] [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: 02/12/2024] [Revised: 06/13/2024] [Accepted: 07/30/2024] [Indexed: 08/27/2024]
Abstract
Voltage-gated sodium (Nav) channels sense membrane potential and drive cellular electrical activity. The deathstalker scorpion α-toxin LqhαIT exerts a strong action potential prolonging effect on Nav channels. To elucidate the mechanism of action of LqhαIT, we determined a 3.9 Å cryoelectron microscopy (cryo-EM) structure of LqhαIT in complex with the Nav channel from Periplaneta americana (NavPas). We found that LqhαIT binds to voltage sensor domain 4 and traps it in an "S4 down" conformation. The functionally essential C-terminal epitope of LqhαIT forms an extensive interface with the glycan scaffold linked to Asn330 of NavPas that augments a small protein-protein interface between NavPas and LqhαIT. A combination of molecular dynamics simulations, structural comparisons, and prior mutagenesis experiments demonstrates the functional importance of this toxin-glycan interaction. These findings establish a structural basis for the specificity achieved by scorpion α-toxins and reveal the conserved glycan as an essential component of the toxin-binding epitope.
Collapse
Affiliation(s)
- Swastik Phulera
- Discovery Sciences, Novartis Biomedical Research, 250 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Callum J Dickson
- Global Discovery Chemistry, Novartis Biomedical Research, 181 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Christopher J Schwalen
- Global Discovery Chemistry, Novartis Biomedical Research, 181 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Maryam Khoshouei
- Discovery Sciences, Novartis Biomedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Samantha J Cassell
- Discovery Sciences, Novartis Biomedical Research, 250 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Yishan Sun
- Neuroscience, Novartis Biomedical Research, 22 Windsor St, Cambridge, MA 02139, USA
| | - Tara Condos
- Discovery Sciences, Novartis Biomedical Research, 250 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Jonathan Whicher
- Discovery Sciences, Novartis Biomedical Research, 250 Massachusetts Avenue, Cambridge, MA 02139, USA.
| | - Wilhelm A Weihofen
- Discovery Sciences, Novartis Biomedical Research, 250 Massachusetts Avenue, Cambridge, MA 02139, USA.
| |
Collapse
|
32
|
Hollingworth D, Thomas F, Page DA, Fouda MA, De Castro RLR, Sula A, Mykhaylyk VB, Kelly G, Ulmschneider MB, Ruben PC, Wallace BA. Structural basis for the rescue of hyperexcitable cells by the amyotrophic lateral sclerosis drug Riluzole. Nat Commun 2024; 15:8426. [PMID: 39341837 PMCID: PMC11438954 DOI: 10.1038/s41467-024-52539-4] [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: 03/22/2024] [Accepted: 09/09/2024] [Indexed: 10/01/2024] Open
Abstract
Neuronal hyperexcitability is a key element of many neurodegenerative disorders including the motor neuron disease Amyotrophic Lateral Sclerosis (ALS), where it occurs associated with elevated late sodium current (INaL). INaL results from incomplete inactivation of voltage-gated sodium channels (VGSCs) after their opening and shapes physiological membrane excitability. However, dysfunctional increases can cause hyperexcitability-associated diseases. Here we reveal the atypical binding mechanism which explains how the neuroprotective ALS-treatment drug riluzole stabilises VGSCs in their inactivated state to cause the suppression of INaL that leads to reversed cellular overexcitability. Riluzole accumulates in the membrane and enters VGSCs through openings to their membrane-accessible fenestrations. Riluzole binds within these fenestrations to stabilise the inactivated channel state, allowing for the selective allosteric inhibition of INaL without the physical block of Na+ conduction associated with traditional channel pore binding VGSC drugs. We further demonstrate that riluzole can reproduce these effects on a disease variant of the non-neuronal VGSC isoform Nav1.4, where pathologically increased INaL is caused directly by mutation. Overall, we identify a model for VGSC inhibition that produces effects consistent with the inhibitory action of riluzole observed in models of ALS. Our findings will aid future drug design and supports research directed towards riluzole repurposing.
Collapse
Affiliation(s)
- David Hollingworth
- School of Natural Sciences, Birkbeck University of London, London, United Kingdom
| | - Frances Thomas
- School of Natural Sciences, Birkbeck University of London, London, United Kingdom
| | - Dana A Page
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
| | - Mohamed A Fouda
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
| | - Raquel Lopez-Rios De Castro
- Department of Chemistry, King's College London, London, United Kingdom
- Biological Physics and Soft Matter Group, Department of Physics, King's College London, London, United Kingdom
| | - Altin Sula
- Syngenta Crop Protection, Jealott's Hill International Research Centre, Bracknell, Berkshire, United Kingdom
| | - Vitaliy B Mykhaylyk
- Diamond Light Source, Harwell Science and Innovation Campus, Chilton, Didcot, UK
| | - Geoff Kelly
- The Medical Research Council Biomedical NMR Centre, The Francis Crick Institute, London, UK
| | | | - Peter C Ruben
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada.
| | - B A Wallace
- School of Natural Sciences, Birkbeck University of London, London, United Kingdom.
| |
Collapse
|
33
|
Akbari Ahangar A, Elhanafy E, Blanton H, Li J. Mapping structural distribution and gating-property impacts of disease-associated mutations in voltage-gated sodium channels. iScience 2024; 27:110678. [PMID: 39286500 PMCID: PMC11404175 DOI: 10.1016/j.isci.2024.110678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 06/18/2024] [Accepted: 08/02/2024] [Indexed: 09/19/2024] Open
Abstract
Thousands of voltage-gated sodium (Nav) channel variants contribute to a variety of disorders, including epilepsy, cardiac arrhythmia, and pain disorders. Yet, the effects of more variants remain unclear. The conventional gain-of-function (GoF) or loss-of-function (LoF) classifications are frequently employed to interpret mutations' effects and guide therapy for sodium channelopathies. Our study challenges this binary classification by analyzing 525 mutations associated with 34 diseases across 366 electrophysiology studies, revealing that diseases with similar GoF/LoF effects can stem from unique molecular mechanisms. Utilizing UniProt data, we mapped over 2,400 disease-associated missense mutations across Nav channels. This analysis pinpoints key mutation hotspots and maps patterns of gating-property impacts for the mutations, respectively, located around the selectivity filter, activation gate, fast inactivation region, and voltage-sensing domains. This study shows great potential to enhance prediction accuracy for mutational effects based on the structural context, paving the way for targeted drug design in precision medicine.
Collapse
Affiliation(s)
- Amin Akbari Ahangar
- Department of Biomolecular Sciences, School of Pharmacy, University of Mississippi, Oxford, MS 38677, USA
| | - Eslam Elhanafy
- Department of Biomolecular Sciences, School of Pharmacy, University of Mississippi, Oxford, MS 38677, USA
| | - Hayden Blanton
- Department of Biomolecular Sciences, School of Pharmacy, University of Mississippi, Oxford, MS 38677, USA
| | - Jing Li
- Department of Biomolecular Sciences, School of Pharmacy, University of Mississippi, Oxford, MS 38677, USA
| |
Collapse
|
34
|
Elhanafy E, Ahangar AA, Roth R, Gamal El-Din TM, Bankston JR, Li J. ELUCIDATING THE DIFFERENTIAL IMPACTS OF EQUIVALENT GATING-CHARGE MUTATIONS IN VOLTAGE-GATED SODIUM CHANNELS. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.09.612021. [PMID: 39314455 PMCID: PMC11419121 DOI: 10.1101/2024.09.09.612021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Voltage-gated sodium (Nav) channels are pivotal for cellular signaling and mutations in Nav channels can lead to excitability disorders in cardiac, muscular, and neural tissues. A major cluster of pathological mutations localizes in the voltage-sensing domains (VSDs), resulting in either gain-of-function (GoF), loss-of-function (LoF) effects, or both. However, the mechanism behind this functional divergence of mutations at equivalent positions remains elusive. Through hotspot analysis, we identified three gating charges (R1, R2, and R3) as major mutational hotspots in VSDs. The same amino-acid substitutions at equivalent gating-charge positions in VSDI and VSDII of the cardiac sodium channel Nav1.5 show differential gating-property impacts in electrophysiology measurements. We conducted 120 μs molecular dynamics (MD) simulations on wild-type and six mutants to elucidate the structural basis of their differential impacts. Our μs-scale MD simulations with applied external electric fields captured VSD state transitions and revealed the differential structural dynamics between equivalent R-to-Q mutants. Notably, we observed transient leaky conformations in some mutants during structural transitions, offering a detailed structural explanation for gating-pore currents. Our salt-bridge network analysis uncovered VSD-specific and state-dependent interactions among gating charges, countercharges, and lipids. This detailed analysis elucidated how mutations disrupt critical electrostatic interactions, thereby altering VSD permeability and modulating gating properties. By demonstrating the crucial importance of considering the specific structural context of each mutation, our study represents a significant leap forward in understanding structure-function relationships in Nav channels. Our work establishes a robust framework for future investigations into the molecular basis of ion channel-related disorders.
Collapse
Affiliation(s)
- Eslam Elhanafy
- Department of Biomolecular Sciences, School of Pharmacy, University of Mississippi, Oxford, MS
| | - Amin Akbari Ahangar
- Department of Biomolecular Sciences, School of Pharmacy, University of Mississippi, Oxford, MS
| | - Rebecca Roth
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical Campus, Aurora, CO
| | | | - John R Bankston
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Jing Li
- Department of Biomolecular Sciences, School of Pharmacy, University of Mississippi, Oxford, MS
| |
Collapse
|
35
|
Tikhonova TB, Sharkov AA, Zhorov BS, Vassilevski AA. Diverse biophysical mechanisms in voltage-gated sodium channel Na v1.4 variants associated with myotonia. FASEB J 2024; 38:e23883. [PMID: 39150825 DOI: 10.1096/fj.202400867r] [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: 04/16/2024] [Revised: 07/14/2024] [Accepted: 07/31/2024] [Indexed: 08/18/2024]
Abstract
Mutations in SCN4A gene encoding Nav1.4 channel α-subunit, are known to cause neuromuscular disorders such as myotonia or paralysis. Here, we study the effect of two amino acid replacements, K1302Q and G1306E, in the DIII-IV loop of the channel, corresponding to mutations found in patients with myotonia. We combine clinical, electrophysiological, and molecular modeling data to provide a holistic picture of the molecular mechanisms operating in mutant channels and eventually leading to pathology. We analyze the existing clinical data for patients with the K1302Q substitution, which was reported for adults with or without myotonia phenotypes, and report two new unrelated patients with the G1306E substitution, who presented with severe neonatal episodic laryngospasm and childhood-onset myotonia. We provide a functional analysis of the mutant channels by expressing Nav1.4 α-subunit in Xenopus oocytes in combination with β1 subunit and recording sodium currents using two-electrode voltage clamp. The K1302Q variant exhibits abnormal voltage dependence of steady-state fast inactivation, being the likely cause of pathology. K1302Q does not lead to decelerated fast inactivation, unlike several other myotonic mutations such as G1306E. For both mutants, we observe increased window currents corresponding to a larger population of channels available for activation. To elaborate the structural rationale for our experimental data, we explore the contacts involving K/Q1302 and E1306 in the AlphaFold2 model of wild-type Nav1.4 and Monte Carlo-minimized models of mutant channels. Our data provide the missing evidence to support the classification of K1302Q variant as likely pathogenic and may be used by clinicians.
Collapse
Affiliation(s)
- Tatiana B Tikhonova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Artem A Sharkov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Veltischev Research and Clinical Institute for Pediatrics and Pediatric Surgery of the Pirogov Russian National Research Medical University, Moscow, Russia
- Genomed Ltd., Moscow, Russia
| | - Boris S Zhorov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Sechenov Institute of Evolutionary Physiology & Biochemistry, Russian Academy of Sciences, St. Petersburg, Russia
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Canada
| | - Alexander A Vassilevski
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Moscow Institute of Physics and Technology, State University, Dolgoprudny, Russia
| |
Collapse
|
36
|
Neumann B, McCarthy S, Gonen S. Structural basis of inhibition of human Na V1.8 by the tarantula venom peptide Protoxin-I. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.27.609828. [PMID: 39253517 PMCID: PMC11383277 DOI: 10.1101/2024.08.27.609828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
Voltage-gated sodium channels (NaVs) selectively permit diffusion of sodium ions across the cell membrane and, in excitable cells, are responsible for propagating action potentials. One of the nine human NaV isoforms, NaV1.8, is a promising target for analgesics, and selective inhibitors are of interest as therapeutics. One such inhibitor, the gating-modifier peptide Protoxin-I derived from tarantula venom, blocks channel opening by shifting the activation voltage threshold to more depolarised potentials, but the structural basis for this inhibition has not previously been determined. Using monolayer graphene grids, we report the cryogenic electron microscopy structures of full-length human apo-NaV1.8 and the Protoxin-I-bound complex at 3.1 Å and 2.8 Å resolution, respectively. The apo structure shows an unexpected movement of the Domain I S4-S5 helix, and VSDI was unresolvable. We find that Protoxin-I binds to and displaces the VSDII S3-S4 linker, hindering translocation of the S4II helix during activation.
Collapse
Affiliation(s)
- Bryan Neumann
- Department of Molecular Biology and Biochemistry, University of California Irvine, CA 92617, USA
| | - Stephen McCarthy
- Department of Molecular Biology and Biochemistry, University of California Irvine, CA 92617, USA
| | - Shane Gonen
- Department of Molecular Biology and Biochemistry, University of California Irvine, CA 92617, USA
| |
Collapse
|
37
|
Palmisano VF, Anguita-Ortiz N, Faraji S, Nogueira JJ. Voltage-Gated Ion Channels: Structure, Pharmacology and Photopharmacology. Chemphyschem 2024; 25:e202400162. [PMID: 38649320 DOI: 10.1002/cphc.202400162] [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: 02/23/2024] [Revised: 04/21/2024] [Accepted: 04/22/2024] [Indexed: 04/25/2024]
Abstract
Voltage-gated ion channels are transmembrane proteins responsible for the generation and propagation of action potentials in excitable cells. Over the last decade, advancements have enabled the elucidation of crystal structures of ion channels. This progress in structural understanding, particularly in identifying the binding sites of local anesthetics, opens avenues for the design of novel compounds capable of modulating ion conduction. However, many traditional drugs lack selectivity and come with adverse side effects. The emergence of photopharmacology has provided an orthogonal way of controlling the activity of compounds, enabling the regulation of ion conduction with light. In this review, we explore the central pore region of voltage-gated sodium and potassium channels, providing insights from both structural and pharmacological perspectives. We discuss the different binding modes of synthetic compounds that can physically occlude the pore and, therefore, block ion conduction. Moreover, we examine recent advances in the photopharmacology of voltage-gated ion channels, introducing molecular approaches aimed at controlling their activity by using photosensitive drugs.
Collapse
Affiliation(s)
- Vito F Palmisano
- Department of Chemistry, Universidad Autónoma de Madrid, 28049, Madrid, Spain
- Theoretical Chemistry Group, Zernike Institute for Advanced Materials, University of Groningen, 9747 AG, Groningen, The Netherlands
| | - Nuria Anguita-Ortiz
- Department of Chemistry, Universidad Autónoma de Madrid, 28049, Madrid, Spain
| | - Shirin Faraji
- Theoretical Chemistry Group, Zernike Institute for Advanced Materials, University of Groningen, 9747 AG, Groningen, The Netherlands
| | - Juan J Nogueira
- Department of Chemistry, Universidad Autónoma de Madrid, 28049, Madrid, Spain
- IADCHEM, Institute for Advanced Research in Chemistry, Universidad Autónoma de Madrid, 28049, Madrid, Spain
| |
Collapse
|
38
|
Catterall WA, Gamal El-Din TM, Wisedchaisri G. The chemistry of electrical signaling in sodium channels from bacteria and beyond. Cell Chem Biol 2024; 31:1405-1421. [PMID: 39151407 PMCID: PMC12084679 DOI: 10.1016/j.chembiol.2024.07.010] [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: 01/12/2024] [Revised: 06/27/2024] [Accepted: 07/22/2024] [Indexed: 08/19/2024]
Abstract
Electrical signaling is essential for all fast processes in biology, but its molecular mechanisms have been uncertain. This review article focuses on studies of bacterial sodium channels in order to home in on the essential molecular and chemical mechanisms underlying transmembrane ion conductance and voltage-dependent gating without the overlay of complex protein interactions and regulatory mechanisms in mammalian sodium channels. This minimalist approach has yielded a nearly complete picture of sodium channel function at the atomic level that are mostly conserved in mammalian sodium channels, including sodium selectivity and conductance, voltage sensing and activation, electromechanical coupling to pore opening and closing, slow inactivation, and pathogenic dysfunction in a debilitating channelopathy. Future studies of nature's simplest sodium channels may continue to yield key insights into the fundamental molecular and chemical principles of their function and further elucidate the chemical basis of electrical signaling.
Collapse
Affiliation(s)
- William A Catterall
- Department of Pharmacology, University of Washington, Seattle WA 98195-7280, USA.
| | - Tamer M Gamal El-Din
- Department of Pharmacology, University of Washington, Seattle WA 98195-7280, USA.
| | - Goragot Wisedchaisri
- Department of Pharmacology, University of Washington, Seattle WA 98195-7280, USA.
| |
Collapse
|
39
|
Jin X, Huang J, Wang H, Wang K, Yan N. A versatile residue numbering scheme for Na v and Ca v channels. Cell Chem Biol 2024; 31:1394-1404. [PMID: 39151406 DOI: 10.1016/j.chembiol.2024.07.008] [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: 02/18/2024] [Revised: 07/01/2024] [Accepted: 07/18/2024] [Indexed: 08/19/2024]
Abstract
Voltage-gated sodium (Nav) and calcium (Cav) channels are responsible for the initiation of electrical signals. They have long been targeted for the treatment of various diseases. The mounting number of cryoelectron microscopy (cryo-EM) structures for diverse subtypes of Nav and Cav channels from multiple organisms necessitates a generic residue numbering system to establish the structure-function relationship and to aid rational drug design or optimization. Here we suggest a structure-based residue numbering scheme, centering around the most conserved residues on each of the functional segments. We elaborate the generic numbers through illustrative examples, focusing on representative drug-binding sites of eukaryotic Nav and Cav channels. We also extend the numbering scheme to compare common disease mutations among different Nav subtypes. Application of the generic residue numbering scheme affords immediate insights into hotspots for pathogenic mutations and critical loci for drug binding and will facilitate drug discovery targeting Nav and Cav channels.
Collapse
Affiliation(s)
- Xueqin Jin
- Beijing Frontier Research Center for Biological Structures, State Key Laboratory of Membrane Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China.
| | - Jian Huang
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA.
| | - Huan Wang
- Beijing Frontier Research Center for Biological Structures, State Key Laboratory of Membrane Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Kan Wang
- Department of Anesthesiology, China-Japan Friendship Hospital, Beijing 100029, China
| | - Nieng Yan
- Beijing Frontier Research Center for Biological Structures, State Key Laboratory of Membrane Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China; Institute of Bio-Architecture and Bio-Interactions (IBABI), Shenzhen Medical Academy of Research and Translation, Guangming District, Shenzhen, Guangdong Province 518107, China; Shenzhen Bay Laboratory, Guangming District, Shenzhen, Guangdong Province 518132, China.
| |
Collapse
|
40
|
Pei S, Wang N, Mei Z, Zhangsun D, Craik DJ, McIntosh JM, Zhu X, Luo S. Conotoxins Targeting Voltage-Gated Sodium Ion Channels. Pharmacol Rev 2024; 76:828-845. [PMID: 38914468 PMCID: PMC11331937 DOI: 10.1124/pharmrev.123.000923] [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: 09/27/2023] [Revised: 06/17/2024] [Accepted: 06/18/2024] [Indexed: 06/26/2024] Open
Abstract
Voltage-gated sodium (NaV) channels are intimately involved in the generation and transmission of action potentials, and dysfunction of these channels may contribute to nervous system diseases, such as epilepsy, neuropathic pain, psychosis, autism, and cardiac arrhythmia. Many venom peptides selectively act on NaV channels. These include conotoxins, which are neurotoxins secreted by cone snails for prey capture or self-defense but which are also valuable pharmacological tools for the identification and/or treatment of human diseases. Typically, conotoxins contain two or three disulfide bonds, and these internal crossbraces contribute to conotoxins having compact, well defined structures and high stability. Of the conotoxins containing three disulfide bonds, some selectively target mammalian NaV channels and can block, stimulate, or modulate these channels. Such conotoxins have great potential to serve as pharmacological tools for studying the functions and characteristics of NaV channels or as drug leads for neurologic diseases related to NaV channels. Accordingly, discovering or designing conotoxins targeting NaV channels with high potency and selectivity is important. The amino acid sequences, disulfide bond connectivity, and three-dimensional structures are key factors that affect the biological activity of conotoxins, and targeted synthetic modifications of conotoxins can greatly improve their activity and selectivity. This review examines NaV channel-targeted conotoxins, focusing on their structures, activities, and designed modifications, with a view toward expanding their applications. SIGNIFICANCE STATEMENT: NaV channels are crucial in various neurologic diseases. Some conotoxins selectively target NaV channels, causing either blockade or activation, thus enabling their use as pharmacological tools for studying the channels' characteristics and functions. Conotoxins also have promising potential to be developed as drug leads. The disulfide bonds in these peptides are important for stabilizing their structures, thus leading to enhanced specificity and potency. Together, conotoxins targeting NaV channels have both immediate research value and promising future application prospects.
Collapse
Affiliation(s)
- Shengrong Pei
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning, China (S.P., N.W., Z.M., D.Z., X.Z., S.L.); Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, Haikou, China (D.Z., S.L.); Institute for Molecular Bioscience, Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, Queensland, Australia (D.J.C.); Departments of Biology and Psychiatry, University of Utah, Salt Lake City, Utah (J.M.M.); and George E. Wahlen Veterans Affairs Medical Center, Salt Lake City, Utah (J.M.M.)
| | - Nan Wang
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning, China (S.P., N.W., Z.M., D.Z., X.Z., S.L.); Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, Haikou, China (D.Z., S.L.); Institute for Molecular Bioscience, Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, Queensland, Australia (D.J.C.); Departments of Biology and Psychiatry, University of Utah, Salt Lake City, Utah (J.M.M.); and George E. Wahlen Veterans Affairs Medical Center, Salt Lake City, Utah (J.M.M.)
| | - Zaoli Mei
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning, China (S.P., N.W., Z.M., D.Z., X.Z., S.L.); Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, Haikou, China (D.Z., S.L.); Institute for Molecular Bioscience, Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, Queensland, Australia (D.J.C.); Departments of Biology and Psychiatry, University of Utah, Salt Lake City, Utah (J.M.M.); and George E. Wahlen Veterans Affairs Medical Center, Salt Lake City, Utah (J.M.M.)
| | - Dongting Zhangsun
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning, China (S.P., N.W., Z.M., D.Z., X.Z., S.L.); Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, Haikou, China (D.Z., S.L.); Institute for Molecular Bioscience, Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, Queensland, Australia (D.J.C.); Departments of Biology and Psychiatry, University of Utah, Salt Lake City, Utah (J.M.M.); and George E. Wahlen Veterans Affairs Medical Center, Salt Lake City, Utah (J.M.M.)
| | - David J Craik
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning, China (S.P., N.W., Z.M., D.Z., X.Z., S.L.); Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, Haikou, China (D.Z., S.L.); Institute for Molecular Bioscience, Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, Queensland, Australia (D.J.C.); Departments of Biology and Psychiatry, University of Utah, Salt Lake City, Utah (J.M.M.); and George E. Wahlen Veterans Affairs Medical Center, Salt Lake City, Utah (J.M.M.)
| | - J Michael McIntosh
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning, China (S.P., N.W., Z.M., D.Z., X.Z., S.L.); Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, Haikou, China (D.Z., S.L.); Institute for Molecular Bioscience, Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, Queensland, Australia (D.J.C.); Departments of Biology and Psychiatry, University of Utah, Salt Lake City, Utah (J.M.M.); and George E. Wahlen Veterans Affairs Medical Center, Salt Lake City, Utah (J.M.M.)
| | - Xiaopeng Zhu
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning, China (S.P., N.W., Z.M., D.Z., X.Z., S.L.); Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, Haikou, China (D.Z., S.L.); Institute for Molecular Bioscience, Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, Queensland, Australia (D.J.C.); Departments of Biology and Psychiatry, University of Utah, Salt Lake City, Utah (J.M.M.); and George E. Wahlen Veterans Affairs Medical Center, Salt Lake City, Utah (J.M.M.)
| | - Sulan Luo
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning, China (S.P., N.W., Z.M., D.Z., X.Z., S.L.); Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, Haikou, China (D.Z., S.L.); Institute for Molecular Bioscience, Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, Queensland, Australia (D.J.C.); Departments of Biology and Psychiatry, University of Utah, Salt Lake City, Utah (J.M.M.); and George E. Wahlen Veterans Affairs Medical Center, Salt Lake City, Utah (J.M.M.)
| |
Collapse
|
41
|
Goodchild SJ, Ahern CA. Conformational photo-trapping in Na V1.5: Inferring local motions at the "inactivation gate". Biophys J 2024; 123:2167-2175. [PMID: 38664963 PMCID: PMC11309974 DOI: 10.1016/j.bpj.2024.04.017] [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: 12/22/2023] [Revised: 03/22/2024] [Accepted: 04/18/2024] [Indexed: 05/12/2024] Open
Abstract
Rapid and effectual inactivation in voltage-gated sodium channels is required for canonical action-potential firing. This "fast" inactivation arises from swift and reversible protein conformational changes that utilize transmembrane segments and the cytoplasmic linker between channel domains III and IV. Until recently, fast inactivation had been accepted to rely on a "ball-and-chain" mechanism whereby a hydrophobic triplet of DIII-IV amino acids (IFM) impairs conductance by binding to a site in central pore of the channel made available by channel opening. New structures of sodium channels have upended this model. Specifically, cryo-electron microscopic structures of eukaryotic sodium channels depict a peripheral binding site for the IFM motif, outside of the pore, opening the possibility of a yet unidentified allosteric mechanism of fast-inactivation gating. We set out to study fast inactivation by photo-trapping human sodium channels in various functional states under voltage control. This was achieved by genetically encoding the crosslinking unnatural amino acid benzophenone phenylalanine at various sites within the DIII-IV linker in the cardiac sodium channel NaV1.5. These data show dynamic state- and positional-dependent trapping of the transient conformations associated with fast inactivation, each yielding different phenotypes and rates of trapping. These data reveal distinct conformational changes that underlie fast inactivation and point to a dynamic environment around the IFM locus.
Collapse
Affiliation(s)
- Samuel J Goodchild
- Department of Anesthesiology, Pharmacology and Therapeutics, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Christopher A Ahern
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, Iowa.
| |
Collapse
|
42
|
Aman TK, Raman IM. Resurgent current in context: Insights from the structure and function of Na and K channels. Biophys J 2024; 123:1924-1941. [PMID: 38130058 PMCID: PMC11309984 DOI: 10.1016/j.bpj.2023.12.016] [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: 10/14/2023] [Revised: 12/07/2023] [Accepted: 12/18/2023] [Indexed: 12/23/2023] Open
Abstract
Discovered just over 25 years ago in cerebellar Purkinje neurons, resurgent Na current was originally described operationally as a component of voltage-gated Na current that flows upon repolarization from relatively depolarized potentials and speeds recovery from inactivation, increasing excitability. Its presence in many excitable cells and absence from others has raised questions regarding its biophysical and molecular mechanisms. Early studies proposed that Na channels capable of generating resurgent current are subject to a rapid open-channel block by an endogenous blocking protein, which binds upon depolarization and unblocks upon repolarization. Since the time that this mechanism was suggested, many physiological and structural studies of both Na and K channels have revealed aspects of gating and conformational states that provide insights into resurgent current. These include descriptions of domain movements for activation and inactivation, solution of cryo-EM structures with pore-blocking compounds, and identification of native blocking domains, proteins, and modulatory subunits. Such results not only allow the open-channel block hypothesis to be refined but also link it more clearly to research that preceded it. This review considers possible mechanisms for resurgent Na current in the context of earlier and later studies of ion channels and suggests a framework for future research.
Collapse
Affiliation(s)
- Teresa K Aman
- Department of Neurobiology, Northwestern University, Evanston, Illinois
| | - Indira M Raman
- Department of Neurobiology, Northwestern University, Evanston, Illinois.
| |
Collapse
|
43
|
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
|
44
|
Moreira-Junior L, Leal-Cardoso JH, Cassola AC, Carvalho-de-Souza JL. Eugenol and lidocaine inhibit voltage-gated Na + channels from dorsal root ganglion neurons with different mechanisms. Front Pharmacol 2024; 15:1354737. [PMID: 38989141 PMCID: PMC11234063 DOI: 10.3389/fphar.2024.1354737] [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: 12/12/2023] [Accepted: 04/08/2024] [Indexed: 07/12/2024] Open
Abstract
Eugenol (EUG) is a bioactive monoterpenoid used as an analgesic, preservative, and flavoring agent. Our new data show EUG as a voltage-gated Na+ channel (VGSC) inhibitor, comparable but not identical to lidocaine (LID). EUG inhibits both total and only TTX-R voltage-activated Na+ currents (INa) recorded from VGSCs naturally expressed on dorsal root ganglion sensory neurons in rats. Inhibition is quick, fully reversible, and dose-dependent. Our biophysical and pharmacological analyses showed that EUG and LID inhibit VGSCs with different mechanisms. EUG inhibits VGSCs with a dose-response relationship characterized by a Hill coefficient of 2, while this parameter for the inhibition by LID is 1. Furthermore, in a different way from LID, EUG modified the voltage dependence of both the VGSC activation and inactivation processes and the recovery from fast inactivated states and the entry to slow inactivated states. In addition, we suggest that EUG, but not LID, interacts with VGSC pre-open-closed states, according to our data.
Collapse
Affiliation(s)
- Luiz Moreira-Junior
- Department of Anesthesiology, University of Arizona, Tucson, AZ, United States
| | | | - Antonio Carlos Cassola
- Department of Physiology and Biophysics, Biomedical Sciences Institute, University of Sao Paulo, São Paulo, Brazil
| | - Joao Luis Carvalho-de-Souza
- Department of Anesthesiology, University of Arizona, Tucson, AZ, United States
- Department of Physiology and Biophysics, Biomedical Sciences Institute, University of Sao Paulo, São Paulo, Brazil
| |
Collapse
|
45
|
Xiao X, Mei Y, Deng W, Zou G, Hou H, Ji X. Electric Eel Biomimetics for Energy Storage and Conversion. SMALL METHODS 2024; 8:e2201435. [PMID: 36840652 DOI: 10.1002/smtd.202201435] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 02/07/2023] [Indexed: 06/18/2023]
Abstract
The electric eel is known as the most powerful creature to generate electricity with a discharge voltage up to 860 V and peak current up to 1 A. These surprising properties are the results of billions of years of evolution on the electrical biological structure and bulk, and now have triggered great research interest in electric eel biomimetics for designing innovated configurations and components of energy storage and conversion devices. In this review, first, the bioelectrical behavior of electric eels is surveyed, followed by the physiological structure to reveal the discharge characteristics and principles of electric organs and electrocytes. Additionally, underlying electrochemical mechanisms and models for calculating the potential and current of electrocytes are presented. Central to this review is the recent progress of electric-eel-inspired innovations and applications for energy storage and conversion, particularly including novel power sources, triboelectric nanogenerators, and nanochannel ion-selective membranes for salinity gradient energy harvesting. Finally, insights on the challenges at the moment and the perspectives on the future research prospects are critically compiled. It is suggested that energy-related electric eel biomimetics will greatly boost the development of next-generation high performance, green, and functional electronics.
Collapse
Affiliation(s)
- Xiangting Xiao
- State Key Laboratory of Powder Metallurgy, College of Chemistry and Chemical Engineering, Central South University, Changsha, 410083, China
| | - Yu Mei
- State Key Laboratory of Powder Metallurgy, College of Chemistry and Chemical Engineering, Central South University, Changsha, 410083, China
| | - Wentao Deng
- State Key Laboratory of Powder Metallurgy, College of Chemistry and Chemical Engineering, Central South University, Changsha, 410083, China
| | - Guoqiang Zou
- State Key Laboratory of Powder Metallurgy, College of Chemistry and Chemical Engineering, Central South University, Changsha, 410083, China
| | - Hongshuai Hou
- State Key Laboratory of Powder Metallurgy, College of Chemistry and Chemical Engineering, Central South University, Changsha, 410083, China
| | - Xiaobo Ji
- State Key Laboratory of Powder Metallurgy, College of Chemistry and Chemical Engineering, Central South University, Changsha, 410083, China
| |
Collapse
|
46
|
Schott K, Usher SG, Serra O, Carnevale V, Pless SA, Chua HC. Unplugging lateral fenestrations of NALCN reveals a hidden drug binding site within the pore region. Proc Natl Acad Sci U S A 2024; 121:e2401591121. [PMID: 38787877 PMCID: PMC11145269 DOI: 10.1073/pnas.2401591121] [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: 01/23/2024] [Accepted: 04/09/2024] [Indexed: 05/26/2024] Open
Abstract
The sodium (Na+) leak channel (NALCN) is a member of the four-domain voltage-gated cation channel family that includes the prototypical voltage-gated sodium and calcium channels (NaVs and CaVs, respectively). Unlike NaVs and CaVs, which have four lateral fenestrations that serve as routes for lipophilic compounds to enter the central cavity to modulate channel function, NALCN has bulky residues (W311, L588, M1145, and Y1436) that block these openings. Structural data suggest that occluded fenestrations underlie the pharmacological resistance of NALCN, but functional evidence is lacking. To test this hypothesis, we unplugged the fenestrations of NALCN by substituting the four aforementioned residues with alanine (AAAA) and compared the effects of NaV, CaV, and NALCN blockers on both wild-type (WT) and AAAA channels. Most compounds behaved in a similar manner on both channels, but phenytoin and 2-aminoethoxydiphenyl borate (2-APB) elicited additional, distinct responses on AAAA channels. Further experiments using single alanine mutants revealed that phenytoin and 2-APB enter the inner cavity through distinct fenestrations, implying structural specificity to their modes of access. Using a combination of computational and functional approaches, we identified amino acid residues critical for 2-APB activity, supporting the existence of drug binding site(s) within the pore region. Intrigued by the activity of 2-APB and its analogues, we tested compounds containing the diphenylmethane/amine moiety on WT channels. We identified clinically used drugs that exhibited diverse activity, thus expanding the pharmacological toolbox for NALCN. While the low potencies of active compounds reiterate the pharmacological resistance of NALCN, our findings lay the foundation for rational drug design to develop NALCN modulators with refined properties.
Collapse
Affiliation(s)
- Katharina Schott
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen2100, Denmark
| | - Samuel George Usher
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen2100, Denmark
| | - Oscar Serra
- Department of Biology, Temple University, Philadelphia, PA19122
- Institute for Genomics and Evolutionary Medicine, Temple University, Philadelphia, PA19122
- Institute of Computational Molecular Science, Temple University, Philadelphia, PA19122
| | - Vincenzo Carnevale
- Department of Biology, Temple University, Philadelphia, PA19122
- Institute for Genomics and Evolutionary Medicine, Temple University, Philadelphia, PA19122
- Institute of Computational Molecular Science, Temple University, Philadelphia, PA19122
| | - Stephan Alexander Pless
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen2100, Denmark
| | - Han Chow Chua
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen2100, Denmark
| |
Collapse
|
47
|
Huffer K, Tan XF, Fernández-Mariño AI, Dhingra S, Swartz KJ. Dilation of ion selectivity filters in cation channels. Trends Biochem Sci 2024; 49:417-430. [PMID: 38514273 PMCID: PMC11069442 DOI: 10.1016/j.tibs.2024.02.004] [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: 12/12/2023] [Revised: 02/19/2024] [Accepted: 02/23/2024] [Indexed: 03/23/2024]
Abstract
Ion channels establish the voltage gradient across cellular membranes by providing aqueous pathways for ions to selectively diffuse down their concentration gradients. The selectivity of any given channel for its favored ions has conventionally been viewed as a stable property, and in many cation channels, it is determined by an ion-selectivity filter within the external end of the ion-permeation pathway. In several instances, including voltage-activated K+ (Kv) channels, ATP-activated P2X receptor channels, and transient receptor potential (TRP) channels, the ion-permeation pathways have been proposed to dilate in response to persistent activation, dynamically altering ion permeation. Here, we discuss evidence for dynamic ion selectivity, examples where ion selectivity filters exhibit structural plasticity, and opportunities to fill gaps in our current understanding.
Collapse
Affiliation(s)
- Kate Huffer
- Molecular Physiology and Biophysics Section, Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Xiao-Feng Tan
- Molecular Physiology and Biophysics Section, Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ana I Fernández-Mariño
- Molecular Physiology and Biophysics Section, Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Surbhi Dhingra
- Molecular Physiology and Biophysics Section, Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kenton J Swartz
- Molecular Physiology and Biophysics Section, Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
48
|
Bodawatta KH, Hu H, Schalk F, Daniel JM, Maiah G, Koane B, Iova B, Beemelmanns C, Poulsen M, Jønsson KA. Multiple mutations in the Nav1.4 sodium channel of New Guinean toxic birds provide autoresistance to deadly batrachotoxin. Mol Ecol 2024; 33:e16878. [PMID: 36779590 DOI: 10.1111/mec.16878] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 02/02/2023] [Accepted: 02/07/2023] [Indexed: 02/14/2023]
Abstract
Toxicity has evolved multiple times across the tree of life and serves important functions related to hunting, defence and parasite deterrence. Toxins are produced either in situ by the toxic organism itself or associated symbionts, or acquired through diet. The ability to exploit toxins from external sources requires adaptations that prevent toxic effects on the consumer (autoresistance). Here, we examine genomic adaptations that could facilitate autoresistance to the diet-acquired potent neurotoxic alkaloid batrachotoxin (BTX) in New Guinean toxic birds. Our work documents two new toxic bird species and shows that toxic birds carry multiple mutations in the SCN4A gene that are under positive selection. This gene encodes the most common vertebrate muscle Nav channel (Nav1.4). Molecular docking results indicate that some of the mutations that are present in the pore-forming segment of the Nav channel, where BTX binds, could reduce its binding affinity. These mutations should therefore prevent the continuous opening of the sodium channels that BTX binding elicits, thereby preventing muscle paralysis and ultimately death. Although these mutations are different from those present in Neotropical Phyllobates poison dart frogs, they occur in the same segments of the Nav1.4 channel. Consequently, in addition to uncovering a greater diversity of toxic bird species than previously known, our work provides an intriguing example of molecular-level convergent adaptations allowing frogs and birds to ingest and use the same neurotoxin. This suggests that genetically modified Nav1.4 channels represent a key adaptation to BTX tolerance and exploitation across vertebrates.
Collapse
Affiliation(s)
- Kasun H Bodawatta
- Natural History Museum of Denmark, University of Copenhagen, Copenhagen, Denmark
| | - Haofu Hu
- Natural History Museum of Denmark, University of Copenhagen, Copenhagen, Denmark
| | - Felix Schalk
- Chemical Biology of Microbe-Host Interactions, Leibniz Institute for Natural Product Research and Infection Biology e. V., Hans-Knöll-Institute, Jena, Germany
| | - Jan-Martin Daniel
- Chemical Biology of Microbe-Host Interactions, Leibniz Institute for Natural Product Research and Infection Biology e. V., Hans-Knöll-Institute, Jena, Germany
- Institute for Pharmaceutical Microbiology, University of Bonn, Bonn, Germany
| | - Gibson Maiah
- The New Guinea Binatang Research Centre, Madang, Papua New Guinea
| | - Bonny Koane
- The New Guinea Binatang Research Centre, Madang, Papua New Guinea
| | - Bulisa Iova
- PNG National Museum and Art Gallery, Port Moresby, Papua New Guinea
| | - Christine Beemelmanns
- Chemical Biology of Microbe-Host Interactions, Leibniz Institute for Natural Product Research and Infection Biology e. V., Hans-Knöll-Institute, Jena, Germany
- Department Anti-infectives from Microbiota, Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarbrücken, Germany
- Universität des Saarlandes, Saarbrücken, Germany
| | - Michael Poulsen
- Section for Ecology and Evolution, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Knud A Jønsson
- Natural History Museum of Denmark, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
49
|
Zou X, Zhang Z, Lu H, Zhao W, Pan L, Chen Y. Functional effects of drugs and toxins interacting with Na V1.4. Front Pharmacol 2024; 15:1378315. [PMID: 38725668 PMCID: PMC11079311 DOI: 10.3389/fphar.2024.1378315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 04/08/2024] [Indexed: 05/12/2024] Open
Abstract
NaV1.4 is a voltage-gated sodium channel subtype that is predominantly expressed in skeletal muscle cells. It is essential for producing action potentials and stimulating muscle contraction, and mutations in NaV1.4 can cause various muscle disorders. The discovery of the cryo-EM structure of NaV1.4 in complex with β1 has opened new possibilities for designing drugs and toxins that target NaV1.4. In this review, we summarize the current understanding of channelopathies, the binding sites and functions of chemicals including medicine and toxins that interact with NaV1.4. These substances could be considered novel candidate compounds or tools to develop more potent and selective drugs targeting NaV1.4. Therefore, studying NaV1.4 pharmacology is both theoretically and practically meaningful.
Collapse
Affiliation(s)
- Xinyi Zou
- Zhejiang Provincial Key Laboratory of Resources Protection and Innovation of Traditional Chinese Medicine, College of Food and Health, Zhejiang Agriculture and Forestry University, Hangzhou, China
| | - Zixuan Zhang
- Zhejiang Provincial Key Laboratory of Resources Protection and Innovation of Traditional Chinese Medicine, College of Food and Health, Zhejiang Agriculture and Forestry University, Hangzhou, China
| | - Hui Lu
- Zhejiang Provincial Key Laboratory of Resources Protection and Innovation of Traditional Chinese Medicine, College of Food and Health, Zhejiang Agriculture and Forestry University, Hangzhou, China
| | - Wei Zhao
- Zhejiang Provincial Key Laboratory of Resources Protection and Innovation of Traditional Chinese Medicine, College of Food and Health, Zhejiang Agriculture and Forestry University, Hangzhou, China
| | - Lanying Pan
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Yuan Chen
- Zhejiang Provincial Key Laboratory of Resources Protection and Innovation of Traditional Chinese Medicine, College of Food and Health, Zhejiang Agriculture and Forestry University, Hangzhou, China
| |
Collapse
|
50
|
Biswas R, López-Serrano A, Huang HL, Ramirez-Navarro A, Grandinetti G, Heissler S, Deschênes I, Chinthalapudi K. Structural basis of human Na v1.5 gating mechanisms. RESEARCH SQUARE 2024:rs.3.rs-3985999. [PMID: 38659812 PMCID: PMC11042394 DOI: 10.21203/rs.3.rs-3985999/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Voltage-gated Nav1.5 channels are central to the generation and propagation of cardiac action potentials1. Aberrations in their function are associated with a wide spectrum of cardiac diseases including arrhythmias and heart failure2-5. Despite decades of progress in Nav1.5 biology6-8, the lack of structural insights into intracellular regions has hampered our understanding of its gating mechanisms. Here we present three cryo-EM structures of human Nav1.5 in previously unanticipated open states, revealing sequential conformational changes in gating charges of the voltage-sensing domains (VSDs) and several intracellular regions. Despite the channel being in the open state, these structures show the IFM motif repositioned in the receptor site but not dislodged. In particular, our structural findings highlight a dynamic C-terminal domain (CTD) and III-IV linker interaction, which regulates the conformation of VSDs and pore opening. Electrophysiological studies confirm that disrupting this interaction results in the fast inactivation of Nav1.5. Together, our structure-function studies establish a foundation for understanding the gating mechanisms of Nav1.5 and the mechanisms underlying CTD-related channelopathies.
Collapse
|