1
|
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
|
2
|
Zhang Y, Ma K, Fang X, Zhang Y, Miao R, Guan H, Tian J. Targeting ion homeostasis in metabolic diseases: Molecular mechanisms and targeted therapies. Pharmacol Res 2025; 212:107579. [PMID: 39756557 DOI: 10.1016/j.phrs.2025.107579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 12/13/2024] [Accepted: 01/01/2025] [Indexed: 01/07/2025]
Abstract
The incidence of metabolic diseases-hypertension, diabetes, obesity, metabolic dysfunction-associated steatotic liver disease (MASLD), and atherosclerosis-is increasing annually, imposing a significant burden on both human health and the social economy. The occurrence and development of these diseases are closely related to the disruption of ion homeostasis, which is crucial for maintaining cellular functions and metabolic equilibrium. However, the specific mechanism of ion homeostasis in metabolic diseases is still unclear. This article reviews the role of ion homeostasis in the pathogenesis of metabolic diseases and assesses its potential as a therapeutic target. Furthermore, the article explores pharmacological strategies that target ion channels and transporters, including existing drugs and emerging drugs under development. Lastly, the article discusses the development direction of future therapeutic strategies, including the possibility of gene therapy targeting specific ion channels and personalized therapy using novel biomarkers. In summary, targeting ion homeostasis provides a new perspective and potential therapeutic approach for the treatment of metabolic diseases.
Collapse
Affiliation(s)
- Yanjiao Zhang
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Kaile Ma
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Xinyi Fang
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China; Graduate College, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yuxin Zhang
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Runyu Miao
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China; Graduate College, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Huifang Guan
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Jiaxing Tian
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| |
Collapse
|
3
|
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: 7] [Impact Index Per Article: 7.0] [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
|
4
|
Knotts GM, Lile SK, Campbell EM, Agee TA, Liyanage SD, Gwaltney SR, Johnson CN. An all-atom model of the human cardiac sodium channel in a lipid bilayer. Sci Rep 2024; 14:26857. [PMID: 39500978 PMCID: PMC11538489 DOI: 10.1038/s41598-024-78466-4] [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: 08/13/2024] [Accepted: 10/31/2024] [Indexed: 11/08/2024] Open
Abstract
Voltage-gated sodium channels (NaV) are complex macromolecular proteins that are responsible for the initial upstroke of an action potential in excitable cells. Appropriate function is necessary for many physiological processes such as heartbeat, voluntary muscle contraction, nerve conduction, and neurological function. Dysfunction can have life-threatening consequences. During the past decade, there have been significant advancements with ion channel structural characterization by CryoEM, yet descriptions of cytosolic components are often lacking. Many investigations have biophysically characterized reconstituted cytosolic components and their interactions. However, extrapolating the structural alterations and allosteric communication within an intact ion channel can be challenging. To address this, we have developed an all-atom model of the human cardiac sodium channel (NaV1.5) in a lipid bilayer with explicit salt and water. Our simulations contain descriptions of cytosolic components that are poorly predicted by AlphaFold and lacking in many CryoEM structures. Leveraging the latest advancements of the Amber force fields (ff19sb and Lipid21) and water model (OPC), our simulations improved protein backbone torsion angles and generated structural information across time (four independent one-microsecond simulations). Our analysis provided descriptions of lipid and solvent contacts and insight into the C-Terminal Domain - inactivation gate and inactivation gate - latch receptor interactions.
Collapse
Affiliation(s)
- Garrett M Knotts
- Department of Chemistry, Mississippi State University, Starkville, MS, 39759, USA
| | - Spencer K Lile
- Department of Chemistry, Mississippi State University, Starkville, MS, 39759, USA
| | - Emily M Campbell
- Department of Chemistry, Mississippi State University, Starkville, MS, 39759, USA
| | - Taylor A Agee
- Department of Chemistry, Mississippi State University, Starkville, MS, 39759, USA
| | - Senal D Liyanage
- Department of Chemistry, Mississippi State University, Starkville, MS, 39759, USA
| | - Steven R Gwaltney
- Department of Chemistry, Mississippi State University, Starkville, MS, 39759, USA
| | | |
Collapse
|
5
|
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: 0] [Impact Index Per Article: 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
|
6
|
Lopez-Mateos D, Narang K, Yarov-Yarovoy V. Exploring voltage-gated sodium channel conformations and protein-protein interactions using AlphaFold2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.15.618559. [PMID: 39463944 PMCID: PMC11507785 DOI: 10.1101/2024.10.15.618559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Voltage-gated sodium (NaV) channels are vital regulators of electrical activity in excitable cells, playing critical roles in generating and propagating action potentials. Given their importance in physiology, NaV channels are key therapeutic targets for treating numerous conditions, yet developing subtype-selective drugs remains challenging due to the high sequence and structural conservation among NaV family members. Recent advances in cryo-electron microscopy have resolved nearly all human NaV channels, providing valuable insights into their structure and function. However, limitations persist in fully capturing the complex conformational states that underlie NaV channel gating and modulation. This study explores the capability of AlphaFold2 to sample multiple NaV channel conformations and assess AlphaFold Multimer's accuracy in modeling interactions between the NaV α-subunit and its protein partners, including auxiliary β-subunits and calmodulin. We enhance conformational sampling to explore NaV channel conformations using a subsampled multiple sequence alignment approach and varying the number of recycles. Our results demonstrate that AlphaFold2 models multiple NaV channel conformations, including those from experimental structures, new states not yet experimentally identified, and potential intermediate states. Furthermore, AlphaFold Multimer models NaV complexes with auxiliary β-subunits and calmodulin with high accuracy, and the presence of protein partners significantly alters the conformational landscape of the NaV α-subunit. These findings highlight the potential of deep learning-based methods to expand our understanding of NaV channel structure, gating, and modulation, with significant implications for future drug discovery efforts.
Collapse
Affiliation(s)
- Diego Lopez-Mateos
- Department of Physiology and Membrane Biology, University of California School of Medicine, Davis, CA 95616
- Biophysics Graduate Group, University of California School of Medicine, Davis, CA 95616
| | - Kush Narang
- Department of Physiology and Membrane Biology, University of California School of Medicine, Davis, CA 95616
| | - Vladimir Yarov-Yarovoy
- Department of Physiology and Membrane Biology, University of California School of Medicine, Davis, CA 95616
- Biophysics Graduate Group, University of California School of Medicine, Davis, CA 95616
- Department of Anesthesiology and Pain Medicine, University of California School of Medicine, Davis, CA 95616
| |
Collapse
|
7
|
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
|
8
|
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
|
9
|
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
|
10
|
Denomme N, Hernandez CC, Bock HA, Ohana RF, Bakshi S, Sherwood AM, McCorvy JD, Daley PF, Callaway WB, Hull JM, Alt A, Isom LL, Cozzi NV. N-(4-Bromo-2,5-Dimethoxyphenethyl)-6-(4-Phenylbutoxy)Hexan-1-Amine (XOB): A Novel Phenylalkylamine Antagonist of Serotonin 2A Receptors and Voltage-Gated Sodium Channels. Mol Pharmacol 2024; 106:92-106. [PMID: 38821630 PMCID: PMC11254453 DOI: 10.1124/molpharm.123.000837] [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: 11/02/2023] [Revised: 05/17/2024] [Accepted: 05/23/2024] [Indexed: 06/02/2024] Open
Abstract
Bipolar disorder impacts millions of patients in the United States but the mechanistic understanding of its pathophysiology and therapeutics is incomplete. Atypical antipsychotic serotonin2A (5-HT2A) receptor antagonists, such as quetiapine and olanzapine, and mood-stabilizing voltage-gated sodium channel (VGSC) blockers, such as lamotrigine, carbamazepine, and valproate, show therapeutic synergy and are often prescribed in combination for the treatment of bipolar disorder. Combination therapy is a complex task for clinicians and patients, often resulting in unexpected difficulties with dosing, drug tolerances, and decreased patient compliance. Thus, an unmet need for bipolar disorder treatment is to develop a therapeutic agent that targets both 5-HT2A receptors and VGSCs. Toward this goal, we developed a novel small molecule that simultaneously antagonizes 5-HT2A receptors and blocks sodium current. The new compound, N-(4-bromo-2,5-dimethoxyphenethyl)-6-(4-phenylbutoxy)hexan-1-amine (XOB) antagonizes 5-HT-stimulated, Gq-mediated, calcium flux at 5-HT2A receptors at low micromolar concentrations while displaying negligible affinity and activity at 5-HT1A, 5-HT2B, and 5-HT2C receptors. At similar concentrations, XOB administration inhibits sodium current in heterologous cells and results in reduced action potential (AP) firing and VGSC-related AP properties in mouse prefrontal cortex layer V pyramidal neurons. Thus, XOB represents a new, proof-of-principle tool that can be used for future preclinical investigations and therapeutic development. This polypharmacology approach of developing a single molecule to act upon two targets, which are currently independently targeted by combination therapies, may lead to safer alternatives for the treatment of psychiatric disorders that are increasingly being found to benefit from the simultaneous targeting of multiple receptors. SIGNIFICANCE STATEMENT: The authors synthesized a novel small molecule (XOB) that simultaneously antagonizes two key therapeutic targets of bipolar disorder, 5-HT2A receptors and voltage-gated sodium channels, in heterologous cells, and inhibits the intrinsic excitability of mouse prefrontal cortex layer V pyramidal neurons in brain slices. XOB represents a valuable new proof-of-principle tool for future preclinical investigations and provides a novel molecular approach to the pharmacological treatment of complex neuropsychiatric disease, which often requires a combination of therapeutics for sufficient patient benefit.
Collapse
Affiliation(s)
- Nicholas Denomme
- Department of Pharmacology (N.D., S.B., A.A., L.L.I.), Michigan Psychedelic Center (M-PsyC) (N.D., L.L.I.), Life Sciences Institute (C.C.H., A.A.), University of Michigan, Ann Arbor, Michigan; Promega Corporation, Fitchburg, Wisconsin (R.F.O.); Usona Institute, Fitchburg, Wisconsin (A.M.S.); Department of Cell Biology, Neurobiology, and Anatomy. Medical College of Wisconsin, Milwaukee, Wisconsin (H.A.B., J.D.M.); Department of Neurology and Neurologic Sciences, Stanford University, Stanford, California (J.M.H.); and Alexander Shulgin Research Institute, Lafayette, California (P.F.D., W.B.C., N.V.C.)
| | - Ciria C Hernandez
- Department of Pharmacology (N.D., S.B., A.A., L.L.I.), Michigan Psychedelic Center (M-PsyC) (N.D., L.L.I.), Life Sciences Institute (C.C.H., A.A.), University of Michigan, Ann Arbor, Michigan; Promega Corporation, Fitchburg, Wisconsin (R.F.O.); Usona Institute, Fitchburg, Wisconsin (A.M.S.); Department of Cell Biology, Neurobiology, and Anatomy. Medical College of Wisconsin, Milwaukee, Wisconsin (H.A.B., J.D.M.); Department of Neurology and Neurologic Sciences, Stanford University, Stanford, California (J.M.H.); and Alexander Shulgin Research Institute, Lafayette, California (P.F.D., W.B.C., N.V.C.)
| | - Hailey A Bock
- Department of Pharmacology (N.D., S.B., A.A., L.L.I.), Michigan Psychedelic Center (M-PsyC) (N.D., L.L.I.), Life Sciences Institute (C.C.H., A.A.), University of Michigan, Ann Arbor, Michigan; Promega Corporation, Fitchburg, Wisconsin (R.F.O.); Usona Institute, Fitchburg, Wisconsin (A.M.S.); Department of Cell Biology, Neurobiology, and Anatomy. Medical College of Wisconsin, Milwaukee, Wisconsin (H.A.B., J.D.M.); Department of Neurology and Neurologic Sciences, Stanford University, Stanford, California (J.M.H.); and Alexander Shulgin Research Institute, Lafayette, California (P.F.D., W.B.C., N.V.C.)
| | - Rachel F Ohana
- Department of Pharmacology (N.D., S.B., A.A., L.L.I.), Michigan Psychedelic Center (M-PsyC) (N.D., L.L.I.), Life Sciences Institute (C.C.H., A.A.), University of Michigan, Ann Arbor, Michigan; Promega Corporation, Fitchburg, Wisconsin (R.F.O.); Usona Institute, Fitchburg, Wisconsin (A.M.S.); Department of Cell Biology, Neurobiology, and Anatomy. Medical College of Wisconsin, Milwaukee, Wisconsin (H.A.B., J.D.M.); Department of Neurology and Neurologic Sciences, Stanford University, Stanford, California (J.M.H.); and Alexander Shulgin Research Institute, Lafayette, California (P.F.D., W.B.C., N.V.C.)
| | - Shreeya Bakshi
- Department of Pharmacology (N.D., S.B., A.A., L.L.I.), Michigan Psychedelic Center (M-PsyC) (N.D., L.L.I.), Life Sciences Institute (C.C.H., A.A.), University of Michigan, Ann Arbor, Michigan; Promega Corporation, Fitchburg, Wisconsin (R.F.O.); Usona Institute, Fitchburg, Wisconsin (A.M.S.); Department of Cell Biology, Neurobiology, and Anatomy. Medical College of Wisconsin, Milwaukee, Wisconsin (H.A.B., J.D.M.); Department of Neurology and Neurologic Sciences, Stanford University, Stanford, California (J.M.H.); and Alexander Shulgin Research Institute, Lafayette, California (P.F.D., W.B.C., N.V.C.)
| | - Alexander M Sherwood
- Department of Pharmacology (N.D., S.B., A.A., L.L.I.), Michigan Psychedelic Center (M-PsyC) (N.D., L.L.I.), Life Sciences Institute (C.C.H., A.A.), University of Michigan, Ann Arbor, Michigan; Promega Corporation, Fitchburg, Wisconsin (R.F.O.); Usona Institute, Fitchburg, Wisconsin (A.M.S.); Department of Cell Biology, Neurobiology, and Anatomy. Medical College of Wisconsin, Milwaukee, Wisconsin (H.A.B., J.D.M.); Department of Neurology and Neurologic Sciences, Stanford University, Stanford, California (J.M.H.); and Alexander Shulgin Research Institute, Lafayette, California (P.F.D., W.B.C., N.V.C.)
| | - John D McCorvy
- Department of Pharmacology (N.D., S.B., A.A., L.L.I.), Michigan Psychedelic Center (M-PsyC) (N.D., L.L.I.), Life Sciences Institute (C.C.H., A.A.), University of Michigan, Ann Arbor, Michigan; Promega Corporation, Fitchburg, Wisconsin (R.F.O.); Usona Institute, Fitchburg, Wisconsin (A.M.S.); Department of Cell Biology, Neurobiology, and Anatomy. Medical College of Wisconsin, Milwaukee, Wisconsin (H.A.B., J.D.M.); Department of Neurology and Neurologic Sciences, Stanford University, Stanford, California (J.M.H.); and Alexander Shulgin Research Institute, Lafayette, California (P.F.D., W.B.C., N.V.C.)
| | - Paul F Daley
- Department of Pharmacology (N.D., S.B., A.A., L.L.I.), Michigan Psychedelic Center (M-PsyC) (N.D., L.L.I.), Life Sciences Institute (C.C.H., A.A.), University of Michigan, Ann Arbor, Michigan; Promega Corporation, Fitchburg, Wisconsin (R.F.O.); Usona Institute, Fitchburg, Wisconsin (A.M.S.); Department of Cell Biology, Neurobiology, and Anatomy. Medical College of Wisconsin, Milwaukee, Wisconsin (H.A.B., J.D.M.); Department of Neurology and Neurologic Sciences, Stanford University, Stanford, California (J.M.H.); and Alexander Shulgin Research Institute, Lafayette, California (P.F.D., W.B.C., N.V.C.)
| | - Wyeth B Callaway
- Department of Pharmacology (N.D., S.B., A.A., L.L.I.), Michigan Psychedelic Center (M-PsyC) (N.D., L.L.I.), Life Sciences Institute (C.C.H., A.A.), University of Michigan, Ann Arbor, Michigan; Promega Corporation, Fitchburg, Wisconsin (R.F.O.); Usona Institute, Fitchburg, Wisconsin (A.M.S.); Department of Cell Biology, Neurobiology, and Anatomy. Medical College of Wisconsin, Milwaukee, Wisconsin (H.A.B., J.D.M.); Department of Neurology and Neurologic Sciences, Stanford University, Stanford, California (J.M.H.); and Alexander Shulgin Research Institute, Lafayette, California (P.F.D., W.B.C., N.V.C.)
| | - Jacob M Hull
- Department of Pharmacology (N.D., S.B., A.A., L.L.I.), Michigan Psychedelic Center (M-PsyC) (N.D., L.L.I.), Life Sciences Institute (C.C.H., A.A.), University of Michigan, Ann Arbor, Michigan; Promega Corporation, Fitchburg, Wisconsin (R.F.O.); Usona Institute, Fitchburg, Wisconsin (A.M.S.); Department of Cell Biology, Neurobiology, and Anatomy. Medical College of Wisconsin, Milwaukee, Wisconsin (H.A.B., J.D.M.); Department of Neurology and Neurologic Sciences, Stanford University, Stanford, California (J.M.H.); and Alexander Shulgin Research Institute, Lafayette, California (P.F.D., W.B.C., N.V.C.)
| | - Andrew Alt
- Department of Pharmacology (N.D., S.B., A.A., L.L.I.), Michigan Psychedelic Center (M-PsyC) (N.D., L.L.I.), Life Sciences Institute (C.C.H., A.A.), University of Michigan, Ann Arbor, Michigan; Promega Corporation, Fitchburg, Wisconsin (R.F.O.); Usona Institute, Fitchburg, Wisconsin (A.M.S.); Department of Cell Biology, Neurobiology, and Anatomy. Medical College of Wisconsin, Milwaukee, Wisconsin (H.A.B., J.D.M.); Department of Neurology and Neurologic Sciences, Stanford University, Stanford, California (J.M.H.); and Alexander Shulgin Research Institute, Lafayette, California (P.F.D., W.B.C., N.V.C.)
| | - Lori L Isom
- Department of Pharmacology (N.D., S.B., A.A., L.L.I.), Michigan Psychedelic Center (M-PsyC) (N.D., L.L.I.), Life Sciences Institute (C.C.H., A.A.), University of Michigan, Ann Arbor, Michigan; Promega Corporation, Fitchburg, Wisconsin (R.F.O.); Usona Institute, Fitchburg, Wisconsin (A.M.S.); Department of Cell Biology, Neurobiology, and Anatomy. Medical College of Wisconsin, Milwaukee, Wisconsin (H.A.B., J.D.M.); Department of Neurology and Neurologic Sciences, Stanford University, Stanford, California (J.M.H.); and Alexander Shulgin Research Institute, Lafayette, California (P.F.D., W.B.C., N.V.C.)
| | - Nicholas V Cozzi
- Department of Pharmacology (N.D., S.B., A.A., L.L.I.), Michigan Psychedelic Center (M-PsyC) (N.D., L.L.I.), Life Sciences Institute (C.C.H., A.A.), University of Michigan, Ann Arbor, Michigan; Promega Corporation, Fitchburg, Wisconsin (R.F.O.); Usona Institute, Fitchburg, Wisconsin (A.M.S.); Department of Cell Biology, Neurobiology, and Anatomy. Medical College of Wisconsin, Milwaukee, Wisconsin (H.A.B., J.D.M.); Department of Neurology and Neurologic Sciences, Stanford University, Stanford, California (J.M.H.); and Alexander Shulgin Research Institute, Lafayette, California (P.F.D., W.B.C., N.V.C.)
| |
Collapse
|
11
|
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] [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
|
12
|
Stary-Weinzinger A. In silico models of the macromolecular Na V1.5-K IR2.1 complex. Front Physiol 2024; 15:1362964. [PMID: 38468705 PMCID: PMC10925717 DOI: 10.3389/fphys.2024.1362964] [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: 12/29/2023] [Accepted: 02/07/2024] [Indexed: 03/13/2024] Open
Abstract
In cardiac cells, the expression of the cardiac voltage-gated Na+ channel (NaV1.5) is reciprocally regulated with the inward rectifying K+ channel (KIR2.1). These channels can form macromolecular complexes that pre-assemble early during forward trafficking (transport to the cell membrane). In this study, we present in silico 3D models of NaV1.5-KIR2.1, generated by rigid-body protein-protein docking programs and deep learning-based AlphaFold-Multimer software. Modeling revealed that the two channels could physically interact with each other along the entire transmembrane region. Structural mapping of disease-associated mutations revealed a hotspot at this interface with several trafficking-deficient variants in close proximity. Thus, examining the role of disease-causing variants is important not only in isolated channels but also in the context of macromolecular complexes. These findings may contribute to a better understanding of the life-threatening cardiovascular diseases underlying KIR2.1 and NaV1.5 malfunctions.
Collapse
Affiliation(s)
- Anna Stary-Weinzinger
- Division of Pharmacology and Toxicology, Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria
| |
Collapse
|
13
|
Kanithi M, Kumari L, Yalakaturi K, Munjal K, Jimitreddy S, Kandamuri M, Veeramachineni P, Chopra H, Junapudi S. Nanoparticle Polymers Influence on Cardiac Health: Good or Bad for Cardiac Physiology? Curr Probl Cardiol 2024; 49:102145. [PMID: 37852559 DOI: 10.1016/j.cpcardiol.2023.102145] [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: 10/06/2023] [Accepted: 10/14/2023] [Indexed: 10/20/2023]
Abstract
Cardiovascular diseases (CVD) are one of the leading causes of death and morbidity worldwide. Lifestyle modifications, medications, and addressing epidemiological factors have long been at the forefront of targeting therapeutics for CVD. Treatments can be further complicated given the intersection of gender, age, unique comorbidities, and healthcare access, among many other factors. Therefore, expanding treatment and diagnostic modalities for CVD is absolutely necessary. Nanoparticles and nanomaterials are increasingly being used as therapeutic and diagnostic modalities in various disciplines of biomedicine. Nanoparticles have multiple ways of interacting with the cardiovascular system. Some of them alter cardiac physiology by impacting ion channels, whereas others influence ions directly or indirectly, improving cellular death via decreasing oxidative stress. While embedding nanoparticles into therapeutics can help enhance healthy cardiovascular function in other scenarios, they can also impair physiology by increasing reactive oxidative species and leading to cardiotoxicity. This review explores different types of nanoparticles, their effects, and the applicable dosages to create a better foundation for understanding the current research findings.
Collapse
Affiliation(s)
- Manasa Kanithi
- Michigan State University College of Osteopathic Medicine, East Lansing, MI
| | - Lata Kumari
- People University of Medical and Health Sciences, Nawab Shah, Sindh, Pakistan
| | | | - Kavita Munjal
- Amity Institute of Pharmacy, Amity University, Noida, Uttar Pradesh, India
| | | | | | | | - Hitesh Chopra
- Department of Biosciences, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu, India.
| | - Sunil Junapudi
- Geethanjali College of Pharmacy, Hyderabad, Telangana, India.
| |
Collapse
|
14
|
Heigl T, Netzer MA, Zanetti L, Ganglberger M, Fernández-Quintero ML, Koschak A. Characterization of two pathological gating-charge substitutions in Cav1.4 L-type calcium channels. Channels (Austin) 2023; 17:2192360. [PMID: 36943941 PMCID: PMC10038055 DOI: 10.1080/19336950.2023.2192360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 03/14/2023] [Indexed: 03/23/2023] Open
Abstract
Cav1.4 L-type calcium channels are predominantly expressed at the photoreceptor terminals and in bipolar cells, mediating neurotransmitter release. Mutations in its gene, CACNA1F, can cause congenital stationary night-blindness type 2 (CSNB2). Due to phenotypic variability in CSNB2, characterization of pathological variants is necessary to better determine pathological mechanism at the site of action. A set of known mutations affects conserved gating charges in the S4 voltage sensor, two of which have been found in male CSNB2 patients. Here, we describe two disease-causing Cav1.4 mutations with gating charge neutralization, exchanging an arginine 964 with glycine (RG) or arginine 1288 with leucine (RL). In both, charge neutralization was associated with a reduction channel expression also reflected in smaller ON gating currents. In RL channels, the strong decrease in whole-cell current densities might additionally be explained by a reduction of single-channel currents. We further identified alterations in their biophysical properties, such as a hyperpolarizing shift of the activation threshold and an increase in slope factor of activation and inactivation. Molecular dynamic simulations in RL substituted channels indicated water wires in both, resting and active, channel states, suggesting the development of omega (ω)currents as a new pathological mechanism in CSNB2. This sum of the respective channel property alterations might add to the differential symptoms in patients beside other factors, such as genomic and environmental deviations.
Collapse
Affiliation(s)
- Thomas Heigl
- University of Innsbruck, Institute of Pharmacy, Pharmacology and Toxicology, Innsbruck, Austria
| | - Michael A. Netzer
- University of Innsbruck, Institute of Pharmacy, Pharmacology and Toxicology, Innsbruck, Austria
| | - Lucia Zanetti
- University of Innsbruck, Institute of Pharmacy, Pharmacology and Toxicology, Innsbruck, Austria
| | - Matthias Ganglberger
- University of Innsbruck, Institute of Pharmacy, Pharmacology and Toxicology, Innsbruck, Austria
| | - Monica L. Fernández-Quintero
- Institute of General, Inorganic and Theoretical Chemistry, Center for Chemistry and Biomedicine, University of Innsbruck, Innsbruck, Austria
- Division of Pharmacology and Toxicology, Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria
| | - Alexandra Koschak
- University of Innsbruck, Institute of Pharmacy, Pharmacology and Toxicology, Innsbruck, Austria
| |
Collapse
|
15
|
Ma JG, Vandenberg JI, Ng CA. Development of automated patch clamp assays to overcome the burden of variants of uncertain significance in inheritable arrhythmia syndromes. Front Physiol 2023; 14:1294741. [PMID: 38089476 PMCID: PMC10712320 DOI: 10.3389/fphys.2023.1294741] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 11/13/2023] [Indexed: 10/16/2024] Open
Abstract
Advances in next-generation sequencing have been exceptionally valuable for identifying variants in medically actionable genes. However, for most missense variants there is insufficient evidence to permit definitive classification of variants as benign or pathogenic. To overcome the deluge of Variants of Uncertain Significance, there is an urgent need for high throughput functional assays to assist with the classification of variants. Advances in parallel planar patch clamp technologies has enabled the development of automated high throughput platforms capable of increasing throughput 10- to 100-fold compared to manual patch clamp methods. Automated patch clamp electrophysiology is poised to revolutionize the field of functional genomics for inheritable cardiac ion channelopathies. In this review, we outline i) the evolution of patch clamping, ii) the development of high-throughput automated patch clamp assays to assess cardiac ion channel variants, iii) clinical application of these assays and iv) where the field is heading.
Collapse
Affiliation(s)
- Joanne G. Ma
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | - Jamie I. Vandenberg
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | - Chai-Ann Ng
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
16
|
Yang W, Wang W, Cai S, Li P, Zhang D, Ning J, Ke J, Hou A, Chen L, Ma Y, Jin W. Synthesis and In Vivo Antiarrhythmic Activity Evaluation of Novel Scutellarein Analogues as Voltage-Gated Nav1.5 and Cav1.2 Channels Blockers. Molecules 2023; 28:7417. [PMID: 37959836 PMCID: PMC10650756 DOI: 10.3390/molecules28217417] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 10/21/2023] [Accepted: 10/31/2023] [Indexed: 11/15/2023] Open
Abstract
Malignant cardiac arrhythmias with high morbidity and mortality have posed a significant threat to our human health. Scutellarein, a metabolite of Scutellarin which is isolated from Scutellaria altissima L., presents excellent therapeutic effects on cardiovascular diseases and could further be metabolized into methylated forms. A series of 22 new scutellarein derivatives with hydroxyl-substitution based on the scutellarin metabolite in vivo was designed, synthesized via the conjugation of the scutellarein scaffold with pharmacophores of FDA-approved antiarrhythmic medications and evaluated for their antiarrhythmic activity through the analyzation of the rat number of arrhythmia recovery, corresponding to the recovery time and maintenance time in the rat model of barium chloride-induced arrhythmia, as well as the cumulative dosage of aconitine required to induce VP, VT, VF and CA in the rat model of aconitine-induced arrhythmia. All designed compounds could shorten the time of the arrhythmia continuum induced by barium chloride, indicating that 4'-hydroxy substituents of scutellarein had rapid-onset antiarrhythmic effects. In addition, nearly all of the compounds could normalize the HR, RR, QRS, QT and QTc interval, as well as the P/T waves' amplitude. The most promising compound 10e showed the best antiarrhythmic activity with long-term efficacy and extremely low cytotoxicity, better than the positive control scutellarein. This result was also approved by the computational docking simulation. Most importantly, patch clamp measurements on Nav1.5 and Cav1.2 channels indicated that compound 10e was able to reduce the INa and ICa in a concentration-dependent manner and left-shifted the inactivation curve of Nav1.5. Taken together, all compounds were considered to be antiarrhythmic. Compound 10e even showed no proarrhythmic effect and could be classified as Ib Vaughan Williams antiarrhythmic agents. What is more, compound 10e did not block the hERG potassium channel which highly associated with cardiotoxicity.
Collapse
Affiliation(s)
- Wei Yang
- Key Laboratory of External Drug Delivery System and Preparation Technology in Universities of Yunnan, Yunnan University of Chinese Medicine, Kunming 650500, China (D.Z.)
- Faculty of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Wenping Wang
- Key Laboratory of External Drug Delivery System and Preparation Technology in Universities of Yunnan, Yunnan University of Chinese Medicine, Kunming 650500, China (D.Z.)
- Faculty of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Song Cai
- Department of Anatomy and Histology, Shenzhen University Medical School, Shenzhen 518060, China
| | - Peng Li
- School of Food and Drug, Shenzhen Polytechnic, Shenzhen 518000, China
| | - Die Zhang
- Key Laboratory of External Drug Delivery System and Preparation Technology in Universities of Yunnan, Yunnan University of Chinese Medicine, Kunming 650500, China (D.Z.)
- Faculty of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Jinhua Ning
- Key Laboratory of External Drug Delivery System and Preparation Technology in Universities of Yunnan, Yunnan University of Chinese Medicine, Kunming 650500, China (D.Z.)
- Faculty of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Jin Ke
- Key Laboratory of External Drug Delivery System and Preparation Technology in Universities of Yunnan, Yunnan University of Chinese Medicine, Kunming 650500, China (D.Z.)
- Faculty of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Anguo Hou
- Key Laboratory of External Drug Delivery System and Preparation Technology in Universities of Yunnan, Yunnan University of Chinese Medicine, Kunming 650500, China (D.Z.)
- Faculty of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Linyun Chen
- Key Laboratory of External Drug Delivery System and Preparation Technology in Universities of Yunnan, Yunnan University of Chinese Medicine, Kunming 650500, China (D.Z.)
- Faculty of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Yunshu Ma
- Key Laboratory of External Drug Delivery System and Preparation Technology in Universities of Yunnan, Yunnan University of Chinese Medicine, Kunming 650500, China (D.Z.)
- Faculty of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Wenbin Jin
- Key Laboratory of External Drug Delivery System and Preparation Technology in Universities of Yunnan, Yunnan University of Chinese Medicine, Kunming 650500, China (D.Z.)
- Faculty of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming 650500, China
- State Key Laboratory of Chemical Biology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR, China
- Drug Discovery and Department of Applied Biology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR, China
- Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR, China
| |
Collapse
|
17
|
Frosio A, Micaglio E, Polsinelli I, Calamaio S, Melgari D, Prevostini R, Ghiroldi A, Binda A, Carrera P, Villa M, Mastrocinque F, Presi S, Salerno R, Boccellino A, Anastasia L, Ciconte G, Ricagno S, Pappone C, Rivolta I. Unravelling Novel SCN5A Mutations Linked to Brugada Syndrome: Functional, Structural, and Genetic Insights. Int J Mol Sci 2023; 24:15089. [PMID: 37894777 PMCID: PMC10606416 DOI: 10.3390/ijms242015089] [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: 09/08/2023] [Revised: 10/08/2023] [Accepted: 10/09/2023] [Indexed: 10/29/2023] Open
Abstract
Brugada Syndrome (BrS) is a rare inherited cardiac arrhythmia causing potentially fatal ventricular tachycardia or fibrillation, mainly occurring during rest or sleep in young individuals without heart structural issues. It increases the risk of sudden cardiac death, and its characteristic feature is an abnormal ST segment elevation on the ECG. While BrS has diverse genetic origins, a subset of cases can be conducted to mutations in the SCN5A gene, which encodes for the Nav1.5 sodium channel. Our study focused on three novel SCN5A mutations (p.A344S, p.N347K, and p.D349N) found in unrelated BrS families. Using patch clamp experiments, we found that these mutations disrupted sodium currents: p.A344S reduced current density, while p.N347K and p.D349N completely abolished it, leading to altered voltage dependence and inactivation kinetics when co-expressed with normal channels. We also explored the effects of mexiletine treatment, which can modulate ion channel function. Interestingly, the p.N347K and p.D349N mutations responded well to the treatment, rescuing the current density, while p.A344S showed a limited response. Structural analysis revealed these mutations were positioned in key regions of the channel, impacting its stability and function. This research deepens our understanding of BrS by uncovering the complex relationship between genetic mutations, ion channel behavior, and potential therapeutic interventions.
Collapse
Affiliation(s)
- Anthony Frosio
- Institute of Molecular and Translational Cardiology (IMTC), IRCCS Policlinico San Donato, 20097 San Donato Milanese, Italy; (A.F.); (E.M.); (I.P.); (S.C.); (D.M.); (R.P.); (A.G.); (M.V.); (L.A.); (G.C.); (S.R.); (C.P.)
| | - Emanuele Micaglio
- Institute of Molecular and Translational Cardiology (IMTC), IRCCS Policlinico San Donato, 20097 San Donato Milanese, Italy; (A.F.); (E.M.); (I.P.); (S.C.); (D.M.); (R.P.); (A.G.); (M.V.); (L.A.); (G.C.); (S.R.); (C.P.)
- Arrhythmia and Electrophysiology Department, IRCCS Policlinico San Donato, 20097 San Donato Milanese, Italy; (F.M.); (A.B.)
| | - Ivan Polsinelli
- Institute of Molecular and Translational Cardiology (IMTC), IRCCS Policlinico San Donato, 20097 San Donato Milanese, Italy; (A.F.); (E.M.); (I.P.); (S.C.); (D.M.); (R.P.); (A.G.); (M.V.); (L.A.); (G.C.); (S.R.); (C.P.)
| | - Serena Calamaio
- Institute of Molecular and Translational Cardiology (IMTC), IRCCS Policlinico San Donato, 20097 San Donato Milanese, Italy; (A.F.); (E.M.); (I.P.); (S.C.); (D.M.); (R.P.); (A.G.); (M.V.); (L.A.); (G.C.); (S.R.); (C.P.)
| | - Dario Melgari
- Institute of Molecular and Translational Cardiology (IMTC), IRCCS Policlinico San Donato, 20097 San Donato Milanese, Italy; (A.F.); (E.M.); (I.P.); (S.C.); (D.M.); (R.P.); (A.G.); (M.V.); (L.A.); (G.C.); (S.R.); (C.P.)
| | - Rachele Prevostini
- Institute of Molecular and Translational Cardiology (IMTC), IRCCS Policlinico San Donato, 20097 San Donato Milanese, Italy; (A.F.); (E.M.); (I.P.); (S.C.); (D.M.); (R.P.); (A.G.); (M.V.); (L.A.); (G.C.); (S.R.); (C.P.)
| | - Andrea Ghiroldi
- Institute of Molecular and Translational Cardiology (IMTC), IRCCS Policlinico San Donato, 20097 San Donato Milanese, Italy; (A.F.); (E.M.); (I.P.); (S.C.); (D.M.); (R.P.); (A.G.); (M.V.); (L.A.); (G.C.); (S.R.); (C.P.)
- Laboratory of Stem Cells for Tissue Engineering, IRCCS Policlinico San Donato, 20097 San Donato Milanese, Italy
| | - Anna Binda
- School of Medicine and Surgery, University of Milano-Bicocca, Via Cadore, 48, 20900 Monza, Italy;
| | - Paola Carrera
- Laboratory of Clinical Molecular Genetics and Cytogenetics, Unit of Genomics for Diagnosis of Human Diseases, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; (P.C.); (S.P.)
| | - Marco Villa
- Institute of Molecular and Translational Cardiology (IMTC), IRCCS Policlinico San Donato, 20097 San Donato Milanese, Italy; (A.F.); (E.M.); (I.P.); (S.C.); (D.M.); (R.P.); (A.G.); (M.V.); (L.A.); (G.C.); (S.R.); (C.P.)
| | - Flavio Mastrocinque
- Arrhythmia and Electrophysiology Department, IRCCS Policlinico San Donato, 20097 San Donato Milanese, Italy; (F.M.); (A.B.)
| | - Silvia Presi
- Laboratory of Clinical Molecular Genetics and Cytogenetics, Unit of Genomics for Diagnosis of Human Diseases, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; (P.C.); (S.P.)
| | - Raffaele Salerno
- Faculty of Medicine and Surgery, Vita-Salute San Raffaele University, Via Olgettina, 58, 20132 Milan, Italy;
| | - Antonio Boccellino
- Arrhythmia and Electrophysiology Department, IRCCS Policlinico San Donato, 20097 San Donato Milanese, Italy; (F.M.); (A.B.)
| | - Luigi Anastasia
- Institute of Molecular and Translational Cardiology (IMTC), IRCCS Policlinico San Donato, 20097 San Donato Milanese, Italy; (A.F.); (E.M.); (I.P.); (S.C.); (D.M.); (R.P.); (A.G.); (M.V.); (L.A.); (G.C.); (S.R.); (C.P.)
- Laboratory of Stem Cells for Tissue Engineering, IRCCS Policlinico San Donato, 20097 San Donato Milanese, Italy
- Faculty of Medicine and Surgery, Vita-Salute San Raffaele University, Via Olgettina, 58, 20132 Milan, Italy;
| | - Giuseppe Ciconte
- Institute of Molecular and Translational Cardiology (IMTC), IRCCS Policlinico San Donato, 20097 San Donato Milanese, Italy; (A.F.); (E.M.); (I.P.); (S.C.); (D.M.); (R.P.); (A.G.); (M.V.); (L.A.); (G.C.); (S.R.); (C.P.)
- Arrhythmia and Electrophysiology Department, IRCCS Policlinico San Donato, 20097 San Donato Milanese, Italy; (F.M.); (A.B.)
- Faculty of Medicine and Surgery, Vita-Salute San Raffaele University, Via Olgettina, 58, 20132 Milan, Italy;
| | - Stefano Ricagno
- Institute of Molecular and Translational Cardiology (IMTC), IRCCS Policlinico San Donato, 20097 San Donato Milanese, Italy; (A.F.); (E.M.); (I.P.); (S.C.); (D.M.); (R.P.); (A.G.); (M.V.); (L.A.); (G.C.); (S.R.); (C.P.)
- Department of Biosciences, Università degli Studi di Milano, Via Celoria, 26, 20133 Milan, Italy
| | - Carlo Pappone
- Institute of Molecular and Translational Cardiology (IMTC), IRCCS Policlinico San Donato, 20097 San Donato Milanese, Italy; (A.F.); (E.M.); (I.P.); (S.C.); (D.M.); (R.P.); (A.G.); (M.V.); (L.A.); (G.C.); (S.R.); (C.P.)
- Arrhythmia and Electrophysiology Department, IRCCS Policlinico San Donato, 20097 San Donato Milanese, Italy; (F.M.); (A.B.)
- Faculty of Medicine and Surgery, Vita-Salute San Raffaele University, Via Olgettina, 58, 20132 Milan, Italy;
| | - Ilaria Rivolta
- Institute of Molecular and Translational Cardiology (IMTC), IRCCS Policlinico San Donato, 20097 San Donato Milanese, Italy; (A.F.); (E.M.); (I.P.); (S.C.); (D.M.); (R.P.); (A.G.); (M.V.); (L.A.); (G.C.); (S.R.); (C.P.)
- School of Medicine and Surgery, University of Milano-Bicocca, Via Cadore, 48, 20900 Monza, Italy;
| |
Collapse
|
18
|
Hussein RA, Ahmed M, Heinemann SH. Selenomethionine mis-incorporation and redox-dependent voltage-gated sodium channel gain of function. J Neurochem 2023; 167:262-276. [PMID: 37679952 DOI: 10.1111/jnc.15957] [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: 05/03/2023] [Revised: 08/21/2023] [Accepted: 08/22/2023] [Indexed: 09/09/2023]
Abstract
Selenomethionine (SeMet) readily replaces methionine (Met) residues in proteins during translation. Long-term dietary SeMet intake results in the accumulation of the amino acid in tissue proteins. Despite the high rates of SeMet incorporation in proteins and its stronger susceptibility to oxidation compared to Met, little is known about the effect of SeMet mis-incorporation on electrical excitability and ion channels. Fast inactivation of voltage-gated sodium (NaV ) channels is essential for exact action potential shaping with even minute impairment of inactivation resulting in a plethora of adverse phenotypes. Met oxidation of the NaV channel inactivation motif (Ile-Phe-Met) and further Met residues causes a marked loss of inactivation. Here, we examined the impact of SeMet mis-incorporation on the function of NaV channels. While extensive SeMet incorporation into recombinant rat NaV 1.4 channels preserved their normal function, it greatly sensitized the channels to mild oxidative stress, resulting in loss of inactivation and diminished maximal current, both reversible by dithiothreitol-induced reduction. SeMet incorporation similarly affected human NaV 1.4, NaV 1.2, NaV 1.5, and NaV 1.7. In mouse dorsal root ganglia (DRG) neurons, 1 day of SeMet exposure exacerbated the oxidation-mediated broadening of action potentials. SeMet-treated DRGs also exhibited a stronger increase in the persistent NaV current in response to oxidation. SeMet incorporation in NaV proteins coinciding with oxidative insults may therefore result in hyperexcitability pathologies, such as cardiac arrhythmias and neuropathies, like congenital NaV channel gain-of-function mutations.
Collapse
Affiliation(s)
- Rama A Hussein
- Center for Molecular Biomedicine, Department of Biophysics, Friedrich Schiller University Jena and Jena University Hospital, Jena, Germany
| | - Marwa Ahmed
- Center for Molecular Biomedicine, Department of Biophysics, Friedrich Schiller University Jena and Jena University Hospital, Jena, Germany
| | - Stefan H Heinemann
- Center for Molecular Biomedicine, Department of Biophysics, Friedrich Schiller University Jena and Jena University Hospital, Jena, Germany
| |
Collapse
|
19
|
Kalinina AD, Rogachevskii IV, Samosvat DM, Zegrya GG, Butkevich IP, Mikhailenko VA, Plakhova VB, Penniyaynen VA, Podzorova SA, Krylov BV. Analgesic Effect of the Lysine-Containing Short Peptide Is Due to Modulation of the Na V1.8 Channel Activation Gating System. Life (Basel) 2023; 13:1800. [PMID: 37763204 PMCID: PMC10533133 DOI: 10.3390/life13091800] [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: 07/08/2023] [Revised: 08/10/2023] [Accepted: 08/22/2023] [Indexed: 09/29/2023] Open
Abstract
The present work continues our recent series of articles that aim to elucidate the ligand-receptor binding mechanism of short cationic peptides to the NaV1.8 channel in the nociceptive neuron. The applied methodological approach has involved several methods: the patch-clamp experimental evaluation of the effective charge of the NaV1.8 channel activation gating system, the organotypic tissue culture method, the formalin test, and theoretical conformational analysis. The lysine-containing short peptide Ac-KEKK-NH2 has been shown to effectively modulate the NaV1.8 channel activation gating system. As demonstrated by the organotypic tissue culture method, the studied short peptide does not trigger the downstream signaling cascades controlling neurite outgrowth and should not be expected to evoke adverse side effects. Conformational analysis of the Ac-KEKK-NH2 molecule has revealed that the distances between the positively charged amino groups of the lysine side chains are equal to 11-12 Å. According to the previously suggested mechanism of ligand-receptor binding of short peptides to the NaV1.8 channel molecule, Ac-KEKK-NH2 should exhibit an analgesic effect, which has been confirmed by the formalin test. The data obtained unequivocally indicate that the studied lysine-containing short peptide is a promising candidate for the role of a novel analgesic medicinal substance.
Collapse
Affiliation(s)
- Arina D. Kalinina
- Pavlov Institute of Physiology, Russian Academy of Sciences, 199034 Saint Petersburg, Russia (V.B.P.)
| | - Ilya V. Rogachevskii
- Pavlov Institute of Physiology, Russian Academy of Sciences, 199034 Saint Petersburg, Russia (V.B.P.)
| | - Dmitriy M. Samosvat
- Ioffe Institute, Russian Academy of Sciences, 194021 Saint Petersburg, Russia
| | - Georgy G. Zegrya
- Pavlov Institute of Physiology, Russian Academy of Sciences, 199034 Saint Petersburg, Russia (V.B.P.)
- Ioffe Institute, Russian Academy of Sciences, 194021 Saint Petersburg, Russia
| | - Irina P. Butkevich
- Pavlov Institute of Physiology, Russian Academy of Sciences, 199034 Saint Petersburg, Russia (V.B.P.)
| | - Viktor A. Mikhailenko
- Pavlov Institute of Physiology, Russian Academy of Sciences, 199034 Saint Petersburg, Russia (V.B.P.)
| | - Vera B. Plakhova
- Pavlov Institute of Physiology, Russian Academy of Sciences, 199034 Saint Petersburg, Russia (V.B.P.)
| | - Valentina A. Penniyaynen
- Pavlov Institute of Physiology, Russian Academy of Sciences, 199034 Saint Petersburg, Russia (V.B.P.)
| | - Svetlana A. Podzorova
- Pavlov Institute of Physiology, Russian Academy of Sciences, 199034 Saint Petersburg, Russia (V.B.P.)
| | - Boris V. Krylov
- Pavlov Institute of Physiology, Russian Academy of Sciences, 199034 Saint Petersburg, Russia (V.B.P.)
| |
Collapse
|
20
|
Ruiz-Fernández AR, Campos L, Villanelo F, Garate JA, Perez-Acle T. Protein-Mediated Electroporation in a Cardiac Voltage-Sensing Domain Due to an nsPEF Stimulus. Int J Mol Sci 2023; 24:11397. [PMID: 37511161 PMCID: PMC10379607 DOI: 10.3390/ijms241411397] [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: 04/29/2023] [Revised: 06/15/2023] [Accepted: 07/05/2023] [Indexed: 07/30/2023] Open
Abstract
This study takes a step in understanding the physiological implications of the nanosecond pulsed electric field (nsPEF) by integrating molecular dynamics simulations and machine learning techniques. nsPEF, a state-of-the-art technology, uses high-voltage electric field pulses with a nanosecond duration to modulate cellular activity. This investigation reveals a relatively new and underexplored phenomenon: protein-mediated electroporation. Our research focused on the voltage-sensing domain (VSD) of the NaV1.5 sodium cardiac channel in response to nsPEF stimulation. We scrutinized the VSD structures that form pores and thereby contribute to the physical chemistry that governs the defibrillation effect of nsPEF. To do so, we conducted a comprehensive analysis involving the clustering of 142 replicas simulated for 50 ns under nsPEF stimuli. We subsequently pinpointed the representative structures of each cluster and computed the free energy between them. We find that the selected VSD of NaV1.5 forms pores under nsPEF stimulation, but in a way that significant differs from the traditional VSD opening. This study not only extends our understanding of nsPEF and its interaction with protein channels but also adds a new effect to further study.
Collapse
Affiliation(s)
| | - Leonardo Campos
- Computational Biology Lab, Fundación Ciencia & Vida, Santiago 7780272, Chile
- Facultad de Ingeniería y Tecnología, Universidad San Sebastián, Santiago 8420524, Chile
| | - Felipe Villanelo
- Computational Biology Lab, Fundación Ciencia & Vida, Santiago 7780272, Chile
- Facultad de Ingeniería y Tecnología, Universidad San Sebastián, Santiago 8420524, Chile
| | - Jose Antonio Garate
- Computational Biology Lab, Fundación Ciencia & Vida, Santiago 7780272, Chile
- Facultad de Ingeniería y Tecnología, Universidad San Sebastián, Santiago 8420524, Chile
- Millennium Nucleus im NanoBioPhysics, Universidad de Valparaiso, Valparaiso 2351319, Chile
| | - Tomas Perez-Acle
- Computational Biology Lab, Fundación Ciencia & Vida, Santiago 7780272, Chile
- Facultad de Ingeniería y Tecnología, Universidad San Sebastián, Santiago 8420524, Chile
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaiso 2360102, Chile
| |
Collapse
|
21
|
Huang J, Fan X, Jin X, Jo S, Zhang HB, Fujita A, Bean BP, Yan N. Cannabidiol inhibits Na v channels through two distinct binding sites. Nat Commun 2023; 14:3613. [PMID: 37330538 PMCID: PMC10276812 DOI: 10.1038/s41467-023-39307-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 06/07/2023] [Indexed: 06/19/2023] Open
Abstract
Cannabidiol (CBD), a major non-psychoactive phytocannabinoid in cannabis, is an effective treatment for some forms of epilepsy and pain. At high concentrations, CBD interacts with a huge variety of proteins, but which targets are most relevant for clinical actions is still unclear. Here we show that CBD interacts with Nav1.7 channels at sub-micromolar concentrations in a state-dependent manner. Electrophysiological experiments show that CBD binds to the inactivated state of Nav1.7 channels with a dissociation constant of about 50 nM. The cryo-EM structure of CBD bound to Nav1.7 channels reveals two distinct binding sites. One is in the IV-I fenestration near the upper pore. The other binding site is directly next to the inactivated "wedged" position of the Ile/Phe/Met (IFM) motif on the short linker between repeats III and IV, which mediates fast inactivation. Consistent with producing a direct stabilization of the inactivated state, mutating residues in this binding site greatly reduced state-dependent binding of CBD. The identification of this binding site may enable design of compounds with improved properties compared to CBD itself.
Collapse
Affiliation(s)
- Jian Huang
- Department of Molecular Biology, Princeton University, Princeton, NJ, 08544, USA
| | - Xiao Fan
- Department of Molecular Biology, Princeton University, Princeton, NJ, 08544, USA
| | - 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
| | - Sooyeon Jo
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA, 02115, USA
| | - Hanxiong Bear Zhang
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA, 02115, USA
| | - Akie Fujita
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA, 02115, USA
| | - Bruce P Bean
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA, 02115, USA.
| | - Nieng Yan
- Department of Molecular Biology, Princeton University, Princeton, NJ, 08544, USA.
- 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.
| |
Collapse
|
22
|
Wu Q, Huang J, Fan X, Wang K, Jin X, Huang G, Li J, Pan X, Yan N. Structural mapping of Na v1.7 antagonists. Nat Commun 2023; 14:3224. [PMID: 37270609 DOI: 10.1038/s41467-023-38942-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 05/22/2023] [Indexed: 06/05/2023] Open
Abstract
Voltage-gated sodium (Nav) channels are targeted by a number of widely used and investigational drugs for the treatment of epilepsy, arrhythmia, pain, and other disorders. Despite recent advances in structural elucidation of Nav channels, the binding mode of most Nav-targeting drugs remains unknown. Here we report high-resolution cryo-EM structures of human Nav1.7 treated with drugs and lead compounds with representative chemical backbones at resolutions of 2.6-3.2 Å. A binding site beneath the intracellular gate (site BIG) accommodates carbamazepine, bupivacaine, and lacosamide. Unexpectedly, a second molecule of lacosamide plugs into the selectivity filter from the central cavity. Fenestrations are popular sites for various state-dependent drugs. We show that vinpocetine, a synthetic derivative of a vinca alkaloid, and hardwickiic acid, a natural product with antinociceptive effect, bind to the III-IV fenestration, while vixotrigine, an analgesic candidate, penetrates the IV-I fenestration of the pore domain. Our results permit building a 3D structural map for known drug-binding sites on Nav channels summarized from the present and previous structures.
Collapse
Affiliation(s)
- Qiurong Wu
- Beijing Frontier Research Center for Biological Structures, 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.
| | - Xiao Fan
- Department of Molecular Biology, Princeton University, Princeton, NJ, 08544, USA.
| | - Kan Wang
- Department of Anesthesiology, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Xueqin Jin
- Beijing Frontier Research Center for Biological Structures, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Gaoxingyu Huang
- Westlake Laboratory of Life Sciences and Biomedicine, Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, 18 Shilongshan Road, Hangzhou, 310024, Zhejiang Province, China
- Institute of Biology, Westlake Institute for Advanced Study, 18 Shilongshan Road, Hangzhou, 310024, Zhejiang Province, China
| | - Jiaao Li
- Beijing Frontier Research Center for Biological Structures, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Xiaojing Pan
- Beijing Frontier Research Center for Biological Structures, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China.
| | - Nieng Yan
- Beijing Frontier Research Center for Biological Structures, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China.
- Department of Molecular Biology, Princeton University, Princeton, NJ, 08544, USA.
- Shenzhen Medical Academy of Research and Translation, Guangming District, Shenzhen, 518107, Guangdong Province, China.
| |
Collapse
|
23
|
Negami T, Terada T. Calculations of the binding free energies of the Comprehensive in vitro Proarrhythmia Assay (CiPA) reference drugs to cardiac ion channels. Biophys Physicobiol 2023; 20:e200016. [PMID: 38496247 PMCID: PMC10941965 DOI: 10.2142/biophysico.bppb-v20.0016] [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: 01/17/2023] [Accepted: 03/24/2023] [Indexed: 03/19/2024] Open
Abstract
The evaluation of the inhibitory activities of drugs on multiple cardiac ion channels is required for the accurate assessment of proarrhythmic risks. Moreover, the in silico prediction of such inhibitory activities of drugs on cardiac channels can improve the efficiency of the drug-development process. Here, we performed molecular docking simulations to predict the complex structures of 25 reference drugs that were proposed by the Comprehensive in vitro Proarrhythmia Assay consortium using two cardiac ion channels, the human ether-a-go-go-related gene (hERG) potassium channel and human NaV1.5 (hNaV1.5) sodium channel, with experimentally available structures. The absolute binding free energy (ΔGbind) values of the predicted structures were calculated by a molecular dynamics-based method and compared with the experimental half-maximal inhibitory concentration (IC50) data. Furthermore, the regression analysis between the calculated values and negative of the common logarithm of the experimental IC50 values (pIC50) revealed that the calculated values of four and ten drugs deviated significantly from the regression lines of the hERG and hNaV1.5 channels, respectively. We reconsidered the docking poses and protonation states of the drugs based on the experimental data and recalculated their ΔGbind values. Finally, the calculated ΔGbind values of 24 and 19 drugs correlated with their experimental pIC50 values (coefficients of determination=0.791 and 0.613 for the hERG and hNaV1.5 channels, respectively). Thus, the regression analysis between the calculated ΔGbind and experimental IC50 data ensured the realization of an increased number of reliable complex structures.
Collapse
Affiliation(s)
- Tatsuki Negami
- Department of Biotechnology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Tohru Terada
- Department of Biotechnology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-8657, Japan
| |
Collapse
|
24
|
O'Neill MJ, Sala L, Denjoy I, Wada Y, Kozek K, Crotti L, Dagradi F, Kotta MC, Spazzolini C, Leenhardt A, Salem JE, Kashiwa A, Ohno S, Tao R, Roden DM, Horie M, Extramiana F, Schwartz PJ, Kroncke BM. Continuous Bayesian variant interpretation accounts for incomplete penetrance among Mendelian cardiac channelopathies. Genet Med 2023; 25:100355. [PMID: 36496179 PMCID: PMC9992222 DOI: 10.1016/j.gim.2022.12.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 12/05/2022] [Accepted: 12/05/2022] [Indexed: 12/12/2022] Open
Abstract
PURPOSE The congenital Long QT Syndrome (LQTS) and Brugada Syndrome (BrS) are Mendelian autosomal dominant diseases that frequently precipitate fatal cardiac arrhythmias. Incomplete penetrance is a barrier to clinical management of heterozygotes harboring variants in the major implicated disease genes KCNQ1, KCNH2, and SCN5A. We apply and evaluate a Bayesian penetrance estimation strategy that accounts for this phenomenon. METHODS We generated Bayesian penetrance models for KCNQ1-LQT1 and SCN5A-LQT3 using variant-specific features and clinical data from the literature, international arrhythmia genetic centers, and population controls. We analyzed the distribution of posterior penetrance estimates across 4 genotype-phenotype relationships and compared continuous estimates with ClinVar annotations. Posterior estimates were mapped onto protein structure. RESULTS Bayesian penetrance estimates of KCNQ1-LQT1 and SCN5A-LQT3 are empirically equivalent to 10 and 5 clinically phenotype heterozygotes, respectively. Posterior penetrance estimates were bimodal for KCNQ1-LQT1 and KCNH2-LQT2, with a higher fraction of missense variants with high penetrance among KCNQ1 variants. There was a wide distribution of variant penetrance estimates among identical ClinVar categories. Structural mapping revealed heterogeneity among "hot spot" regions and featured high penetrance estimates for KCNQ1 variants in contact with calmodulin and the S6 domain. CONCLUSIONS Bayesian penetrance estimates provide a continuous framework for variant interpretation.
Collapse
Affiliation(s)
- Matthew J O'Neill
- Vanderbilt University School of Medicine, Medical Scientist Training Program, Vanderbilt University, Nashville, TN
| | - Luca Sala
- IRCCS, Istituto Auxologico Italiano, Center for Cardiac Arrhythmias of Genetic Origin and Laboratory of Cardiovascular Genetics, Milano, Italy
| | - Isabelle Denjoy
- Department of Cardiovascular Medicine, Hôpital Bichat, APHP, Université de Paris Cité, Paris, France
| | - Yuko Wada
- Vanderbilt Center for Arrhythmia Research and Therapeutics (VanCART), Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Krystian Kozek
- Vanderbilt University School of Medicine, Medical Scientist Training Program, Vanderbilt University, Nashville, TN
| | - Lia Crotti
- IRCCS, Istituto Auxologico Italiano, Center for Cardiac Arrhythmias of Genetic Origin and Laboratory of Cardiovascular Genetics, Milano, Italy
| | - Federica Dagradi
- IRCCS, Istituto Auxologico Italiano, Center for Cardiac Arrhythmias of Genetic Origin and Laboratory of Cardiovascular Genetics, Milano, Italy
| | - Maria-Christina Kotta
- IRCCS, Istituto Auxologico Italiano, Center for Cardiac Arrhythmias of Genetic Origin and Laboratory of Cardiovascular Genetics, Milano, Italy
| | - Carla Spazzolini
- IRCCS, Istituto Auxologico Italiano, Center for Cardiac Arrhythmias of Genetic Origin and Laboratory of Cardiovascular Genetics, Milano, Italy
| | - Antoine Leenhardt
- Department of Cardiovascular Medicine, Hôpital Bichat, APHP, Université de Paris Cité, Paris, France
| | - Joe-Elie Salem
- Department of Cardiovascular Medicine, Hôpital Bichat, APHP, Université de Paris Cité, Paris, France
| | - Asami Kashiwa
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine Kyoto, Japan
| | - Seiko Ohno
- Department of Bioscience and Genetics, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Ran Tao
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN
| | - Dan M Roden
- Vanderbilt Center for Arrhythmia Research and Therapeutics (VanCART), Departments of Medicine, Pharmacology, and Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN
| | - Minoru Horie
- Department of Cardiovascular Medicine, Shiga University of Medical Science, Shiga, Japan
| | - Fabrice Extramiana
- Department of Cardiovascular Medicine, Hôpital Bichat, APHP, Université de Paris Cité, Paris, France
| | - Peter J Schwartz
- IRCCS, Istituto Auxologico Italiano, Center for Cardiac Arrhythmias of Genetic Origin and Laboratory of Cardiovascular Genetics, Milano, Italy
| | - Brett M Kroncke
- Vanderbilt Center for Arrhythmia Research and Therapeutics (VanCART), Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN.
| |
Collapse
|
25
|
Salvage SC, Jeevaratnam K, Huang CL, Jackson AP. Cardiac sodium channel complexes and arrhythmia: structural and functional roles of the β1 and β3 subunits. J Physiol 2023; 601:923-940. [PMID: 36354758 PMCID: PMC10953345 DOI: 10.1113/jp283085] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 11/04/2022] [Indexed: 11/12/2022] Open
Abstract
In cardiac myocytes, the voltage-gated sodium channel NaV 1.5 opens in response to membrane depolarisation and initiates the action potential. The NaV 1.5 channel is typically associated with regulatory β-subunits that modify gating and trafficking behaviour. These β-subunits contain a single extracellular immunoglobulin (Ig) domain, a single transmembrane α-helix and an intracellular region. Here we focus on the role of the β1 and β3 subunits in regulating NaV 1.5. We catalogue β1 and β3 domain specific mutations that have been associated with inherited cardiac arrhythmia, including Brugada syndrome, long QT syndrome, atrial fibrillation and sudden death. We discuss how new structural insights into these proteins raises new questions about physiological function.
Collapse
Affiliation(s)
| | | | - Christopher L.‐H. Huang
- Department of BiochemistryUniversity of CambridgeCambridgeUK
- Department of PhysiologyDevelopment and NeuroscienceUniversity of CambridgeCambridgeUK
| | | |
Collapse
|
26
|
Wang G, Xu L, Chen H, Liu Y, Pan P, Hou T. Recent advances in computational studies on voltage‐gated sodium channels: Drug design and mechanism studies. WIRES COMPUTATIONAL MOLECULAR SCIENCE 2023. [DOI: 10.1002/wcms.1641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/18/2023]
Affiliation(s)
- Gaoang Wang
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University College of Pharmaceutical Sciences, Zhejiang University Hangzhou Zhejiang China
| | - Lei Xu
- Institute of Bioinformatics and Medical Engineering School of Electrical and Information Engineering, Jiangsu University of Technology Changzhou Jiangsu China
| | - Haiyi Chen
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University College of Pharmaceutical Sciences, Zhejiang University Hangzhou Zhejiang China
| | - Yifei Liu
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University College of Pharmaceutical Sciences, Zhejiang University Hangzhou Zhejiang China
| | - Peichen Pan
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University College of Pharmaceutical Sciences, Zhejiang University Hangzhou Zhejiang China
| | - Tingjun Hou
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University College of Pharmaceutical Sciences, Zhejiang University Hangzhou Zhejiang China
| |
Collapse
|
27
|
Abstract
Voltage-gated sodium channel Nav1.6 plays a crucial role in neuronal firing in the central nervous system (CNS). Aberrant function of Nav1.6 may lead to epilepsy and other neurological disorders. Specific inhibitors of Nav1.6 thus have therapeutic potentials. Here we present the cryo-EM structure of human Nav1.6 in the presence of auxiliary subunits β1 and fibroblast growth factor homologous factor 2B (FHF2B) at an overall resolution of 3.1 Å. The overall structure represents an inactivated state with closed pore domain (PD) and all "up" voltage-sensing domains. A conserved carbohydrate-aromatic interaction involving Trp302 and Asn326, together with the β1 subunit, stabilizes the extracellular loop in repeat I. Apart from regular lipids that are resolved in the EM map, an unprecedented Y-shaped density that belongs to an unidentified molecule binds to the PD, revealing a potential site for developing Nav1.6-specific blockers. Structural mapping of disease-related Nav1.6 mutations provides insights into their pathogenic mechanism.
Collapse
|
28
|
Gu Y, Wang J, Li M, Zhong F, Xiang J, Xu Z. Inhibitory Effects of Nobiletin on Voltage-Gated Na + Channel in Rat Ventricular Myocytes Based on Electrophysiological Analysis and Molecular Docking Method. Int J Mol Sci 2022; 23:ijms232315175. [PMID: 36499507 PMCID: PMC9736168 DOI: 10.3390/ijms232315175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 11/27/2022] [Accepted: 11/30/2022] [Indexed: 12/07/2022] Open
Abstract
Nobiletin (NOB) has attracted much attention owing to its outstanding bioactivities. This study aimed to investigate its anti-arrhythmic effect through electrophysiological and molecular docking studies. We assessed the anti-arrhythmic effects of NOB using aconitine-induced ventricular arrhythmia in a rat model and the electrophysiological effects of NOB on rat cardiomyocytes utilizing whole-cell patch-clamp techniques. Moreover, we investigated the binding characters of NOB with rNav1.5, rNav1.5/QQQ, and hNaV1.5 via docking analysis, comparing them with amiodarone and aconitine. NOB pretreatment delayed susceptibility to ventricular premature and ventricular tachycardia and decreased the incidence of fatal ventricular fibrillation. Whole-cell patch-clamp assays demonstrated that the peak current density of the voltage-gated Na+ channel current was reversibly reduced by NOB in a concentration-dependent manner. The steady-state activation and recovery curves were shifted in the positive direction along the voltage axis, and the steady-state inactivation curve was shifted in the negative direction along the voltage axis, as shown by gating kinetics. The molecular docking study showed NOB formed a π-π stacking interaction with rNav1.5 and rNav1.5/QQQ upon Phe-1762, which is the homolog to Phe-1760 in hNaV1.5 and plays an important role in antiarrhythmic action This study reveals that NOB may act as a class I sodium channel anti-arrhythmia agent.
Collapse
Affiliation(s)
- Youwei Gu
- Department of Pharmacology, School of Medicine, Yangzhou University, Yangzhou 225009, China
| | - Jieru Wang
- Department of Pharmacology, School of Medicine, Yangzhou University, Yangzhou 225009, China
| | - Mengting Li
- Department of Pharmacology, School of Medicine, Yangzhou University, Yangzhou 225009, China
| | - Fei Zhong
- Department of Pharmacology, School of Medicine, Yangzhou University, Yangzhou 225009, China
| | - Jie Xiang
- Department of Pharmacology, School of Medicine, Yangzhou University, Yangzhou 225009, China
- Correspondence: (Z.X.); (J.X.)
| | - Zhengxin Xu
- Department of Pharmacology, School of Medicine, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
- Jingsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou 225001, China
- Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research, Yangzhou 225009, China
- Yeda Institute of Gene and Cell Therapy, Taizhou 318000, China
- Correspondence: (Z.X.); (J.X.)
| |
Collapse
|
29
|
Gamal El-Din TM. When the Gates Swing Open Only: Arrhythmia Mutations That Target the Fast Inactivation Gate of Na v1.5. Cells 2022; 11:cells11233714. [PMID: 36496974 PMCID: PMC9735811 DOI: 10.3390/cells11233714] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 11/17/2022] [Accepted: 11/17/2022] [Indexed: 11/23/2022] Open
Abstract
Nav1.5 is the main voltage-gated sodium channel found in cardiac muscle, where it facilitates the fast influx of Na+ ions across the cell membrane, resulting in the fast depolarization phase-phase 0 of the cardiac action potential. As a result, it plays a major role in determining the amplitude and the upstroke velocity of the cardiac impulse. Quantitively, cardiac sodium channel activates in less than a millisecond to trigger the cardiac action potential and inactivates within 2-3 ms to facilitate repolarization and return to the resting state in preparation for firing the next action potential. Missense mutations in the gene that encodes Nav1.5 (SCN5A), change these time constants which leads to a wide spectrum of cardiac diseases ranging from long QT syndrome type 3 (LQT3) to sudden cardiac death. In this mini-review I will focus on the missense mutations in the inactivation gate of Nav1.5 that results in arrhythmia, attempting to correlate the location of the missense mutation to their specific phenotype.
Collapse
|
30
|
Wang G, Yu J, Du H, Shen C, Zhang X, Liu Y, Zhang Y, Cao D, Pan P, Hou T. VGSC-DB: an online database of voltage-gated sodium channels. J Cheminform 2022; 14:75. [PMID: 36320030 PMCID: PMC9628066 DOI: 10.1186/s13321-022-00655-y] [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: 09/01/2022] [Accepted: 10/18/2022] [Indexed: 11/26/2022] Open
Abstract
As an important member of ion channels family, the voltage-gated sodium channel (VGSC/Nav) is associated with a variety of diseases, including epilepsy, migraine, ataxia, etc., and has always been a hot target for drug design and discovery. Many subtype-selective modulators targeting VGSCs have been reported, and some of them have been approved for clinical applications. However, the drug design resources related to VGSCs are insufficient, especially the lack of accurate and extensive compound data toward VGSCs. To fulfill this demand, we develop the Voltage-gated Sodium Channels Database (VGSC-DB). VGSC-DB is the first open-source database for VGSCs, which provides open access to 6055 data records, including 3396 compounds from 173 references toward nine subtypes of Navs (Nav1.1 ~ Nav1.9). A total of 28 items of information is included in each data record, including the chemical structure, biological activity (IC50/EC50), target, binding site, organism, chemical and physical properties, etc. VGSC-DB collects the data from small-molecule compounds, toxins and various derivatives. Users can search the information of compounds by text or structure, and the advanced search function is also supported to realize batch query. VGSC-DB is freely accessible at http://cadd.zju.edu.cn/vgsc/ , and all the data can be downloaded in XLSX/SDF file formats.
Collapse
Affiliation(s)
- Gaoang Wang
- grid.13402.340000 0004 1759 700XInnovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Zhejiang University, Hangzhou, 310058 Zhejiang China
| | - Jiahui Yu
- grid.13402.340000 0004 1759 700XInnovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Zhejiang University, Hangzhou, 310058 Zhejiang China
| | - Hongyan Du
- grid.13402.340000 0004 1759 700XInnovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Zhejiang University, Hangzhou, 310058 Zhejiang China
| | - Chao Shen
- grid.13402.340000 0004 1759 700XInnovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Zhejiang University, Hangzhou, 310058 Zhejiang China
| | - Xujun Zhang
- grid.13402.340000 0004 1759 700XInnovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Zhejiang University, Hangzhou, 310058 Zhejiang China
| | - Yifei Liu
- grid.13402.340000 0004 1759 700XInnovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Zhejiang University, Hangzhou, 310058 Zhejiang China
| | - Yangyang Zhang
- grid.13402.340000 0004 1759 700XInnovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Zhejiang University, Hangzhou, 310058 Zhejiang China
| | - Dongsheng Cao
- grid.216417.70000 0001 0379 7164Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410004 Hunan China
| | - Peichen Pan
- grid.13402.340000 0004 1759 700XInnovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Zhejiang University, Hangzhou, 310058 Zhejiang China
| | - Tingjun Hou
- grid.13402.340000 0004 1759 700XInnovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Zhejiang University, Hangzhou, 310058 Zhejiang China
| |
Collapse
|
31
|
Unwinding and spiral sliding of S4 and domain rotation of VSD during the electromechanical coupling in Na v1.7. Proc Natl Acad Sci U S A 2022; 119:e2209164119. [PMID: 35878056 PMCID: PMC9388133 DOI: 10.1073/pnas.2209164119] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Nav1.7 has been targeted for pain management for its well-established role in pain sensation. Hundreds of mutations of Nav1.7 have been found in patients with pain disorders. Structures of Nav1.7 captured in different conformations will reveal its working mechanism and facilitate drug discovery. Here we present the rational design of a Nav1.7 variant, Nav1.7-M11, that may be trapped in the closed-state inactivation conformation at 0 mV. Cryoelectron microscopy analysis of Nav1.7-M11 reveals voltage-sensing domain in the first repeat (VSDI) in the completely down conformation, VSDII at an intermediate state, and the pore domain tightly closed. Structural comparison of Nav1.7-M11 with the WT channel provides unprecedented insight into the electromechanical coupling details and affords mechanistic interpretation for a number of pain-related mutations. Voltage-gated sodium (Nav) channel Nav1.7 has been targeted for the development of nonaddictive pain killers. Structures of Nav1.7 in distinct functional states will offer an advanced mechanistic understanding and aid drug discovery. Here we report the cryoelectron microscopy analysis of a human Nav1.7 variant that, with 11 rationally introduced point mutations, has a markedly right-shifted activation voltage curve with V1/2 reaching 69 mV. The voltage-sensing domain in the first repeat (VSDI) in a 2.7-Å resolution structure displays a completely down (deactivated) conformation. Compared to the structure of WT Nav1.7, three gating charge (GC) residues in VSDI are transferred to the cytosolic side through a combination of helix unwinding and spiral sliding of S4I and ∼20° domain rotation. A conserved WNФФD motif on the cytoplasmic end of S3I stabilizes the down conformation of VSDI. One GC residue is transferred in VSDII mainly through helix sliding. Accompanying GC transfer in VSDI and VSDII, rearrangement and contraction of the intracellular gate is achieved through concerted movements of adjacent segments, including S4-5I, S4-5II, S5II, and all S6 segments. Our studies provide important insight into the electromechanical coupling mechanism of the single-chain voltage-gated ion channels and afford molecular interpretations for a number of pain-associated mutations whose pathogenic mechanism cannot be revealed from previously reported Nav structures.
Collapse
|
32
|
Structural basis for high-voltage activation and subtype-specific inhibition of human Na v1.8. Proc Natl Acad Sci U S A 2022; 119:e2208211119. [PMID: 35858452 PMCID: PMC9335304 DOI: 10.1073/pnas.2208211119] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Pain management represents an unmet healthcare need in many countries. Nav1.8 represents a potential target for developing nonaddictive analgesics. Here we present the cryogenic electron microscopy (cryo-EM) structures of human Nav1.8 alone and bound to a selective pore blocker, A-803467. Unlike reported structures of eukaryotic Nav channels wherein the first voltage-sensing domain (VSDI) is well-resolved in one stable conformation, different conformations of VSDI are observed in the cryo-EM maps of Nav1.8. An extracellular interface between VSDI and the pore domain was identified to be a determinant for Nav1.8’s dependence on higher voltage for activation. A-803467 clenches S6IV within the central cavity. Unexpectedly, the channel selectivity for A-803467 is determined by nonligand coordinating residues through an allosteric mechanism. The dorsal root ganglia–localized voltage-gated sodium (Nav) channel Nav1.8 represents a promising target for developing next-generation analgesics. A prominent characteristic of Nav1.8 is the requirement of more depolarized membrane potential for activation. Here we present the cryogenic electron microscopy structures of human Nav1.8 alone and bound to a selective pore blocker, A-803467, at overall resolutions of 2.7 to 3.2 Å. The first voltage-sensing domain (VSDI) displays three different conformations. Structure-guided mutagenesis identified the extracellular interface between VSDI and the pore domain (PD) to be a determinant for the high-voltage dependence of activation. A-803467 was clearly resolved in the central cavity of the PD, clenching S6IV. Our structure-guided functional characterizations show that two nonligand binding residues, Thr397 on S6I and Gly1406 on S6III, allosterically modulate the channel’s sensitivity to A-803467. Comparison of available structures of human Nav channels suggests the extracellular loop region to be a potential site for developing subtype-specific pore-blocking biologics.
Collapse
|
33
|
Jiang D, Zhang J, Xia Z. Structural Advances in Voltage-Gated Sodium Channels. Front Pharmacol 2022; 13:908867. [PMID: 35721169 PMCID: PMC9204039 DOI: 10.3389/fphar.2022.908867] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 05/23/2022] [Indexed: 11/17/2022] Open
Abstract
Voltage-gated sodium (NaV) channels are responsible for the rapid rising-phase of action potentials in excitable cells. Over 1,000 mutations in NaV channels are associated with human diseases including epilepsy, periodic paralysis, arrhythmias and pain disorders. Natural toxins and clinically-used small-molecule drugs bind to NaV channels and modulate their functions. Recent advances from cryo-electron microscopy (cryo-EM) structures of NaV channels reveal invaluable insights into the architecture, activation, fast inactivation, electromechanical coupling, ligand modulation and pharmacology of eukaryotic NaV channels. These structural analyses not only demonstrate molecular mechanisms for NaV channel structure and function, but also provide atomic level templates for rational development of potential subtype-selective therapeutics. In this review, we summarize recent structural advances of eukaryotic NaV channels, highlighting the structural features of eukaryotic NaV channels as well as distinct modulation mechanisms by a wide range of modulators from natural toxins to synthetic small-molecules.
Collapse
Affiliation(s)
- Daohua Jiang
- Laboratory of Soft Matter Physics, Institute of Physics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- *Correspondence: Daohua Jiang,
| | - Jiangtao Zhang
- Laboratory of Soft Matter Physics, Institute of Physics, Chinese Academy of Sciences, Beijing, China
- Key Laboratory of Molecular Biophysics of MOE, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Zhanyi Xia
- Laboratory of Soft Matter Physics, Institute of Physics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
34
|
Körner J, Albani S, Sudha Bhagavath Eswaran V, Roehl AB, Rossetti G, Lampert A. Sodium Channels and Local Anesthetics-Old Friends With New Perspectives. Front Pharmacol 2022; 13:837088. [PMID: 35418860 PMCID: PMC8996304 DOI: 10.3389/fphar.2022.837088] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 02/16/2022] [Indexed: 12/11/2022] Open
Abstract
The long history of local anesthetics (LAs) starts out in the late 19th century when the content of coca plant leaves was discovered to alleviate pain. Soon after, cocaine was established and headed off to an infamous career as a substance causing addiction. Today, LAs and related substances-in modified form-are indispensable in our clinical everyday life for pain relief during and after minor and major surgery, and dental practices. In this review, we elucidate on the interaction of modern LAs with their main target, the voltage-gated sodium channel (Navs), in the light of the recently published channel structures. Knowledge of the 3D interaction sites of the drug with the protein will allow to mechanistically substantiate the comprehensive data available on LA gating modification. In the 1970s it was suggested that LAs can enter the channel pore from the lipid phase, which was quite prospective at that time. Today we know from cryo-electron microscopy structures and mutagenesis experiments, that indeed Navs have side fenestrations facing the membrane, which are likely the entrance for LAs to induce tonic block. In this review, we will focus on the effects of LA binding on fast inactivation and use-dependent inhibition in the light of the proposed new allosteric mechanism of fast inactivation. We will elaborate on subtype and species specificity and provide insights into modelling approaches that will help identify the exact molecular binding orientation, access pathways and pharmacokinetics. With this comprehensive overview, we will provide new perspectives in the use of the drug, both clinically and as a tool for basic ion channel research.
Collapse
Affiliation(s)
- Jannis Körner
- Institute of Physiology, Aachen, Germany.,Clinic of Anesthesiology, Medical Faculty, Uniklinik RWTH Aachen, Aachen, Germany
| | - Simone Albani
- Institute for Neuroscience and Medicine (INM-9/IAS-5), Forschungszentrum Jülich, Jülich, Germany.,Faculty of Mathematics, Computer Science and Natural Sciences, RWTH Aachen, Aachen, Germany
| | | | - Anna B Roehl
- Clinic of Anesthesiology, Medical Faculty, Uniklinik RWTH Aachen, Aachen, Germany
| | - Giulia Rossetti
- Institute for Neuroscience and Medicine (INM-9/IAS-5), Forschungszentrum Jülich, Jülich, Germany.,Jülich Supercomputing Center (JSC), Forschungszentrum Jülich, Aachen, Germany.,Department of Neurology, University Hospital Aachen, RWTH Aachen University, Aachen, Germany
| | | |
Collapse
|
35
|
Guardiani C, Cecconi F, Chiodo L, Cottone G, Malgaretti P, Maragliano L, Barabash ML, Camisasca G, Ceccarelli M, Corry B, Roth R, Giacomello A, Roux B. Computational methods and theory for ion channel research. ADVANCES IN PHYSICS: X 2022; 7:2080587. [PMID: 35874965 PMCID: PMC9302924 DOI: 10.1080/23746149.2022.2080587] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 05/15/2022] [Indexed: 06/15/2023] Open
Abstract
Ion channels are fundamental biological devices that act as gates in order to ensure selective ion transport across cellular membranes; their operation constitutes the molecular mechanism through which basic biological functions, such as nerve signal transmission and muscle contraction, are carried out. Here, we review recent results in the field of computational research on ion channels, covering theoretical advances, state-of-the-art simulation approaches, and frontline modeling techniques. We also report on few selected applications of continuum and atomistic methods to characterize the mechanisms of permeation, selectivity, and gating in biological and model channels.
Collapse
Affiliation(s)
- C. Guardiani
- Dipartimento di Ingegneria Meccanica e Aerospaziale, Sapienza Università di Roma, Rome, Italy
| | - F. Cecconi
- CNR - Istituto dei Sistemi Complessi, Rome, Italy and Istituto Nazionale di Fisica Nucleare, INFN, Roma1 section. 00185, Roma, Italy
| | - L. Chiodo
- Department of Engineering, Campus Bio-Medico University, Rome, Italy
| | - G. Cottone
- Department of Physics and Chemistry-Emilio Segrè, University of Palermo, Palermo, Italy
| | - P. Malgaretti
- Helmholtz Institute Erlangen-Nürnberg for Renewable Energy (IEK-11), Forschungszentrum Jülich, Erlangen, Germany
| | - L. Maragliano
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Ancona, Italy, and Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova, Italy
| | - M. L. Barabash
- Department of Materials Science and Nanoengineering, Rice University, Houston, TX 77005, USA
| | - G. Camisasca
- Dipartimento di Ingegneria Meccanica e Aerospaziale, Sapienza Università di Roma, Rome, Italy
- Dipartimento di Fisica, Università Roma Tre, Rome, Italy
| | - M. Ceccarelli
- Department of Physics and CNR-IOM, University of Cagliari, Monserrato 09042-IT, Italy
| | - B. Corry
- Research School of Biology, The Australian National University, Canberra, ACT 2600, Australia
| | - R. Roth
- Institut Für Theoretische Physik, Eberhard Karls Universität Tübingen, Tübingen, Germany
| | - A. Giacomello
- Dipartimento di Ingegneria Meccanica e Aerospaziale, Sapienza Università di Roma, Rome, Italy
| | - B. Roux
- Department of Biochemistry & Molecular Biology, University of Chicago, Chicago IL, USA
| |
Collapse
|
36
|
Cheng J, Wei W, Fang Y, Zhou N, Wu Q, Zhao Q. Sudden cardiac death and cardiac sodium channel diseases. JOURNAL OF FORENSIC SCIENCE AND MEDICINE 2022. [DOI: 10.4103/jfsm.jfsm_123_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
|
37
|
Angsutararux P, Zhu W, Voelker TL, Silva JR. Molecular Pathology of Sodium Channel Beta-Subunit Variants. Front Pharmacol 2021; 12:761275. [PMID: 34867379 PMCID: PMC8640220 DOI: 10.3389/fphar.2021.761275] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 10/28/2021] [Indexed: 12/19/2022] Open
Abstract
The voltage-gated Na+ channel regulates the initiation and propagation of the action potential in excitable cells. The major cardiac isoform NaV1.5, encoded by SCN5A, comprises a monomer with four homologous repeats (I-IV) that each contain a voltage sensing domain (VSD) and pore domain. In native myocytes, NaV1.5 forms a macromolecular complex with NaVβ subunits and other regulatory proteins within the myocyte membrane to maintain normal cardiac function. Disturbance of the NaV complex may manifest as deadly cardiac arrhythmias. Although SCN5A has long been identified as a gene associated with familial atrial fibrillation (AF) and Brugada Syndrome (BrS), other genetic contributors remain poorly understood. Emerging evidence suggests that mutations in the non-covalently interacting NaVβ1 and NaVβ3 are linked to both AF and BrS. Here, we investigated the molecular pathologies of 8 variants in NaVβ1 and NaVβ3. Our results reveal that NaVβ1 and NaVβ3 variants contribute to AF and BrS disease phenotypes by modulating both NaV1.5 expression and gating properties. Most AF-linked variants in the NaVβ1 subunit do not alter the gating kinetics of the sodium channel, but rather modify the channel expression. In contrast, AF-related NaVβ3 variants directly affect channel gating, altering voltage-dependent activation and the time course of recovery from inactivation via the modulation of VSD activation.
Collapse
Affiliation(s)
- Paweorn Angsutararux
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, United States
| | - Wandi Zhu
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, United States.,Department of Medicine, Brigham and Women's Hospital, Boston, MA, United States
| | - Taylor L Voelker
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, United States
| | - Jonathan R Silva
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, United States
| |
Collapse
|
38
|
Zaytseva AK, Boitsov AS, Kostareva AA, Zhorov BS. Possible Interactions of Extracellular Loop IVP2-S6 With Voltage-Sensing Domain III in Cardiac Sodium Channel. Front Pharmacol 2021; 12:742508. [PMID: 34721031 PMCID: PMC8551724 DOI: 10.3389/fphar.2021.742508] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 09/13/2021] [Indexed: 11/30/2022] Open
Abstract
Motion transmission from voltage sensors to inactivation gates is an important problem in the general physiology of ion channels. In a cryo-EM structure of channel hNav1.5, residues N1736 and R1739 in the extracellular loop IVP2-S6 approach glutamates E1225 and E1295, respectively, in the voltage-sensing domain III (VSD-III). ClinVar-reported variants E1230K, E1295K, and R1739W/Q and other variants in loops IVP2-S6, IIIS1-S2, and IIIS3-S4 are associated with cardiac arrhythmias, highlighting the interface between IVP2-S6 and VSD-III as a hot spot of disease mutations. Atomic mechanisms of the channel dysfunction caused by these mutations are unknown. Here, we generated mutants E1295R, R1739E, E1295R/R1739E, and N1736R, expressed them in HEK-293T cells, and explored biophysical properties. Mutation E1295R reduced steady-state fast inactivation and enhanced steady-state slow inactivation. In contrast, mutation R1739E slightly enhanced fast inactivation and attenuated slow inactivation. Characteristics of the double mutant E1295R/R1739E were rather similar to those of the wild-type channel. Mutation N1736R attenuated slow inactivation. Molecular modeling predicted salt bridging of R1739E with the outermost lysine in the activated voltage-sensing helix IIIS4. In contrast, the loss-of-function substitution E1295R repelled R1739, thus destabilizing the activated VSD-III in agreement with our data that E1295R caused a depolarizing shift of the G-V curve. In silico deactivation of VSD-III with constraint-maintained salt bridge E1295-R1739 resulted in the following changes: 1) contacts between IIIS4 and IVS5 were switched; 2) contacts of the linker-helix IIIS4-S5 with IVS5, IVS6, and fast inactivation tripeptide IFM were modified; 3) contacts of the IFM tripeptide with helices IVS5 and IVS6 were altered; 4) mobile loop IVP2-S6 shifted helix IVP2 that contributes to the slow inactivation gate and helix IVS6 that contributes to the fast inactivation gate. The likelihood of salt bridge E1295-R1739 in deactivated VSD-III is supported by Poisson–Boltzmann calculations and state-dependent energetics of loop IVP2-S6. Taken together, our results suggest that loop IVP2-S6 is involved in motion transmission from VSD-III to the inactivation gates.
Collapse
Affiliation(s)
- Anastasia K Zaytseva
- Almazov National Medical Research Centre, St. Petersburg, Russia.,Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St. Petersburg, Russia
| | | | - Anna A Kostareva
- Almazov National Medical Research Centre, St. Petersburg, Russia.,Department of Women's and Children's Health, Karolinska Institute, Stockholm, Sweden
| | - Boris S Zhorov
- Almazov National Medical Research Centre, St. Petersburg, Russia.,Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St. Petersburg, Russia.,Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
39
|
Ca2+-dependent modulation of voltage-gated myocyte sodium channels. Biochem Soc Trans 2021; 49:1941-1961. [PMID: 34643236 PMCID: PMC8589445 DOI: 10.1042/bst20200604] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 08/01/2021] [Accepted: 08/31/2021] [Indexed: 12/19/2022]
Abstract
Voltage-dependent Na+ channel activation underlies action potential generation fundamental to cellular excitability. In skeletal and cardiac muscle this triggers contraction via ryanodine-receptor (RyR)-mediated sarcoplasmic reticular (SR) Ca2+ release. We here review potential feedback actions of intracellular [Ca2+] ([Ca2+]i) on Na+ channel activity, surveying their structural, genetic and cellular and functional implications, translating these to their possible clinical importance. In addition to phosphorylation sites, both Nav1.4 and Nav1.5 possess potentially regulatory binding sites for Ca2+ and/or the Ca2+-sensor calmodulin in their inactivating III–IV linker and C-terminal domains (CTD), where mutations are associated with a range of skeletal and cardiac muscle diseases. We summarize in vitro cell-attached patch clamp studies reporting correspondingly diverse, direct and indirect, Ca2+ effects upon maximal Nav1.4 and Nav1.5 currents (Imax) and their half-maximal voltages (V1/2) characterizing channel gating, in cellular expression systems and isolated myocytes. Interventions increasing cytoplasmic [Ca2+]i down-regulated Imax leaving V1/2 constant in native loose patch clamped, wild-type murine skeletal and cardiac myocytes. They correspondingly reduced action potential upstroke rates and conduction velocities, causing pro-arrhythmic effects in intact perfused hearts. Genetically modified murine RyR2-P2328S hearts modelling catecholaminergic polymorphic ventricular tachycardia (CPVT), recapitulated clinical ventricular and atrial pro-arrhythmic phenotypes following catecholaminergic challenge. These accompanied reductions in action potential conduction velocities. The latter were reversed by flecainide at RyR-blocking concentrations specifically in RyR2-P2328S as opposed to wild-type hearts, suggesting a basis for its recent therapeutic application in CPVT. We finally explore the relevance of these mechanisms in further genetic paradigms for commoner metabolic and structural cardiac disease.
Collapse
|
40
|
Wu X, Hong L. Calmodulin Interactions with Voltage-Gated Sodium Channels. Int J Mol Sci 2021; 22:ijms22189798. [PMID: 34575961 PMCID: PMC8472079 DOI: 10.3390/ijms22189798] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Revised: 09/06/2021] [Accepted: 09/07/2021] [Indexed: 02/06/2023] Open
Abstract
Calmodulin (CaM) is a small protein that acts as a ubiquitous signal transducer and regulates neuronal plasticity, muscle contraction, and immune response. It interacts with ion channels and plays regulatory roles in cellular electrophysiology. CaM modulates the voltage-gated sodium channel gating process, alters sodium current density, and regulates sodium channel protein trafficking and expression. Many mutations in the CaM-binding IQ domain give rise to diseases including epilepsy, autism, and arrhythmias by interfering with CaM interaction with the channel. In the present review, we discuss CaM interactions with the voltage-gated sodium channel and modulators involved in CaM regulation, as well as summarize CaM-binding IQ domain mutations associated with human diseases in the voltage-gated sodium channel family.
Collapse
|
41
|
Angsutararux P, Kang PW, Zhu W, Silva JR. Conformations of voltage-sensing domain III differentially define NaV channel closed- and open-state inactivation. J Gen Physiol 2021; 153:212533. [PMID: 34347027 PMCID: PMC8348240 DOI: 10.1085/jgp.202112891] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 07/14/2021] [Indexed: 11/21/2022] Open
Abstract
Voltage-gated Na+ (NaV) channels underlie the initiation and propagation of action potentials (APs). Rapid inactivation after NaV channel opening, known as open-state inactivation, plays a critical role in limiting the AP duration. However, NaV channel inactivation can also occur before opening, namely closed-state inactivation, to tune the cellular excitability. The voltage-sensing domain (VSD) within repeat IV (VSD-IV) of the pseudotetrameric NaV channel α-subunit is known to be a critical regulator of NaV channel inactivation. Yet, the two processes of open- and closed-state inactivation predominate at different voltage ranges and feature distinct kinetics. How inactivation occurs over these different ranges to give rise to the complexity of NaV channel dynamics is unclear. Past functional studies and recent cryo-electron microscopy structures, however, reveal significant inactivation regulation from other NaV channel components. In this Hypothesis paper, we propose that the VSD of NaV repeat III (VSD-III), together with VSD-IV, orchestrates the inactivation-state occupancy of NaV channels by modulating the affinity of the intracellular binding site of the IFMT motif on the III-IV linker. We review and outline substantial evidence that VSD-III activates in two distinct steps, with the intermediate and fully activated conformation regulating closed- and open-state inactivation state occupancy by altering the formation and affinity of the IFMT crevice. A role of VSD-III in determining inactivation-state occupancy and recovery from inactivation suggests a regulatory mechanism for the state-dependent block by small-molecule anti-arrhythmic and anesthetic therapies.
Collapse
Affiliation(s)
- Paweorn Angsutararux
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO
| | - Po Wei Kang
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO
| | - Wandi Zhu
- Department of Medicine, Brigham and Women's Hospital, Boston, MA
| | - Jonathan R Silva
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO
| |
Collapse
|
42
|
Functional cross-talk between phosphorylation and disease-causing mutations in the cardiac sodium channel Na v1.5. Proc Natl Acad Sci U S A 2021; 118:2025320118. [PMID: 34373326 PMCID: PMC8379932 DOI: 10.1073/pnas.2025320118] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
The cardiac sodium channel (Nav1.5) is crucial for generating a regular heartbeat. It is thus not surprising that Nav1.5 mutations have been linked to life-threatening arrhythmias. Interestingly, Nav1.5 activity can also be altered by posttranslational modifications, such as tyrosine phosphorylation. Our combination of protein engineering and molecular modeling has revealed that the detrimental effect of a long QT3 patient mutation is only exposed when a proximal tyrosine is phosphorylated. This suggests a dynamic cross-talk between the genetic mutation and a neighboring phosphorylation, a phenomenon that could be important in other classes of proteins. Additionally, we show that phosphorylation can affect the channel’s sensitivity toward clinically relevant drugs, a finding that may prove important when devising patient-specific treatment plans. The voltage-gated sodium channel Nav1.5 initiates the cardiac action potential. Alterations of its activation and inactivation properties due to mutations can cause severe, life-threatening arrhythmias. Yet despite intensive research efforts, many functional aspects of this cardiac channel remain poorly understood. For instance, Nav1.5 undergoes extensive posttranslational modification in vivo, but the functional significance of these modifications is largely unexplored, especially under pathological conditions. This is because most conventional approaches are unable to insert metabolically stable posttranslational modification mimics, thus preventing a precise elucidation of the contribution by these modifications to channel function. Here, we overcome this limitation by using protein semisynthesis of Nav1.5 in live cells and carry out complementary molecular dynamics simulations. We introduce metabolically stable phosphorylation mimics on both wild-type (WT) and two pathogenic long-QT mutant channel backgrounds and decipher functional and pharmacological effects with unique precision. We elucidate the mechanism by which phosphorylation of Y1495 impairs steady-state inactivation in WT Nav1.5. Surprisingly, we find that while the Q1476R patient mutation does not affect inactivation on its own, it enhances the impairment of steady-state inactivation caused by phosphorylation of Y1495 through enhanced unbinding of the inactivation particle. We also show that both phosphorylation and patient mutations can impact Nav1.5 sensitivity toward the clinically used antiarrhythmic drugs quinidine and ranolazine, but not flecainide. The data highlight that functional effects of Nav1.5 phosphorylation can be dramatically amplified by patient mutations. Our work is thus likely to have implications for the interpretation of mutational phenotypes and the design of future drug regimens.
Collapse
|
43
|
Structure of human Ca v2.2 channel blocked by the painkiller ziconotide. Nature 2021; 596:143-147. [PMID: 34234349 PMCID: PMC8529174 DOI: 10.1038/s41586-021-03699-6] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 06/07/2021] [Indexed: 02/06/2023]
Abstract
The neuronal-type (N-type) voltage-gated calcium (Cav) channels, which are designated Cav2.2, have an important role in the release of neurotransmitters1-3. Ziconotide is a Cav2.2-specific peptide pore blocker that has been clinically used for treating intractable pain4-6. Here we present cryo-electron microscopy structures of human Cav2.2 (comprising the core α1 and the ancillary α2δ-1 and β3 subunits) in the presence or absence of ziconotide. Ziconotide is thoroughly coordinated by helices P1 and P2, which support the selectivity filter, and the extracellular loops (ECLs) in repeats II, III and IV of α1. To accommodate ziconotide, the ECL of repeat III and α2δ-1 have to tilt upward concertedly. Three of the voltage-sensing domains (VSDs) are in a depolarized state, whereas the VSD of repeat II exhibits a down conformation that is stabilized by Cav2-unique intracellular segments and a phosphatidylinositol 4,5-bisphosphate molecule. Our studies reveal the molecular basis for Cav2.2-specific pore blocking by ziconotide and establish the framework for investigating electromechanical coupling in Cav channels.
Collapse
|