1
|
Nguyen PT, Harris BJ, Mateos DL, González AH, Murray AM, Yarov-Yarovoy V. Structural modeling of ion channels using AlphaFold2, RoseTTAFold2, and ESMFold. Channels (Austin) 2024; 18:2325032. [PMID: 38445990 PMCID: PMC10936637 DOI: 10.1080/19336950.2024.2325032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 01/14/2024] [Indexed: 03/07/2024] Open
Abstract
Ion channels play key roles in human physiology and are important targets in drug discovery. The atomic-scale structures of ion channels provide invaluable insights into a fundamental understanding of the molecular mechanisms of channel gating and modulation. Recent breakthroughs in deep learning-based computational methods, such as AlphaFold, RoseTTAFold, and ESMFold have transformed research in protein structure prediction and design. We review the application of AlphaFold, RoseTTAFold, and ESMFold to structural modeling of ion channels using representative voltage-gated ion channels, including human voltage-gated sodium (NaV) channel - NaV1.8, human voltage-gated calcium (CaV) channel - CaV1.1, and human voltage-gated potassium (KV) channel - KV1.3. We compared AlphaFold, RoseTTAFold, and ESMFold structural models of NaV1.8, CaV1.1, and KV1.3 with corresponding cryo-EM structures to assess details of their similarities and differences. Our findings shed light on the strengths and limitations of the current state-of-the-art deep learning-based computational methods for modeling ion channel structures, offering valuable insights to guide their future applications for ion channel research.
Collapse
Affiliation(s)
- Phuong Tran Nguyen
- Department of Physiology and Membrane Biology, University of California School of Medicine, Davis, CA, USA
| | - Brandon John Harris
- Department of Physiology and Membrane Biology, University of California School of Medicine, Davis, CA, USA
- Biophysics Graduate Group, University of California School of Medicine, Davis, CA, USA
| | - Diego Lopez Mateos
- Department of Physiology and Membrane Biology, University of California School of Medicine, Davis, CA, USA
- Biophysics Graduate Group, University of California School of Medicine, Davis, CA, USA
| | - Adriana Hernández González
- Department of Physiology and Membrane Biology, University of California School of Medicine, Davis, CA, USA
- Biophysics Graduate Group, University of California School of Medicine, Davis, CA, USA
| | | | - Vladimir Yarov-Yarovoy
- Department of Physiology and Membrane Biology, University of California School of Medicine, Davis, CA, USA
- Department of Anesthesiology and Pain Medicine, University of California School of Medicine, Davis, CA, USA
| |
Collapse
|
2
|
Rusina E, Simonti M, Duprat F, Cestèle S, Mantegazza M. Voltage-gated sodium channels in genetic epilepsy: up and down of excitability. J Neurochem 2024; 168:3872-3890. [PMID: 37654020 PMCID: PMC11591406 DOI: 10.1111/jnc.15947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 08/11/2023] [Accepted: 08/13/2023] [Indexed: 09/02/2023]
Abstract
The past two decades have witnessed a wide range of studies investigating genetic variants of voltage-gated sodium (NaV) channels, which are involved in a broad spectrum of diseases, including several types of epilepsy. We have reviewed here phenotypes and pathological mechanisms of genetic epilepsies caused by variants in NaV α and β subunits, as well as of some relevant interacting proteins (FGF12/FHF1, PRRT2, and Ankyrin-G). Notably, variants of all these genes can induce either gain- or loss-of-function of NaV leading to either neuronal hyperexcitability or hypoexcitability. We present the results of functional studies obtained with different experimental models, highlighting that they should be interpreted considering the features of the experimental system used. These systems are models, but they have allowed us to better understand pathophysiological issues, ameliorate diagnostics, orientate genetic counseling, and select/develop therapies within a precision medicine framework. These studies have also allowed us to gain insights into the physiological roles of different NaV channels and of the cells that express them. Overall, our review shows the progress that has been made, but also the need for further studies on aspects that have not yet been clarified. Finally, we conclude by highlighting some significant themes of general interest that can be gleaned from the results of the work of the last two decades.
Collapse
Affiliation(s)
- Evgeniia Rusina
- University Cote d'AzurValbonne‐Sophia AntipolisFrance
- CNRS UMR 7275Institute of Molecular and Cellular Pharmacology (IPMC)Valbonne‐Sophia AntipolisFrance
| | - Martina Simonti
- University Cote d'AzurValbonne‐Sophia AntipolisFrance
- CNRS UMR 7275Institute of Molecular and Cellular Pharmacology (IPMC)Valbonne‐Sophia AntipolisFrance
| | - Fabrice Duprat
- University Cote d'AzurValbonne‐Sophia AntipolisFrance
- CNRS UMR 7275Institute of Molecular and Cellular Pharmacology (IPMC)Valbonne‐Sophia AntipolisFrance
- InsermValbonne‐Sophia AntipolisFrance
| | - Sandrine Cestèle
- University Cote d'AzurValbonne‐Sophia AntipolisFrance
- CNRS UMR 7275Institute of Molecular and Cellular Pharmacology (IPMC)Valbonne‐Sophia AntipolisFrance
| | - Massimo Mantegazza
- University Cote d'AzurValbonne‐Sophia AntipolisFrance
- CNRS UMR 7275Institute of Molecular and Cellular Pharmacology (IPMC)Valbonne‐Sophia AntipolisFrance
- InsermValbonne‐Sophia AntipolisFrance
| |
Collapse
|
3
|
Aluru N, Chapman DP, W Becker K, Van Mooy BAS, Karchner SI, Stegeman JJ, Hahn ME. Developmental exposure of zebrafish to saxitoxin causes altered expression of genes associated with axonal growth. Neurotoxicology 2024; 105:303-312. [PMID: 39571800 PMCID: PMC11645194 DOI: 10.1016/j.neuro.2024.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 09/19/2024] [Accepted: 11/14/2024] [Indexed: 11/24/2024]
Abstract
Saxitoxin (STX) is a potent neurotoxin naturally produced by dinoflagellates and cyanobacteria. STX inhibits voltage-gated sodium channels (VGSCs), affecting the propagation of action potentials. Consumption of seafood contaminated with STX is responsible for paralytic shellfish poisoning (PSP). Humans are among the species most sensitive to PSP; neurological symptoms of exposure range from tingling of the extremities to severe paralysis. The objective of this study was to determine the effects of STX exposure on developmental processes during early embryogenesis. This study was designed to test the hypothesis that early developmental exposure to STX would disrupt key processes, particularly those related to neural development. Zebrafish embryos were exposed to STX (24 or 48 pg) or vehicle (0.3 mM HCl) at 6 h post fertilization (hpf) via microinjection. There was no overt toxicity but starting at 36 hpf there was a temporary lack of pigmentation in STX-injected embryos, which resolved by 72 hpf. Using high performance liquid chromatography, we found that STX was retained in embryos up to 72 hpf in a dose-dependent manner. Temporal transcriptional profiling of embryos exposed to 48 pg STX per embryo revealed no differentially expressed genes (DEGs) at 24 hpf, but at 36 and 48 hpf, there were 3547 and 3356 DEGs, respectively. KEGG pathway analysis revealed significant enrichment of genes related to focal adhesion, adherens junction and regulation of actin cytoskeleton, suggesting that cell-cell and cell-extracellular matrix interactions were affected by STX. Genes affected are critical for axonal growth and the development of functional neural networks. We confirmed these findings by visualizing axonal defects in transgenic zebrafish with fluorescently labeled sensory neurons. In addition, our gene expression results suggest that STX exposure affects both canonical and noncanonical functions of VGSCs. Given the fundamental role of VGSCs in both physiology and development, these findings offer valuable insights into effects of exposure to neurotoxins.
Collapse
Affiliation(s)
- Neelakanteswar Aluru
- Biology Department and Center for Oceans and Human Health,Woods Hole Oceanographic Institution, Woods Hole, Massachusetts 02543, USA.
| | - Daniel P Chapman
- Biology Department and Center for Oceans and Human Health,Woods Hole Oceanographic Institution, Woods Hole, Massachusetts 02543, USA; Eckerd College, 4200 54th Ave S, St. Petersburg, FL 33711, USA; Georgetown University School of Medicine, 3970 Reservoir Road NW, Washington DC 20057, USA
| | - Kevin W Becker
- Department of Marine Chemistry and Geochemistry, Woods Hole Oceanographic Institution, Woods Hole, Massachusetts 02543, USA; GEOMAR Helmholtz Centre for Ocean Research Kiel, Wischhofstr. 1-3, Kiel 24148, Germany
| | - Benjamin A S Van Mooy
- Department of Marine Chemistry and Geochemistry, Woods Hole Oceanographic Institution, Woods Hole, Massachusetts 02543, USA
| | - Sibel I Karchner
- Biology Department and Center for Oceans and Human Health,Woods Hole Oceanographic Institution, Woods Hole, Massachusetts 02543, USA
| | - John J Stegeman
- Biology Department and Center for Oceans and Human Health,Woods Hole Oceanographic Institution, Woods Hole, Massachusetts 02543, USA
| | - Mark E Hahn
- Biology Department and Center for Oceans and Human Health,Woods Hole Oceanographic Institution, Woods Hole, Massachusetts 02543, USA
| |
Collapse
|
4
|
Isaac Guillén F, Geist MA, Cheng SY, Harris AM, Treviño ME, Brumback AC, Nishiyama H, Howard MA. A novel mouse model for developmental and epileptic encephalopathy by Purkinje cell-specific deletion of Scn1b. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.19.624370. [PMID: 39605540 PMCID: PMC11601654 DOI: 10.1101/2024.11.19.624370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Loss of function variants of SCN1B are associated with a range of developmental and epileptic encephalopathies (DEEs), including Dravet syndrome. These DEEs feature a wide range of severe neurological disabilities, including changes to social, motor, mood, sleep, and cognitive function which are notoriously difficult to treat, and high rates of early mortality. While the symptomology of SCN1B -associated DEEs indicates broad changes in neural function, most research has focused on epilepsy-related brain structures and function. Mechanistic studies of SCN1B / Scn1b have delineated diverse roles in development and adult maintenance of neural function, via cell adhesion, ion channel regulation, and other intra- and extra-cellular actions. However, use of mouse models is limited as knockout of Scn1b , globally and even in some cell-specific models (e.g., Parvalbumin+ interneuron-specific knockout) in adult mice, leads to severe and progressive epilepsy, health deterioration, and 100% mortality within weeks. Here, we report findings of a novel transgenic mouse line in which Scn1b was specifically deleted in cerebellar Purkinje cells. Unlike most existing models, these mice did not show failure to thrive or early mortality. However, we quantified marked decrements to Purkinje cell physiology as well as motor, social, and cognitive dysfunction. Our data indicate that cerebellar Purkinje cells are an important node for dysfunction and neural disabilities in SCN1B -related DEEs, and combined with previous work identify this as a potentially vital site for understanding mechanisms of DEEs and developing therapies that can treat these disorders holistically.
Collapse
|
5
|
Koster AK, Yarishkin O, Dubin AE, Kefauver JM, Pak RA, Cravatt BF, Patapoutian A. Chemical mapping of the surface interactome of PIEZO1 identifies CADM1 as a modulator of channel inactivation. Proc Natl Acad Sci U S A 2024; 121:e2415934121. [PMID: 39356664 PMCID: PMC11474052 DOI: 10.1073/pnas.2415934121] [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: 08/07/2024] [Accepted: 09/03/2024] [Indexed: 10/04/2024] Open
Abstract
The propeller-shaped blades of the PIEZO1 and PIEZO2 ion channels partition into the plasma membrane and respond to indentation or stretching of the lipid bilayer, thus converting mechanical forces into signals that can be interpreted by cells, in the form of calcium flux and changes in membrane potential. While PIEZO channels participate in diverse physiological processes, from sensing the shear stress of blood flow in the vasculature to detecting touch through mechanoreceptors in the skin, the molecular details that enable these mechanosensors to tune their responses over a vast dynamic range of forces remain largely uncharacterized. To survey the molecular landscape surrounding PIEZO channels at the cell surface, we employed a mass spectrometry-based proteomic approach to capture and identify extracellularly exposed proteins in the vicinity of PIEZO1. This PIEZO1-proximal interactome was enriched in surface proteins localized to cell junctions and signaling hubs within the plasma membrane. Functional screening of these interaction candidates by calcium imaging and electrophysiology in an overexpression system identified the adhesion molecule CADM1/SynCAM that slows the inactivation kinetics of PIEZO1 with little effect on PIEZO2. Conversely, we found that CADM1 knockdown accelerates inactivation of endogenous PIEZO1 in Neuro-2a cells. Systematic deletion of CADM1 domains indicates that the transmembrane region is critical for the observed effects on PIEZO1, suggesting that modulation of inactivation is mediated by interactions in or near the lipid bilayer.
Collapse
Affiliation(s)
- Anna K. Koster
- HHMI, Scripps Research, La JollaCA92037
- Department of Neuroscience, Scripps Research, La Jolla, CA92037
- Department of Chemistry, Scripps Research, La Jolla, CA92037
| | - Oleg Yarishkin
- HHMI, Scripps Research, La JollaCA92037
- Department of Neuroscience, Scripps Research, La Jolla, CA92037
| | - Adrienne E. Dubin
- HHMI, Scripps Research, La JollaCA92037
- Department of Neuroscience, Scripps Research, La Jolla, CA92037
| | - Jennifer M. Kefauver
- HHMI, Scripps Research, La JollaCA92037
- Department of Neuroscience, Scripps Research, La Jolla, CA92037
| | - Ryan A. Pak
- HHMI, Scripps Research, La JollaCA92037
- Department of Neuroscience, Scripps Research, La Jolla, CA92037
| | | | - Ardem Patapoutian
- HHMI, Scripps Research, La JollaCA92037
- Department of Neuroscience, Scripps Research, La Jolla, CA92037
| |
Collapse
|
6
|
Aldridge JL, Alexander ED, Franklin AA, Frasier CR. Decreased ability to manage increases in reactive oxygen species may underlie susceptibility to arrhythmias in mice lacking Scn1b. Am J Physiol Heart Circ Physiol 2024; 327:H723-H732. [PMID: 39120465 PMCID: PMC11482272 DOI: 10.1152/ajpheart.00265.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 07/31/2024] [Accepted: 08/05/2024] [Indexed: 08/10/2024]
Abstract
Scn1b plays essential roles in the heart, where it encodes β1-subunits that serve as modifiers of gene expression, cell surface channel activity, and cardiac conductivity. Reduced β1 function is linked to electrical instability in various diseases with cardiac manifestations and increased susceptibility to arrhythmias. Recently, we demonstrated that loss of Scn1b in mice leads to compromised mitochondria energetics and reactive oxygen species (ROS) production. In this study, we examined the link between increased ROS and arrhythmia susceptibility in Scn1b-/- mice. In addition, ROS-scavenging capacity can be overwhelmed during prolonged oxidative stress, increasing arrhythmia susceptibility. Therefore, we isolated whole hearts and cardiomyocytes from Scn1b-/- and Scn1b+/+ mice and subjected them to an oxidative challenge with diamide, a glutathione oxidant. Next, we analyzed gene expression and activity of antioxidant enzymes in Scn1b-/- hearts. Cells isolated from Scn1b-/- hearts died faster and displayed higher rates of ROS accumulation preceding cell death compared with those from Scn1b+/+. Furthermore, Scn1b-/- hearts showed higher arrhythmia scores and spent less time free of arrhythmia. Lastly, we found that protein expression and enzymatic activity of glutathione peroxidase is increased in Scn1b-/- hearts compared with wild type. Our results indicate that Scn1b-/- mice have decreased capability to manage ROS during prolonged oxidative stress. ROS accumulation is elevated and appears to overwhelm ROS scavenging through the glutathione system. This imbalance creates the potential for altered cell energetics that may underlie increased susceptibility to arrhythmias or other adverse cardiac outcomes.NEW & NOTEWORTHY Using an oxidative challenge, we demonstrated that isolated cells from Scn1b-/- mice are more susceptible to cell death and surges in reactive oxygen species accumulation. At the whole organ level, they were also more susceptible to the formation of cardiac arrhythmias. This may in part be due to changes to the glutathione antioxidant system.
Collapse
Affiliation(s)
- Jessa L Aldridge
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee, United States
| | - Emily Davis Alexander
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee, United States
| | - Allison A Franklin
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee, United States
| | - Chad R Frasier
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee, United States
| |
Collapse
|
7
|
Cortada E, Brugada R, Verges M. Role of protein domains in trafficking and localization of the voltage-gated sodium channel β2 subunit. J Biol Chem 2024; 300:107833. [PMID: 39343005 PMCID: PMC11532958 DOI: 10.1016/j.jbc.2024.107833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 08/25/2024] [Accepted: 09/18/2024] [Indexed: 10/01/2024] Open
Abstract
The voltage-gated sodium (NaV) channel is critical for cardiomyocyte function since it is responsible for action potential initiation and its propagation throughout the cell. It consists of a protein complex made of a pore forming α subunit and associated β subunits, which regulate α subunit function and subcellular localization. We previously showed the implication of N-linked glycosylation and S-acylation of β2 in its polarized trafficking. Here, we present evidence of β2 dimerization. Moreover, we demonstrate the implication of the cytoplasmic tail, extracellular loop, and transmembrane domain on proper β2 folding and export to the cell surface of polarized Madin-Darby canine kidney cells. Substantial alteration, or lack of any of these domains, leads to accumulation of β2 in the endoplasmic reticulum, along with impaired complex N-glycosylation, which is needed for its efficient surface delivery. We also show that these alterations to β2 affected to a certain extent NaV1.5 surface localization. Conversely, however, NaV1.5 had little or no influence on β2 trafficking, its localization to the surface, or homodimer formation. Altogether, our data link the architecture of the β2 domains to the establishment of its proper subcellular localization. These findings could provide valuable insights to gain a deeper comprehension of the elusive biology of β subunits in excitable cells, such as neurons and cardiomyocytes.
Collapse
Affiliation(s)
- Eric Cortada
- Cardiovascular Genetics Group, Girona Biomedical Research Institute (IDIBGI-CERCA), Edifici IDIBGI, Salt, Province of Girona, Spain; Biomedical Research Networking Center on Cardiovascular Diseases (CIBERCV), Salt, Province of Girona, Spain
| | - Ramon Brugada
- Cardiovascular Genetics Group, Girona Biomedical Research Institute (IDIBGI-CERCA), Edifici IDIBGI, Salt, Province of Girona, Spain; Biomedical Research Networking Center on Cardiovascular Diseases (CIBERCV), Salt, Province of Girona, Spain; Department of Medical Sciences, University of Girona Medical School, Girona, Spain; Department of Cardiology, Hospital Josep Trueta, University of Girona, Girona, Spain
| | - Marcel Verges
- Cardiovascular Genetics Group, Girona Biomedical Research Institute (IDIBGI-CERCA), Edifici IDIBGI, Salt, Province of Girona, Spain; Biomedical Research Networking Center on Cardiovascular Diseases (CIBERCV), Salt, Province of Girona, Spain; Department of Medical Sciences, University of Girona Medical School, Girona, Spain.
| |
Collapse
|
8
|
Wang X, Luo H, Peng X, Chen J. Spider and scorpion knottins targeting voltage-gated sodium ion channels in pain signaling. Biochem Pharmacol 2024; 227:116465. [PMID: 39102991 DOI: 10.1016/j.bcp.2024.116465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 07/30/2024] [Accepted: 08/02/2024] [Indexed: 08/07/2024]
Abstract
In sensory neurons that transmit pain signals, whether acute or chronic, voltage-gated sodium channels (VGSCs) are crucial for regulating excitability. NaV1.1, NaV1.3, NaV1.6, NaV1.7, NaV1.8, and NaV1.9 have been demonstrated and defined their functional roles in pain signaling based on their biophysical properties and distinct patterns of expression in each subtype of sensory neurons. Scorpions and spiders are traditional Chinese medicinal materials, belonging to the arachnid class. Most of the studied species of them have evolved venom peptides that exhibit a wide variety of knottins specifically targeting VGSCs with subtype selectivity and conformational specificity. This review provides an overview on the exquisite knottins from scorpion and spider venoms targeting pain-related NaV channels, describing the sequences and the structural features as well as molecular determinants that influence their selectivity on special subtype and at particular conformation, with an aim for the development of novel research tools on NaV channels and analgesics with minimal adverse effects.
Collapse
Affiliation(s)
- Xiting Wang
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, Hunan 410128, China
| | - Huan Luo
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, Hunan 410128, China
| | - Xiaozhen Peng
- School of Public Health & Laboratory Medicine, Hunan University of Medicine, Huaihua 418000, China.
| | - Jinjun Chen
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, Hunan 410128, China; Hunan Provincial Engineering Technology Research Center for Cell Mechanics and Function Analysis, Changsha 418000, China.
| |
Collapse
|
9
|
Catterall WA, Gamal El-Din TM, Wisedchaisri G. The chemistry of electrical signaling in sodium channels from bacteria and beyond. Cell Chem Biol 2024; 31:1405-1421. [PMID: 39151407 DOI: 10.1016/j.chembiol.2024.07.010] [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: 01/12/2024] [Revised: 06/27/2024] [Accepted: 07/22/2024] [Indexed: 08/19/2024]
Abstract
Electrical signaling is essential for all fast processes in biology, but its molecular mechanisms have been uncertain. This review article focuses on studies of bacterial sodium channels in order to home in on the essential molecular and chemical mechanisms underlying transmembrane ion conductance and voltage-dependent gating without the overlay of complex protein interactions and regulatory mechanisms in mammalian sodium channels. This minimalist approach has yielded a nearly complete picture of sodium channel function at the atomic level that are mostly conserved in mammalian sodium channels, including sodium selectivity and conductance, voltage sensing and activation, electromechanical coupling to pore opening and closing, slow inactivation, and pathogenic dysfunction in a debilitating channelopathy. Future studies of nature's simplest sodium channels may continue to yield key insights into the fundamental molecular and chemical principles of their function and further elucidate the chemical basis of electrical signaling.
Collapse
Affiliation(s)
- William A Catterall
- Department of Pharmacology, University of Washington, Seattle WA 98195-7280, USA.
| | - Tamer M Gamal El-Din
- Department of Pharmacology, University of Washington, Seattle WA 98195-7280, USA.
| | - Goragot Wisedchaisri
- Department of Pharmacology, University of Washington, Seattle WA 98195-7280, USA.
| |
Collapse
|
10
|
Moreira-Junior L, Leal-Cardoso JH, Cassola AC, Carvalho-de-Souza JL. Eugenol and lidocaine inhibit voltage-gated Na + channels from dorsal root ganglion neurons with different mechanisms. Front Pharmacol 2024; 15:1354737. [PMID: 38989141 PMCID: PMC11234063 DOI: 10.3389/fphar.2024.1354737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 04/08/2024] [Indexed: 07/12/2024] Open
Abstract
Eugenol (EUG) is a bioactive monoterpenoid used as an analgesic, preservative, and flavoring agent. Our new data show EUG as a voltage-gated Na+ channel (VGSC) inhibitor, comparable but not identical to lidocaine (LID). EUG inhibits both total and only TTX-R voltage-activated Na+ currents (INa) recorded from VGSCs naturally expressed on dorsal root ganglion sensory neurons in rats. Inhibition is quick, fully reversible, and dose-dependent. Our biophysical and pharmacological analyses showed that EUG and LID inhibit VGSCs with different mechanisms. EUG inhibits VGSCs with a dose-response relationship characterized by a Hill coefficient of 2, while this parameter for the inhibition by LID is 1. Furthermore, in a different way from LID, EUG modified the voltage dependence of both the VGSC activation and inactivation processes and the recovery from fast inactivated states and the entry to slow inactivated states. In addition, we suggest that EUG, but not LID, interacts with VGSC pre-open-closed states, according to our data.
Collapse
Affiliation(s)
- Luiz Moreira-Junior
- Department of Anesthesiology, University of Arizona, Tucson, AZ, United States
| | | | - Antonio Carlos Cassola
- Department of Physiology and Biophysics, Biomedical Sciences Institute, University of Sao Paulo, São Paulo, Brazil
| | - Joao Luis Carvalho-de-Souza
- Department of Anesthesiology, University of Arizona, Tucson, AZ, United States
- Department of Physiology and Biophysics, Biomedical Sciences Institute, University of Sao Paulo, São Paulo, Brazil
| |
Collapse
|
11
|
Williams ZJ, Payne LB, Wu X, Gourdie RG. New focus on cardiac voltage-gated sodium channel β1 and β1B: Novel targets for treating and understanding arrhythmias? Heart Rhythm 2024:S1547-5271(24)02742-5. [PMID: 38908461 DOI: 10.1016/j.hrthm.2024.06.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 06/09/2024] [Accepted: 06/17/2024] [Indexed: 06/24/2024]
Abstract
Voltage-gated sodium channels (VGSCs) are transmembrane protein complexes that are vital to the generation and propagation of action potentials in nerve and muscle fibers. The canonical VGSC is generally conceived as a heterotrimeric complex formed by 2 classes of membrane-spanning subunit: an α-subunit (pore forming) and 2 β-subunits (non-pore forming). NaV1.5 is the main sodium channel α-subunit of mammalian ventricle, with lower amounts of other α-subunits, including NaV1.6, being present. There are 4 β-subunits (β1-β4) encoded by 4 genes (SCN1B-SCN4B), each of which is expressed in cardiac tissues. Recent studies suggest that in addition to assignments in channel gating and trafficking, products of Scn1b may have novel roles in conduction of action potential in the heart and intracellular signaling. This includes evidence that the β-subunit extracellular amino-terminal domain facilitates adhesive interactions in intercalated discs and that its carboxyl-terminal region is a substrate for a regulated intramembrane proteolysis (RIP) signaling pathway, with a carboxyl-terminal peptide generated by β1 RIP trafficked to the nucleus and altering transcription of various genes, including NaV1.5. In addition to β1, the Scn1b gene encodes for an alternative splice variant, β1B, which contains an identical extracellular adhesion domain to β1 but has a unique carboxyl-terminus. Although β1B is generally understood to be a secreted variant, evidence indicates that when co-expressed with NaV1.5, it is maintained at the cell membrane, suggesting potential unique roles for this understudied protein. In this review, we focus on what is known of the 2 β-subunit variants encoded by Scn1b in heart, with particular focus on recent findings and the questions raised by this new information. We also explore data that indicate β1 and β1B may be attractive targets for novel antiarrhythmic therapeutics.
Collapse
Affiliation(s)
- Zachary J Williams
- Fralin Biomedical Research Institute, Virginia Polytechnic University, Roanoke, Virginia
| | - Laura Beth Payne
- Fralin Biomedical Research Institute, Virginia Polytechnic University, Roanoke, Virginia
| | - Xiaobo Wu
- Fralin Biomedical Research Institute, Virginia Polytechnic University, Roanoke, Virginia
| | - Robert G Gourdie
- Fralin Biomedical Research Institute, Virginia Polytechnic University, Roanoke, Virginia; School of Medicine, Virgina Polytechnic University, Roanoke, Virginia; Department of Biomedical Engineering and Mechanics, Virginia Polytechnic University, Blacksburg, Virginia.
| |
Collapse
|
12
|
Zhang J, Zou L, Tan F, Wang H, Wen Z, Wang H, Li L. Screening of co-expressed genes in hypopharyngeal carcinoma with esophageal carcinoma based on RNA sequencing and Clinical Research. Sci Rep 2024; 14:13796. [PMID: 38877096 PMCID: PMC11178892 DOI: 10.1038/s41598-024-64162-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 06/05/2024] [Indexed: 06/16/2024] Open
Abstract
To explore the hub comorbidity genes and potential pathogenic mechanisms of hypopharyngeal carcinoma with esophageal carcinoma, and evaluate their diagnostic value for hypopharyngeal carcinoma with co-morbid esophageal carcinoma. We performed gene sequencing on tumor tissues from 6 patients with hypopharyngeal squamous cell carcinoma with esophageal squamous cell carcinoma (hereafter referred to as "group A") and 6 patients with pure hypopharyngeal squamous cell carcinoma (hereafter referred to as "group B"). We analyzed the mechanism of hub genes in the development and progression of hypopharyngeal squamous cell carcinoma with esophageal squamous cell carcinoma through bioinformatics, and constructed an ROC curve and Nomogram prediction model to analyze the value of hub genes in clinical diagnosis and treatment. 44,876 genes were sequenced in 6 patients with group A and 6 patients with group B. Among them, 76 genes showed significant statistical differences between the group A and the group B.47 genes were expressed lower in the group A than in the group B, and 29 genes were expressed higher. The top five hub genes were GABRG2, CACNA1A, CNTNAP2, NOS1, and SCN4B. GABRG2, CNTNAP2, and SCN4B in the hub genes have high diagnostic value in determining whether hypopharyngeal carcinoma patients have combined esophageal carcinoma (AUC: 0.944, 0.944, 0.972). These genes could possibly be used as potential molecular markers for assessing the risk of co-morbidity of hypopharyngeal carcinoma combined with esophageal carcinoma.
Collapse
Affiliation(s)
- Jianing Zhang
- Department of Otorhinolaryngology Head and Neck Surgery, Central Hospital of Chaoyang, Liaoning, 122000, China
| | - Liangyu Zou
- Department of Otorhinolaryngology Head and Neck Surgery, Central Hospital of Chaoyang, Liaoning, 122000, China
| | - Fuxian Tan
- Department of Otorhinolaryngology Head and Neck Surgery, Central Hospital of Chaoyang, Liaoning, 122000, China
| | - Hongmin Wang
- Department of Otorhinolaryngology Head and Neck Surgery, Central Hospital of Chaoyang, Liaoning, 122000, China
| | - Zhenlei Wen
- Department of Otorhinolaryngology Head and Neck Surgery, Central Hospital of Chaoyang, Liaoning, 122000, China
| | - Hongmei Wang
- Department of Otorhinolaryngology Head and Neck Surgery, Central Hospital of Chaoyang, Liaoning, 122000, China
| | - Lianhe Li
- Department of Otorhinolaryngology Head and Neck Surgery, Central Hospital of Chaoyang, Liaoning, 122000, China.
| |
Collapse
|
13
|
Marchal GA, Rivaud MR, Wolswinkel R, Basso C, van Veen TAB, Bezzina CR, Remme CA. Genetic background determines the severity of age-dependent cardiac structural abnormalities and arrhythmia susceptibility in Scn5a-1798insD mice. Europace 2024; 26:euae153. [PMID: 38875491 PMCID: PMC11203918 DOI: 10.1093/europace/euae153] [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: 02/09/2024] [Accepted: 05/12/2024] [Indexed: 06/16/2024] Open
Abstract
AIMS Patients with mutations in SCN5A encoding NaV1.5 often display variable severity of electrical and structural alterations, but the underlying mechanisms are not fully elucidated. We here investigate the combined modulatory effect of genetic background and age on disease severity in the Scn5a1798insD/+ mouse model. METHODS AND RESULTS In vivo electrocardiogram and echocardiograms, ex vivo electrical and optical mapping, and histological analyses were performed in adult (2-7 months) and aged (8-28 months) wild-type (WT) and Scn5a1798insD/+ (mutant, MUT) mice from the FVB/N and 129P2 inbred strains. Atrio-ventricular (AV) conduction, ventricular conduction, and ventricular repolarization are modulated by strain, genotype, and age. An aging effect was present in MUT mice, with aged MUT mice of both strains showing prolonged QRS interval and right ventricular (RV) conduction slowing. 129P2-MUT mice were severely affected, with adult and aged 129P2-MUT mice displaying AV and ventricular conduction slowing, prolonged repolarization, and spontaneous arrhythmias. In addition, the 129P2 strain appeared particularly susceptible to age-dependent electrical, functional, and structural alterations including RV conduction slowing, reduced left ventricular (LV) ejection fraction, RV dilatation, and myocardial fibrosis as compared to FVB/N mice. Overall, aged 129P2-MUT mice displayed the most severe conduction defects, RV dilatation, and myocardial fibrosis, in addition to the highest frequency of spontaneous arrhythmia and inducible arrhythmias. CONCLUSION Genetic background and age both modulate disease severity in Scn5a1798insD/+ mice and hence may explain, at least in part, the variable disease expressivity observed in patients with SCN5A mutations. Age- and genetic background-dependent development of cardiac structural alterations furthermore impacts arrhythmia risk. Our findings therefore emphasize the importance of continued assessment of cardiac structure and function in patients carrying SCN5A mutations.
Collapse
Affiliation(s)
- Gerard A Marchal
- Department of Experimental Cardiology, Heart Centre, Amsterdam UMC location University of Amsterdam, Meibergdreef 15, PO Box 22660, 1100 DD Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Meibergdreef 15, PO Box 22660, 1100 DD Amsterdam, The Netherlands
- OptoCARD Lab, Institute of Clinical Physiology (IFC-CNR), Florence, Italy
| | - Mathilde R Rivaud
- Department of Experimental Cardiology, Heart Centre, Amsterdam UMC location University of Amsterdam, Meibergdreef 15, PO Box 22660, 1100 DD Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Meibergdreef 15, PO Box 22660, 1100 DD Amsterdam, The Netherlands
| | - Rianne Wolswinkel
- Department of Experimental Cardiology, Heart Centre, Amsterdam UMC location University of Amsterdam, Meibergdreef 15, PO Box 22660, 1100 DD Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Meibergdreef 15, PO Box 22660, 1100 DD Amsterdam, The Netherlands
| | - Cristina Basso
- Department of Cardiac, Thoracic and Vascular Sciences and Public Health, University of Padova, Padua, Italy
| | - Toon A B van Veen
- Department of Medical Physiology, Division Heart & Lungs, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Connie R Bezzina
- Department of Experimental Cardiology, Heart Centre, Amsterdam UMC location University of Amsterdam, Meibergdreef 15, PO Box 22660, 1100 DD Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Meibergdreef 15, PO Box 22660, 1100 DD Amsterdam, The Netherlands
| | - Carol Ann Remme
- Department of Experimental Cardiology, Heart Centre, Amsterdam UMC location University of Amsterdam, Meibergdreef 15, PO Box 22660, 1100 DD Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Meibergdreef 15, PO Box 22660, 1100 DD Amsterdam, The Netherlands
| |
Collapse
|
14
|
Yang X, Liu Q, Li G. Anti-NSCLC role of SCN4B by negative regulation of the cGMP-PKG pathway: Integrated utilization of bioinformatics analysis and in vitro assay validation. Drug Dev Res 2024; 85:e22192. [PMID: 38678552 DOI: 10.1002/ddr.22192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 04/04/2024] [Accepted: 04/12/2024] [Indexed: 05/01/2024]
Abstract
Non-small cell lung cancer (NSCLC) is a malignant tumor with low overall cure and survival rates. Uncovering abnormally expressed genes is significantly important for developing novel targeted therapies in NSCLC. This study aimed to discover new differentially expressed genes (DEGs) of NSCLC. The DEGs of NSCLC were identified in eight data sets from Gene Expression Omnibus (GEO) database. The expression profiles and the prognostic significance of SCN4B in LUAD and LUSC were analyzed using GEPIA database. LinkedOmics was used to identify co-expressed genes with SCN4B, which were further subjected to KEGG pathway enrichment analysis. SCN4B-overexpressing plasmid (pcDNA/SCN4B) was transfected into A549 and NCI-H2170 cells to elevate the expression of SCN4B. MTT and TUNEL assays were performed to evaluate cell viability and apoptosis. Relying on the screened DEGs from GEO database, we identified that SCN4B was significantly downregulated in LUAD and LUSC. We confirmed the downregulation of SCN4B in NSCLC tissues using GEPIA database. SCN4B has a prognostic value in LUAD, but not LUSC. KEGG pathway enrichment analysis of SCN4B-related genes showed that cGMP-PKG signaling pathway might be involved in the role of SCN4B in NSCLC. Overexpression of SCN4B in A549 and NCI-H2170 cells inhibited the cell viability. Besides, SCN4B overexpression induced apoptosis of A549 and NCI-H2170 cells. SCN4B inhibited the expression of PKG1 and p-CREB in NSCLC cells. Moreover, the inhibitory effects of SCN4B on tumor malignancy were attenuated by the activator of PKG. In conclusion, integrated bioinformatical analysis proved that SCN4B was downregulated and had a prognostic significance in NSCLC. In vitro experimental studies demonstrated that SCN4B regulated NSCLC cells viability and apoptosis via inhibiting cGMP-PKG signaling pathway.
Collapse
Affiliation(s)
- Xiujun Yang
- Department of Respiratory and Critical Care Medicine, Huai'an People's Hospital of Hongze District, Huai'an, China
| | - Qun Liu
- Medical Ward 20, Lianshui County People's Hospital, Huai'an, China
| | - Gang Li
- Department of Respiratory and Critical Care Medicine, Huai'an Second People's Hospital, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, China
| |
Collapse
|
15
|
Janes ME, Kinlein A, Flajnik MF, Du Pasquier L, Ohta Y. Genomic view of the origins of cell-mediated immunity. Immunogenetics 2023; 75:479-493. [PMID: 37735270 PMCID: PMC11019866 DOI: 10.1007/s00251-023-01319-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 08/10/2023] [Indexed: 09/23/2023]
Abstract
NKp30 is an activating natural killer cell receptor (NKR) with a single-exon variable (VJ)-type immunoglobulin superfamily (IgSF) domain. Such VJ-IgSF domains predate the emergence of the antigen receptors (immunoglobulin and T cell receptor), which possess the same domain but undergo gene rearrangement. NCR3, the gene encoding NKp30, is present in jawed vertebrates from sharks to mammals; thus, unlike most NKR that are highly divergent among vertebrate taxa, NKp30 is uniquely conserved. We previously hypothesized that an ancestral NCR3 gene was encoded in the proto-major histocompatibility complex (MHC), the region where many immune-related genes have accumulated. Herein, we searched in silico databases to identify NCR3 paralogues and examined their genomic locations. We found a paralogue, NCR3H, in many vertebrates but was lost in mammals. Additionally, we identified a set of voltage-gated sodium channel beta (SCNB) genes as NCR3-distantly-related genes. Like NCR3, both NCR3H and SCNB proteins contain a single VJ-IgSF domain followed by a transmembrane region. These genes map to MHC paralogous regions, originally described in an invertebrate, along with genes encoding cell adhesion molecules involved in NK cell recognition networks. Other genes having no obvious relationship to immunity also map to these paralogous regions. These gene complexes were traced to several invertebrates, suggesting that the foundation of these cellular networks emerged before the genome-wide duplications in early gnathostome history. Here, we propose that this ancestral region was involved in cell-mediated immunity prior to the emergence of adaptive immunity and that NCR3 piggybacked onto this primordial complex, heralding the emergence of vertebrate NK cell/T cells.
Collapse
Affiliation(s)
- Morgan E Janes
- Department of Microbiology and Immunology, University of Maryland, Baltimore, MD, 21201, USA
| | - Allison Kinlein
- Department of Microbiology and Immunology, University of Maryland, Baltimore, MD, 21201, USA
| | - Martin F Flajnik
- Department of Microbiology and Immunology, University of Maryland, Baltimore, MD, 21201, USA
| | - Louis Du Pasquier
- Department of Environmental Sciences, Zoology, University of Basel, Vesalgasse 1, 4051, Basel, Switzerland
| | - Yuko Ohta
- Department of Microbiology and Immunology, University of Maryland, Baltimore, MD, 21201, USA.
| |
Collapse
|
16
|
Spafford JD. A governance of ion selectivity based on the occupancy of the "beacon" in one- and four-domain calcium and sodium channels. Channels (Austin) 2023; 17:2191773. [PMID: 37075164 PMCID: PMC10120453 DOI: 10.1080/19336950.2023.2191773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/21/2023] Open
Abstract
One of nature's exceptions was discovered when a Cav3 T-type channel was observed to switch phenotype from a calcium channel into a sodium channel by neutralizing an aspartate residue in the high field strength (HFS) +1 position within the ion selectivity filter. The HFS+1 site is dubbed a "beacon" for its location at the entryway just above the constricted, minimum radius of the HFS site's electronegative ring. A classification is proposed based on the occupancy of the HFS+1 "beacon" which correlates with the calcium- or sodium-selectivity phenotype. If the beacon is a glycine, or neutral, non-glycine residue, then the cation channel is calcium-selective or sodium-permeable, respectively (Class I). Occupancy of a beacon aspartate are calcium-selective channels (Class II) or possessing a strong calcium block (Class III). A residue lacking in position of the sequence alignment for the beacon are sodium channels (Class IV). The extent to which animal channels are sodium-selective is dictated in the occupancy of the HFS site with a lysine residue (Class III/IV). Governance involving the beacon solves the quandary the HFS site as a basis for ion selectivity, where an electronegative ring of glutamates at the HFS site generates a sodium-selective channel in one-domain channels but generates a calcium-selective channel in four-domain channels. Discovery of a splice variant in an exceptional channel revealed nature's exploits, highlighting the "beacon" as a principal determinant for calcium and sodium selectivity, encompassing known ion channels composed of one and four domains, from bacteria to animals.
Collapse
Affiliation(s)
- J David Spafford
- Department of Biology, University of Waterloo, Waterloo, Ontario, Canada
| |
Collapse
|
17
|
Llanos MA, Enrique N, Esteban-López V, Scioli-Montoto S, Sánchez-Benito D, Ruiz ME, Milesi V, López DE, Talevi A, Martín P, Gavernet L. A Combined Ligand- and Structure-Based Virtual Screening To Identify Novel NaV1.2 Blockers: In Vitro Patch Clamp Validation and In Vivo Anticonvulsant Activity. J Chem Inf Model 2023; 63:7083-7096. [PMID: 37917937 DOI: 10.1021/acs.jcim.3c00645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2023]
Abstract
Epilepsy is a neurological disorder characterized by recurrent seizures that arise from abnormal electrical activity in the brain. Voltage-gated sodium channels (NaVs), responsible for the initiation and propagation of action potentials in neurons, play a critical role in the pathogenesis of epilepsy. This study sought to discover potential anticonvulsant compounds that interact with NaVs, specifically, the brain subtype hNaV1.2. A ligand-based QSAR model and a docking model were constructed, validated, and applied in a parallel virtual screening over the DrugBank database. Montelukast, Novobiocin, and Cinnarizine were selected for in vitro testing, using the patch-clamp technique, and all of them proved to inhibit hNaV1.2 channels heterologously expressed in HEK293 cells. Two hits were evaluated in the GASH/Sal model of audiogenic seizures and demonstrated promising activity, reducing the severity of sound-induced seizures at the doses tested. The combination of ligand- and structure-based models presents a valuable approach for identifying potential NaV inhibitors. These findings may provide a basis for further research into the development of new antiseizure drugs for the treatment of epilepsy.
Collapse
Affiliation(s)
- Manuel A Llanos
- Laboratory of Bioactive Compounds Research and Development (LIDeB), Department of Biological Sciences, Faculty of Exact Sciences, National University of La Plata (UNLP), La Plata B1900ADU, Argentina
| | - Nicolás Enrique
- Facultad de Ciencias Exactas, Departamento de Ciencias Biológicas, Instituto de Estudios Inmunológicos y Fisiopatológicos (IIFP), UNLP, CONICET, asociado CIC PBA, La Plata B1900BJW, Buenos Aires, Argentina
| | - Vega Esteban-López
- Institute for Neuroscience of Castilla y León (INCyL), University of Salamanca, Salamanca 37008, Spain
- Institute for Biomedical Research of Salamanca (IBSAL), Salamanca 37007, Spain
| | - Sebastian Scioli-Montoto
- Laboratory of Bioactive Compounds Research and Development (LIDeB), Department of Biological Sciences, Faculty of Exact Sciences, National University of La Plata (UNLP), La Plata B1900ADU, Argentina
| | - David Sánchez-Benito
- Institute for Neuroscience of Castilla y León (INCyL), University of Salamanca, Salamanca 37008, Spain
- Institute for Biomedical Research of Salamanca (IBSAL), Salamanca 37007, Spain
| | - María E Ruiz
- Laboratory of Bioactive Compounds Research and Development (LIDeB), Department of Biological Sciences, Faculty of Exact Sciences, National University of La Plata (UNLP), La Plata B1900ADU, Argentina
| | - Veronica Milesi
- Facultad de Ciencias Exactas, Departamento de Ciencias Biológicas, Instituto de Estudios Inmunológicos y Fisiopatológicos (IIFP), UNLP, CONICET, asociado CIC PBA, La Plata B1900BJW, Buenos Aires, Argentina
| | - Dolores E López
- Institute for Neuroscience of Castilla y León (INCyL), University of Salamanca, Salamanca 37008, Spain
- Institute for Biomedical Research of Salamanca (IBSAL), Salamanca 37007, Spain
| | - Alan Talevi
- Laboratory of Bioactive Compounds Research and Development (LIDeB), Department of Biological Sciences, Faculty of Exact Sciences, National University of La Plata (UNLP), La Plata B1900ADU, Argentina
| | - Pedro Martín
- Facultad de Ciencias Exactas, Departamento de Ciencias Biológicas, Instituto de Estudios Inmunológicos y Fisiopatológicos (IIFP), UNLP, CONICET, asociado CIC PBA, La Plata B1900BJW, Buenos Aires, Argentina
| | - Luciana Gavernet
- Laboratory of Bioactive Compounds Research and Development (LIDeB), Department of Biological Sciences, Faculty of Exact Sciences, National University of La Plata (UNLP), La Plata B1900ADU, Argentina
| |
Collapse
|
18
|
Viswanathan LG, Alapati S, Nagappa M, Mundlamuri R, Kenchaiah R, Asranna A, Padmanabha H, Seshagiri DV, Sinha S. Phenotypic features of epilepsy due to sodium channelopathies - A single center experience from India. J Neurosci Rural Pract 2023; 14:603-609. [PMID: 38059254 PMCID: PMC10696347 DOI: 10.25259/jnrp_329_2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Accepted: 09/19/2023] [Indexed: 12/08/2023] Open
Abstract
Objectives Nearly 40% of pediatric epilepsies have a genetic basis. There is significant phenotypic and genotypic heterogeneity, especially in epilepsy syndromes caused by sodium channelopathies. Sodium channel subunit 1A (SCN1A)-related epilepsy represents the archetypical channel-associated gene that has been linked to a wide spectrum of epilepsies of varying severity. Subsequently, other sodium channels have also been implicated in epilepsy and other neurodevelopmental disorders. This study aims to describe the phenotypes in children with sodium channelopathies from a center in Southern India. Materials and Methods This is a retrospective, descriptive, and single-center study. Out of 112 children presenting with epilepsy who underwent genetic testing between 2017 and 2021, 23 probands (M: F = 12:11) were identified to have clinically significant sodium channel mutations. Clinical presentation, electroencephalography, and imaging features of these patients were recorded. The utility of genetic test results (e.g., in planning another child, withdrawal of medications, or change in treatment) was also recorded. Results Age at onset of seizures ranged from day 4 of life to 3.5 years. Clinical epilepsy syndromes included generalized epilepsy with febrile seizures plus (n = 3), Dravet syndrome (n = 5), early infantile epileptic encephalopathy (n = 7), drug-resistant epilepsy (n = 5), and epilepsy with associated movement disorders (n = 3). The most common type of seizure was focal with impaired awareness (n = 18, 78.2%), followed by myoclonic jerks (n = 8, 34.78%), epileptic spasms (n = 7, 30.4%), bilateral tonic-clonic seizures/generalized tonic-clonic seizures (n = 3, 13%), and atonic seizures (n = 5, 23.8%). In addition to epilepsy, other phenotypic features that were discerned were microcephaly (n = 1), cerebellar ataxia (n = 2), and chorea and dystonia (n = 1). Conclusion Sodium channelopathies may present with seizure phenotypes that vary in severity. In addition to epilepsy, patients may also have other clinical features such as movement disorders. Early clinical diagnosis may aid in tailoring treatment for the given patient.
Collapse
Affiliation(s)
| | - Sandhya Alapati
- Department of Neurology, National Institute of Mental Health and Neurosciences, Bengaluru, Karnataka, India
| | - Madhu Nagappa
- Department of Neurology, National Institute of Mental Health and Neurosciences, Bengaluru, Karnataka, India
| | - Ravindranadh Mundlamuri
- Department of Neurology, National Institute of Mental Health and Neurosciences, Bengaluru, Karnataka, India
| | - Raghavendra Kenchaiah
- Department of Neurology, National Institute of Mental Health and Neurosciences, Bengaluru, Karnataka, India
| | - Ajay Asranna
- Department of Neurology, National Institute of Mental Health and Neurosciences, Bengaluru, Karnataka, India
| | - Hansashree Padmanabha
- Department of Neurology, National Institute of Mental Health and Neurosciences, Bengaluru, Karnataka, India
| | - Doniparthi V. Seshagiri
- Department of Neurology, National Institute of Mental Health and Neurosciences, Bengaluru, Karnataka, India
| | - Sanjib Sinha
- Department of Neurology, National Institute of Mental Health and Neurosciences, Bengaluru, Karnataka, India
| |
Collapse
|
19
|
Beneki E, Dimitriadis K, Tsatiris K, Aggeli K, Tsioufis K. Left Ventricular Noncompaction Cardiomyopathy Diagnosis in a Patient Presenting with Epileptic Seizure: A "Double-edged Sword". INNOVATIONS IN CLINICAL NEUROSCIENCE 2023; 20:9-11. [PMID: 38193099 PMCID: PMC10773597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/10/2024]
Abstract
Transient loss of consciousness (TLOC) is a common presentation to emergency departments and may be due to syncope or epileptic seizures. The distinction between both entities can be challenging. This case illustrates the need for a multidisciplinary team approach in TLOC to avoid misdiagnosis leading to improper treatment.
Collapse
Affiliation(s)
- Eirini Beneki
- Drs. Beneki, Dimitriadis, Aggeli, and Tsioufis are with First Cardiology Department, School of Medicine, Hippokration General Hospital, National and Kapodistrian University of Athens in Athens, Greece
| | - Kyriakos Dimitriadis
- Drs. Beneki, Dimitriadis, Aggeli, and Tsioufis are with First Cardiology Department, School of Medicine, Hippokration General Hospital, National and Kapodistrian University of Athens in Athens, Greece
| | - Konstantinos Tsatiris
- Dr. Tsatiris is with Department of Cardiology, Karditsa General Hospital in Karditsa, Greece
| | - Konstantina Aggeli
- Drs. Beneki, Dimitriadis, Aggeli, and Tsioufis are with First Cardiology Department, School of Medicine, Hippokration General Hospital, National and Kapodistrian University of Athens in Athens, Greece
| | - Konstantinos Tsioufis
- Drs. Beneki, Dimitriadis, Aggeli, and Tsioufis are with First Cardiology Department, School of Medicine, Hippokration General Hospital, National and Kapodistrian University of Athens in Athens, Greece
| |
Collapse
|
20
|
Banderali U, Moreno M, Martina M. The elusive Na v1.7: From pain to cancer. CURRENT TOPICS IN MEMBRANES 2023; 92:47-69. [PMID: 38007269 DOI: 10.1016/bs.ctm.2023.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2023]
Abstract
Voltage-gated sodium channels (Nav) are protein complexes that play fundamental roles in the transmission of signals in the nervous system, at the neuromuscular junction and in the heart. They are mainly present in excitable cells where they are responsible for triggering action potentials. Dysfunctions in Nav ion conduction give rise to a wide range of conditions, including neurological disorders, hypertension, arrhythmia, pain and cancer. Nav family 1 is composed of nine members, named numerically from 1 to 9. A Nax family also exists and is involved in body-fluid homeostasis. Of particular interest is Nav1.7 which is highly expressed in the sensory neurons of the dorsal root ganglions, where it is involved in the propagation of pain sensation. Gain-of-function mutations in Nav1.7 cause pathologies associated with increased pain sensitivity, while loss-of-function mutations cause reduced sensitivity to pain. The last decade has seen considerable effort in developing highly specific Nav1.7 blockers as pain medications, nonetheless, sufficient efficacy has yet to be achieved. Evidence is now conclusively showing that Navs are also present in many types of cancer cells, where they are involved in cell migration and invasiveness. Nav1.7 is anomalously expressed in endometrial, ovarian and lung cancers. Nav1.7 is also involved in Chemotherapy Induced Peripheral Neuropathy (CIPN). We propose that the knowledge and tools developed to study the role of Nav1.7 in pain can be exploited to develop novel cancer therapies. In this chapter, we illustrate the various aspects of Nav1.7 function in pain, cancer and CIPN, and outline therapeutic approaches.
Collapse
Affiliation(s)
- Umberto Banderali
- Human Health Therapeutics Research Centre, National Research Council of Canada, Montreal road, Ottawa, ON, Canada.
| | - Maria Moreno
- Human Health Therapeutics Research Centre, National Research Council of Canada, Montreal road, Ottawa, ON, Canada
| | - Marzia Martina
- Human Health Therapeutics Research Centre, National Research Council of Canada, Montreal road, Ottawa, ON, Canada
| |
Collapse
|
21
|
Minard AY, Clark CJ, Ahern CA, Piper RC. Beta-subunit-eliminated eHAP expression (BeHAPe) cells reveal subunit regulation of the cardiac voltage-gated sodium channel. J Biol Chem 2023; 299:105132. [PMID: 37544648 PMCID: PMC10506104 DOI: 10.1016/j.jbc.2023.105132] [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: 02/10/2023] [Revised: 07/27/2023] [Accepted: 07/31/2023] [Indexed: 08/08/2023] Open
Abstract
Voltage-gated sodium (NaV) channels drive the upstroke of the action potential and are comprised of a pore-forming α-subunit and regulatory β-subunits. The β-subunits modulate the gating, trafficking, and pharmacology of the α-subunit. These functions are routinely assessed by ectopic expression in heterologous cells. However, currently available expression systems may not capture the full range of these effects since they contain endogenous β-subunits. To better reveal β-subunit functions, we engineered a human cell line devoid of endogenous NaV β-subunits and their immediate phylogenetic relatives. This new cell line, β-subunit-eliminated eHAP expression (BeHAPe) cells, were derived from haploid eHAP cells by engineering inactivating mutations in the β-subunits SCN1B, SCN2B, SCN3B, and SCN4B, and other subfamily members MPZ (myelin protein zero(P0)), MPZL1, MPZL2, MPZL3, and JAML. In diploid BeHAPe cells, the cardiac NaV α-subunit, NaV1.5, was highly sensitive to β-subunit modulation and revealed that each β-subunit and even MPZ imparted unique gating properties. Furthermore, combining β1 and β2 with NaV1.5 generated a sodium channel with hybrid properties, distinct from the effects of the individual subunits. Thus, this approach revealed an expanded ability of β-subunits to regulate NaV1.5 activity and can be used to improve the characterization of other α/β NaV complexes.
Collapse
Affiliation(s)
- Annabel Y Minard
- Department of Molecular Physiology and Biophysics, University of Iowa College of Medicine, Iowa City, Iowa, United States
| | - Colin J Clark
- Department of Molecular Physiology and Biophysics, University of Iowa College of Medicine, Iowa City, Iowa, United States
| | - Christopher A Ahern
- Department of Molecular Physiology and Biophysics, University of Iowa College of Medicine, Iowa City, Iowa, United States.
| | - Robert C Piper
- Department of Molecular Physiology and Biophysics, University of Iowa College of Medicine, Iowa City, Iowa, United States.
| |
Collapse
|
22
|
Sanchez-Sandoval AL, Hernández-Plata E, Gomora JC. Voltage-gated sodium channels: from roles and mechanisms in the metastatic cell behavior to clinical potential as therapeutic targets. Front Pharmacol 2023; 14:1206136. [PMID: 37456756 PMCID: PMC10348687 DOI: 10.3389/fphar.2023.1206136] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 06/21/2023] [Indexed: 07/18/2023] Open
Abstract
During the second half of the last century, the prevalent knowledge recognized the voltage-gated sodium channels (VGSCs) as the proteins responsible for the generation and propagation of action potentials in excitable cells. However, over the last 25 years, new non-canonical roles of VGSCs in cancer hallmarks have been uncovered. Their dysregulated expression and activity have been associated with aggressive features and cancer progression towards metastatic stages, suggesting the potential use of VGSCs as cancer markers and prognostic factors. Recent work has elicited essential information about the signalling pathways modulated by these channels: coupling membrane activity to transcriptional regulation pathways, intracellular and extracellular pH regulation, invadopodia maturation, and proteolytic activity. In a promising scenario, the inhibition of VGSCs with FDA-approved drugs as well as with new synthetic compounds, reduces cancer cell invasion in vitro and cancer progression in vivo. The purpose of this review is to present an update regarding recent advances and ongoing efforts to have a better understanding of molecular and cellular mechanisms on the involvement of both pore-forming α and auxiliary β subunits of VGSCs in the metastatic processes, with the aim at proposing VGSCs as new oncological markers and targets for anticancer treatments.
Collapse
Affiliation(s)
- Ana Laura Sanchez-Sandoval
- Departamento de Neuropatología Molecular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Medicina Genómica, Hospital General de México “Dr Eduardo Liceaga”, Mexico City, Mexico
| | - Everardo Hernández-Plata
- Consejo Nacional de Humanidades, Ciencias y Tecnologías and Instituto Nacional de Medicina Genómica, Mexico City, Mexico
| | - Juan Carlos Gomora
- Departamento de Neuropatología Molecular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| |
Collapse
|
23
|
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: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 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
|
24
|
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
|
25
|
Alsaloum M, Labau JIR, Liu S, Effraim PR, Waxman SG. Stem cell-derived sensory neurons modelling inherited erythromelalgia: normalization of excitability. Brain 2023; 146:359-371. [PMID: 35088838 DOI: 10.1093/brain/awac031] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 12/30/2021] [Accepted: 01/06/2022] [Indexed: 01/11/2023] Open
Abstract
Effective treatment of pain remains an unmet healthcare need that requires new and effective therapeutic approaches. NaV1.7 has been genetically and functionally validated as a mediator of pain. Preclinical studies of NaV1.7-selective blockers have shown limited success and translation to clinical studies has been limited. The degree of NaV1.7 channel blockade necessary to attenuate neuronal excitability and ameliorate pain is an unanswered question important for drug discovery. Here, we utilize dynamic clamp electrophysiology and induced pluripotent stem cell-derived sensory neurons (iPSC-SNs) to answer this question for inherited erythromelalgia, a pain disorder caused by gain-of-function mutations in Nav1.7. We show that dynamic clamp can produce hyperexcitability in iPSC-SNs associated with two different inherited erythromelalgia mutations, NaV1.7-S241T and NaV1.7-I848T. We further show that blockade of approximately 50% of NaV1.7 currents can reverse neuronal hyperexcitability to baseline levels.
Collapse
Affiliation(s)
- Matthew Alsaloum
- Department of Neurology, Yale School of Medicine, New Haven, CT 06510, USA.,Center for Neuroscience and Regeneration Research, Yale University, West Haven, CT 06516, USA.,Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT 06516, USA.,Yale Medical Scientist Training Program, Yale School of Medicine, New Haven, CT 06510, USA.,Interdepartmental Neuroscience Program, Yale School of Medicine, New Haven, CT 06510, USA
| | - Julie I R Labau
- Department of Neurology, Yale School of Medicine, New Haven, CT 06510, USA.,Center for Neuroscience and Regeneration Research, Yale University, West Haven, CT 06516, USA.,Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT 06516, USA.,Department of Clinical Epidemiology and Medical Technology Assessment (KEMTA), Maastricht University Medical Centre, Maastricht, The Netherlands.,Department of Clinical Genetics, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Shujun Liu
- Department of Neurology, Yale School of Medicine, New Haven, CT 06510, USA.,Center for Neuroscience and Regeneration Research, Yale University, West Haven, CT 06516, USA.,Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Philip R Effraim
- Center for Neuroscience and Regeneration Research, Yale University, West Haven, CT 06516, USA.,Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT 06516, USA.,Department of Anesthesiology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Stephen G Waxman
- Department of Neurology, Yale School of Medicine, New Haven, CT 06510, USA.,Center for Neuroscience and Regeneration Research, Yale University, West Haven, CT 06516, USA.,Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT 06516, USA
| |
Collapse
|
26
|
Ssempijja F, Dare SS, Bukenya EEM, Kasozi KI, Kenganzi R, Fernandez EM, Vicente-Crespo M. Attenuation of Seizures, Cognitive Deficits, and Brain Histopathology by Phytochemicals of Imperata cylindrica (L.) P. Beauv (Poaceae) in Acute and Chronic Mutant Drosophila melanogaster Epilepsy Models. J Evid Based Integr Med 2023; 28:2515690X231160191. [PMID: 36866635 PMCID: PMC9989407 DOI: 10.1177/2515690x231160191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 08/09/2022] [Accepted: 02/06/2023] [Indexed: 03/04/2023] Open
Abstract
Imperata cylindrica is a globally distributed plant known for its antiepileptic attributes, but there is a scarcity of robust evidence for its efficacy. The study investigated neuroprotective attributes of Imperata cylindrica root extract on neuropathological features of epilepsy in a Drosophila melanogaster mutant model of epilepsy. It was conducted on 10-day-old (at the initiation of study) male post-eclosion bang-senseless paralytic Drosophila (parabss1) involved acute (1-3 h) and chronic (6-18 days) experiments; n = 50 flies per group (convulsions tests); n = 100 flies per group (learning/memory tests and histological examination). Administrations were done in 1 g standard fly food, per os. The mutant flies of study (parabss1) showed marked age-dependent progressive brain neurodegeneration and axonal degeneration, significant (P < 0.05) bang sensitivity and convulsions, and cognitive deficits due to up-regulation of the paralytic gene in our mutants. The neuropathological findings were significantly (P < 0.05) alleviated in dose and duration-dependent fashions to near normal/normal after acute and chronic treatment with extract similar to sodium valproate. Therefore, para is expressed in neurons of brain tissues in our mutant flies to bring about epilepsy phenotypes and behaviors of the current juvenile and old-adult mutant D. melanogaster models of epilepsy. The herb exerts neuroprotection by anticonvulsant and antiepileptogenic mechanisms in mutant D. melanogaster due to plant flavonoids, polyphenols, and chromones (1 and 2) which exert antioxidative and receptor or voltage-gated sodium ion channels' inhibitory properties, and thus causing reduced inflammation and apoptosis, increased tissue repair, and improved cell biology in the brain of mutant flies. The methanol root extract provides anticonvulsant and antiepileptogenic medicinal values which protect epileptic D. melanogaster. Therefore, the herb should be advanced for more experimental and clinical studies to confirm its efficacy in treating epilepsy.
Collapse
Affiliation(s)
- Fred Ssempijja
- Department of Anatomy, Faculty of Medicine, Mbarara University of Science and Technology, P.O Box 1410, Mbarara, Uganda
- Department of Anatomy, Faculty of Biomedical Sciences, Kampala International University, Western Campus, P.O Box 71, Ishaka, Bushenyi, Uganda
| | - Samuel Sunday Dare
- Department of Anatomy, Faculty of Biomedical Sciences, Kampala International University, Western Campus, P.O Box 71, Ishaka, Bushenyi, Uganda
- School of Medicine, Kabale University, P.O Box 317, Kabale, Uganda
| | - Edmund E. M. Bukenya
- Department of Anatomy, Faculty of Biomedical Sciences, Kampala International University, Western Campus, P.O Box 71, Ishaka, Bushenyi, Uganda
- School of Medicine, Kabale University, P.O Box 317, Kabale, Uganda
| | | | - Ritah Kenganzi
- Department of Medical Laboratory Sciences, School of Allied Health Sciences, Kampala International University Teaching Hospital, P.O Box 71, Ishaka, Bushenyi, Uganda
| | - Edgar Mario Fernandez
- Department of Anatomy, Faculty of Biomedical Sciences, Kampala International University, Western Campus, P.O Box 71, Ishaka, Bushenyi, Uganda
| | - Marta Vicente-Crespo
- Institute of Biomedical Research, Kampala International University Western Campus, P.O Box 71, Bushenyi, Uganda
- Department of Biochemistry, Faculty of Biomedical Sciences, Kampala International University Western Campus, P.O Box 71, Bushenyi, Uganda
| |
Collapse
|
27
|
Ye F, Du L, Huang W, Wang S. Shared Genetic Regulatory Networks Contribute to Neuropathic and Inflammatory Pain: Multi-Omics Systems Analysis. Biomolecules 2022; 12:1454. [PMID: 36291662 PMCID: PMC9599593 DOI: 10.3390/biom12101454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 09/27/2022] [Accepted: 10/07/2022] [Indexed: 11/17/2022] Open
Abstract
The mechanisms of chronic pain are complex, and genetic factors play an essential role in the development of chronic pain. Neuropathic pain (NP) and inflammatory pain (IP) are two primary components of chronic pain. Previous studies have uncovered some common biological processes in NP and IP. However, the shared genetic mechanisms remained poorly studied. We utilized multi-omics systematic analyses to investigate the shared genetic mechanisms of NP and IP. First, by integrating several genome-wide association studies (GWASs) with multi-omics data, we revealed the significant overlap of the gene co-expression modules in NP and IP. Further, we uncovered the shared biological pathways, including the previously reported mitochondrial electron transport and ATP metabolism, and stressed the role of genetic factors in chronic pain with neurodegenerative diseases. Second, we identified 24 conservative key drivers (KDs) contributing to NP and IP, containing two well-established pain genes, IL1B and OPRM1, and some novel potential pain genes, such as C5AR1 and SERPINE1. The subnetwork of those KDs highlighted the processes involving the immune system. Finally, gene expression analysis of the KDs in mouse models underlined two of the KDs, SLC6A15 and KCNQ5, with unidirectional regulatory functions in NP and IP. Our study provides strong evidence to support the current understanding of the shared genetic regulatory networks underlying NP and IP and potentially benefit the future common therapeutic avenues for chronic pain.
Collapse
Affiliation(s)
- Fang Ye
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Li Du
- Vitalant Research Institute, San Francisco, CA 94118, USA
- Department of Laboratory Medicine, University of California, San Francisco, CA 94143, USA
| | - Wenqi Huang
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Sheng Wang
- Institute for Neurodegenerative Diseases, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA 94143, USA
| |
Collapse
|
28
|
Zhu Z, Bolt E, Newmaster K, Osei-Bonsu W, Cohen S, Cuddapah VA, Gupta S, Paudel S, Samanta D, Dang LT, Carney PR, Naik S. SCN1B Genetic Variants: A Review of the Spectrum of Clinical Phenotypes and a Report of Early Myoclonic Encephalopathy. CHILDREN (BASEL, SWITZERLAND) 2022; 9:1507. [PMID: 36291443 PMCID: PMC9600564 DOI: 10.3390/children9101507] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 09/26/2022] [Accepted: 09/30/2022] [Indexed: 11/06/2022]
Abstract
Background: Pathogenic variants in SCN1B, the gene encoding voltage-gated sodium channel b1/b1B subunits are associated with a spectrum of epileptic disorders. This study describes a child with early myoclonic encephalopathy and a compound heterozygous variant in the SCN1B gene (p.Arg85Cys and c.3G>C/p.Met1), along with the child’s clinical response to anti-seizure medications (ASMs) and the ketogenic diet. We reviewed the current clinical literature pertinent to SCN1B-related epilepsy. Methods: We described the evaluation and management of a patient with SCN1B-related developmental and epileptic encephalopathy (DEE). We used the Medline and Pubmed databases to review the various neurological manifestations associated with SCN1B genetic variants, and summarize the functional studies performed on SCN1B variants. Results: We identified 20 families and six individuals (including the index case described herein) reported to have SCN1B-related epilepsy. Individuals with monoallelic pathogenic variants in SCN1B often present with genetic epilepsy with febrile seizures plus (GEFS+), while those with biallelic pathogenic variants may present with developmental and epileptic encephalopathy (DEE). Individuals with DEE present with seizures of various semiologies (commonly myoclonic seizures) and status epilepticus at early infancy and are treated with various antiseizure medications. In our index case, adjunctive fenfluramine was started at 8 months of age at 0.2 mg/kg/day with gradual incremental increases to the final dose of 0.7 mg/kg/day over 5 weeks. Fenfluramine was effective in the treatment of seizures, resulting in a 50% reduction in myoclonic seizures, status epilepticus, and generalized tonic-clonic seizures, as well as a 70−90% reduction in focal seizures, with no significant adverse effects. Following the initiation of fenfluramine at eight months of age, there was also a 50% reduction in the rate of hospitalizations. Conclusions: SCN1B pathogenic variants cause epilepsy and neurodevelopmental impairment with variable expressivity and incomplete penetrance. The severity of disease is associated with the zygosity of the pathogenic variants. Biallelic variants in SCN1B can result in early myoclonic encephalopathy, and adjunctive treatment with fenfluramine may be an effective treatment for SCN1B-related DEE. Further research on the efficacy and safety of using newer ASMs, such as fenfluramine in patients under the age of 2 years is needed.
Collapse
Affiliation(s)
- Zahra Zhu
- College of Medicine, Penn State University, Hershey, PA 17033, USA
| | - Elizabeth Bolt
- College of Medicine, Penn State University, Hershey, PA 17033, USA
| | - Kyra Newmaster
- College of Medicine, Penn State University, Hershey, PA 17033, USA
| | - Wendy Osei-Bonsu
- College of Medicine, Penn State University, Hershey, PA 17033, USA
| | - Stacey Cohen
- Epilepsy Neurogenetics Initiative, Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Vishnu Anand Cuddapah
- Epilepsy Neurogenetics Initiative, Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Siddharth Gupta
- Kennedy Krieger Institute, Department of Neurology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Sita Paudel
- Department of Pediatrics and Neurology, Penn State Health Milton Hershey Medical Center, Hershey, PA 17033, USA
| | - Debopam Samanta
- Division of Pediatric Neurology, Arkansas Children’s Hospital, Little Rock, AR 72202, USA
| | - Louis T. Dang
- Department of Pediatrics, Division of Pediatric Neurology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Paul R. Carney
- Pediatric Neurology Division, University of Missouri Health Care, Columbia, MO 65212, USA
| | - Sunil Naik
- Department of Pediatrics and Neurology, Penn State Health Milton Hershey Medical Center, Hershey, PA 17033, USA
| |
Collapse
|
29
|
Hodges SL, Bouza AA, Isom LL. Therapeutic Potential of Targeting Regulated Intramembrane Proteolysis Mechanisms of Voltage-Gated Ion Channel Subunits and Cell Adhesion Molecules. Pharmacol Rev 2022; 74:1028-1048. [PMID: 36113879 PMCID: PMC9553118 DOI: 10.1124/pharmrev.121.000340] [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: 11/08/2021] [Accepted: 05/13/2022] [Indexed: 10/03/2023] Open
Abstract
Several integral membrane proteins undergo regulated intramembrane proteolysis (RIP), a tightly controlled process through which cells transmit information across and between intracellular compartments. RIP generates biologically active peptides by a series of proteolytic cleavage events carried out by two primary groups of enzymes: sheddases and intramembrane-cleaving proteases (iCLiPs). Following RIP, fragments of both pore-forming and non-pore-forming ion channel subunits, as well as immunoglobulin super family (IgSF) members, have been shown to translocate to the nucleus to function in transcriptional regulation. As an example, the voltage-gated sodium channel β1 subunit, which is also an IgSF-cell adhesion molecule (CAM), is a substrate for RIP. β1 RIP results in generation of a soluble intracellular domain, which can regulate gene expression in the nucleus. In this review, we discuss the proposed RIP mechanisms of voltage-gated sodium, potassium, and calcium channel subunits as well as the roles of their generated proteolytic products in the nucleus. We also discuss other RIP substrates that are cleaved by similar sheddases and iCLiPs, such as IgSF macromolecules, including CAMs, whose proteolytically generated fragments function in the nucleus. Importantly, dysfunctional RIP mechanisms are linked to human disease. Thus, we will also review how understanding RIP events and subsequent signaling processes involving ion channel subunits and IgSF proteins may lead to the discovery of novel therapeutic targets. SIGNIFICANCE STATEMENT: Several ion channel subunits and immunoglobulin superfamily molecules have been identified as substrates of regulated intramembrane proteolysis (RIP). This signal transduction mechanism, which generates polypeptide fragments that translocate to the nucleus, is an important regulator of gene transcription. RIP may impact diseases of excitability, including epilepsy, cardiac arrhythmia, and sudden death syndromes. A thorough understanding of the role of RIP in gene regulation is critical as it may reveal novel therapeutic strategies for the treatment of previously intractable diseases.
Collapse
Affiliation(s)
- Samantha L Hodges
- Departments of Pharmacology (S.L.H., A.A.B., L.L.I.), Neurology (L.L.I.), and Molecular & Integrative Physiology (L.L.I.), University of Michigan Medical School, Ann Arbor, Michigan
| | - Alexandra A Bouza
- Departments of Pharmacology (S.L.H., A.A.B., L.L.I.), Neurology (L.L.I.), and Molecular & Integrative Physiology (L.L.I.), University of Michigan Medical School, Ann Arbor, Michigan
| | - Lori L Isom
- Departments of Pharmacology (S.L.H., A.A.B., L.L.I.), Neurology (L.L.I.), and Molecular & Integrative Physiology (L.L.I.), University of Michigan Medical School, Ann Arbor, Michigan
| |
Collapse
|
30
|
Subcellular dynamics and functional activity of the cleaved intracellular domain of the Na + channel β1 subunit. J Biol Chem 2022; 298:102174. [PMID: 35752364 PMCID: PMC9304784 DOI: 10.1016/j.jbc.2022.102174] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 06/08/2022] [Accepted: 06/16/2022] [Indexed: 11/24/2022] Open
Abstract
The voltage-gated Na+ channel β1 subunit, encoded by SCN1B, regulates cell surface expression and gating of α subunits and participates in cell adhesion. β1 is cleaved by α/β and γ-secretases, releasing an extracellular domain and intracellular domain (ICD), respectively. Abnormal SCN1B expression/function is linked to pathologies including epilepsy, cardiac arrhythmia, and cancer. In this study, we sought to determine the effect of secretase cleavage on β1 function in breast cancer cells. Using a series of GFP-tagged β1 constructs, we show that β1-GFP is mainly retained intracellularly, particularly in the endoplasmic reticulum and endolysosomal pathway, and accumulates in the nucleus. Reduction in endosomal β1-GFP levels occurred following γ-secretase inhibition, implicating endosomes and/or the preceding plasma membrane as important sites for secretase processing. Using live-cell imaging, we also report β1ICD-GFP accumulation in the nucleus. Furthermore, β1-GFP and β1ICD-GFP both increased Na+ current, whereas β1STOP-GFP, which lacks the ICD, did not, thus highlighting that the β1-ICD is necessary and sufficient to increase Na+ current measured at the plasma membrane. Importantly, although the endogenous Na+ current expressed in MDA-MB-231 cells is tetrodotoxin (TTX)-resistant (carried by Nav1.5), the Na+ current increased by β1-GFP or β1ICD-GFP was TTX-sensitive. Finally, we found β1-GFP increased mRNA levels of the TTX-sensitive α subunits SCN1A/Nav1.1 and SCN9A/Nav1.7. Taken together, this work suggests that the β1-ICD is a critical regulator of α subunit function in cancer cells. Our data further highlight that γ-secretase may play a key role in regulating β1 function in breast cancer.
Collapse
|
31
|
Borowicz-Reutt KK. Effects of Antiarrhythmic Drugs on Antiepileptic Drug Action-A Critical Review of Experimental Findings. Int J Mol Sci 2022; 23:ijms23052891. [PMID: 35270033 PMCID: PMC8911389 DOI: 10.3390/ijms23052891] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 02/28/2022] [Accepted: 03/04/2022] [Indexed: 01/27/2023] Open
Abstract
Severe cardiac arrhythmias developing in the course of seizures increase the risk of SUDEP (sudden unexpected death in epilepsy). Hence, epilepsy patients with pre-existing arrhythmias should receive appropriate pharmacotherapy. Concomitant treatment with antiarrhythmic and antiseizure medications creates, however, the possibility of drug–drug interactions. This is due, among other reasons, to a similar mechanism of action. Both groups of drugs inhibit the conduction of electrical impulses in excitable tissues. The aim of this review was the analysis of such interactions in animal seizure models, including the maximal electroshock (MES) test in mice, a widely accepted screening test for antiepileptic drugs.
Collapse
Affiliation(s)
- Kinga K Borowicz-Reutt
- Independent Unit of Experimental Neuropathophysiology, Department of Toxicology, Medical University of Lublin, 20-090 Lublin, Poland
| |
Collapse
|
32
|
Horváth B, Szentandrássy N, Almássy J, Dienes C, Kovács ZM, Nánási PP, Banyasz T. Late Sodium Current of the Heart: Where Do We Stand and Where Are We Going? Pharmaceuticals (Basel) 2022; 15:ph15020231. [PMID: 35215342 PMCID: PMC8879921 DOI: 10.3390/ph15020231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 02/07/2022] [Accepted: 02/09/2022] [Indexed: 02/05/2023] Open
Abstract
Late sodium current has long been linked to dysrhythmia and contractile malfunction in the heart. Despite the increasing body of accumulating information on the subject, our understanding of its role in normal or pathologic states is not complete. Even though the role of late sodium current in shaping action potential under physiologic circumstances is debated, it’s unquestioned role in arrhythmogenesis keeps it in the focus of research. Transgenic mouse models and isoform-specific pharmacological tools have proved useful in understanding the mechanism of late sodium current in health and disease. This review will outline the mechanism and function of cardiac late sodium current with special focus on the recent advances of the area.
Collapse
Affiliation(s)
- Balázs Horváth
- Department of Physiology, University of Debrecen, 4032 Debrecen, Hungary; (B.H.); (N.S.); (J.A.); (C.D.); (Z.M.K.); (P.P.N.)
| | - Norbert Szentandrássy
- Department of Physiology, University of Debrecen, 4032 Debrecen, Hungary; (B.H.); (N.S.); (J.A.); (C.D.); (Z.M.K.); (P.P.N.)
- Department of Basic Medical Sciences, Faculty of Dentistry, University of Debrecen, 4032 Debrecen, Hungary
| | - János Almássy
- Department of Physiology, University of Debrecen, 4032 Debrecen, Hungary; (B.H.); (N.S.); (J.A.); (C.D.); (Z.M.K.); (P.P.N.)
| | - Csaba Dienes
- Department of Physiology, University of Debrecen, 4032 Debrecen, Hungary; (B.H.); (N.S.); (J.A.); (C.D.); (Z.M.K.); (P.P.N.)
| | - Zsigmond Máté Kovács
- Department of Physiology, University of Debrecen, 4032 Debrecen, Hungary; (B.H.); (N.S.); (J.A.); (C.D.); (Z.M.K.); (P.P.N.)
| | - Péter P. Nánási
- Department of Physiology, University of Debrecen, 4032 Debrecen, Hungary; (B.H.); (N.S.); (J.A.); (C.D.); (Z.M.K.); (P.P.N.)
- Department of Dental Physiology and Pharmacology, University of Debrecen, 4032 Debrecen, Hungary
| | - Tamas Banyasz
- Department of Physiology, University of Debrecen, 4032 Debrecen, Hungary; (B.H.); (N.S.); (J.A.); (C.D.); (Z.M.K.); (P.P.N.)
- Correspondence: ; Tel.: +36-(52)-255-575; Fax: +36-(52)-255-116
| |
Collapse
|
33
|
Alsaloum M, Waxman SG. iPSCs and DRGs: stepping stones to new pain therapies. Trends Mol Med 2022; 28:110-122. [PMID: 34933815 PMCID: PMC8810720 DOI: 10.1016/j.molmed.2021.11.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 11/23/2021] [Accepted: 11/24/2021] [Indexed: 02/03/2023]
Abstract
There is a pressing need for more effective nonaddictive treatment options for pain. Pain signals are transmitted from the periphery into the spinal cord via dorsal root ganglion (DRG) neurons, whose excitability is driven by voltage-gated sodium (NaV) channels. Three NaV channels (NaV1.7, NaV1.8, and NaV1.9), preferentially expressed in DRG neurons, play important roles in pain signaling in humans. Blockade of these channels may provide a novel approach to the treatment of pain, but clinical translation of preclinical results has been challenging, in part due to differences between rodent and human DRG neurons. Human DRG neurons and iPSC-derived sensory neurons (iPSC-SNs) provide new preclinical platforms that may facilitate the development of novel pain therapeutics.
Collapse
Affiliation(s)
- Matthew Alsaloum
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA; Center for Neuroscience & Regeneration Research, Yale University, West Haven, CT, USA; Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT, USA; Yale Medical Scientist Training Program, Yale School of Medicine, New Haven, CT, USA; Interdepartmental Neuroscience Program, Yale School of Medicine, New Haven, CT, USA
| | - Stephen G Waxman
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA; Center for Neuroscience & Regeneration Research, Yale University, West Haven, CT, USA; Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT, USA.
| |
Collapse
|
34
|
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: 5.0] [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
|
35
|
Sen S, Hallee L, Lam CK. The Potential of Gamma Secretase as a Therapeutic Target for Cardiac Diseases. J Pers Med 2021; 11:jpm11121294. [PMID: 34945766 PMCID: PMC8703931 DOI: 10.3390/jpm11121294] [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: 10/31/2021] [Revised: 11/24/2021] [Accepted: 12/01/2021] [Indexed: 11/16/2022] Open
Abstract
Heart diseases are some of the most common and pressing threats to human health worldwide. The American Heart Association and the National Institute of Health jointly work to annually update data on cardiac diseases. In 2018, 126.9 million Americans were reported as having some form of cardiac disorder, with an estimated direct and indirect total cost of USD 363.4 billion. This necessitates developing therapeutic interventions for heart diseases to improve human life expectancy and economic relief. In this review, we look into gamma-secretase as a potential therapeutic target for cardiac diseases. Gamma-secretase, an aspartyl protease enzyme, is responsible for the cleavage and activation of a number of substrates that are relevant to normal cardiac development and function as found in mutation studies. Some of these substrates are involved in downstream signaling processes and crosstalk with pathways relevant to heart diseases. Most of the substrates and signaling events we explored were found to be potentially beneficial to maintain cardiac function in diseased conditions. This review presents an updated overview of the current knowledge on gamma-secretase processing of cardiac-relevant substrates and seeks to understand if the modulation of gamma-secretase activity would be beneficial to combat cardiac diseases.
Collapse
Affiliation(s)
- Sujoita Sen
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA;
| | - Logan Hallee
- Department of Mathematical Sciences, University of Delaware, Newark, DE 19716, USA;
| | - Chi Keung Lam
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA;
- Correspondence: ; Tel.: +1-302-831-3165
| |
Collapse
|
36
|
Torrealba-Acosta G, Butt H, Edmondson EA, Willaert R, Viswanathan A, Goldman AM. A Neurostimulation-Triggered Trigeminal Neuralgia-like Pain: Risk Factors and Management. Neurol Clin Pract 2021; 11:e760-e762. [PMID: 34840901 DOI: 10.1212/cpj.0000000000001050] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Accepted: 12/14/2020] [Indexed: 11/15/2022]
Affiliation(s)
- Gabriel Torrealba-Acosta
- Department of Neurology (GT-A, HB, EAE, AMG), Baylor College of Medicine, Houston, TX; GeneDx (RW); and Department of Neurosurgery (AV), Baylor College of Medicine, Houston, TX
| | - Haroon Butt
- Department of Neurology (GT-A, HB, EAE, AMG), Baylor College of Medicine, Houston, TX; GeneDx (RW); and Department of Neurosurgery (AV), Baylor College of Medicine, Houston, TX
| | - Everton A Edmondson
- Department of Neurology (GT-A, HB, EAE, AMG), Baylor College of Medicine, Houston, TX; GeneDx (RW); and Department of Neurosurgery (AV), Baylor College of Medicine, Houston, TX
| | - Rebecca Willaert
- Department of Neurology (GT-A, HB, EAE, AMG), Baylor College of Medicine, Houston, TX; GeneDx (RW); and Department of Neurosurgery (AV), Baylor College of Medicine, Houston, TX
| | - Ashwin Viswanathan
- Department of Neurology (GT-A, HB, EAE, AMG), Baylor College of Medicine, Houston, TX; GeneDx (RW); and Department of Neurosurgery (AV), Baylor College of Medicine, Houston, TX
| | - Alica M Goldman
- Department of Neurology (GT-A, HB, EAE, AMG), Baylor College of Medicine, Houston, TX; GeneDx (RW); and Department of Neurosurgery (AV), Baylor College of Medicine, Houston, TX
| |
Collapse
|
37
|
Sahly AN, Shevell M, Sadleir LG, Myers KA. SUDEP risk and autonomic dysfunction in genetic epilepsies. Auton Neurosci 2021; 237:102907. [PMID: 34773737 DOI: 10.1016/j.autneu.2021.102907] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 10/11/2021] [Accepted: 11/06/2021] [Indexed: 01/02/2023]
Abstract
The underlying pathophysiology of sudden unexpected death in epilepsy (SUDEP) remains unclear. This phenomenon is likely multifactorial, and there is considerable evidence that genetic factors play a role. There are certain genetic causes of epilepsy in which the risk of SUDEP appears to be increased relative to epilepsy overall. For individuals with pathogenic variants in genes including SCN1A, SCN1B, SCN8A, SCN2A, GNB5, KCNA1 and DEPDC5, there are varying degrees of evidence to suggest an increased risk for sudden death. Why the risk for sudden death is higher is not completely clear; however, in many cases pathogenic variants in these genes are also associated with autonomic dysfunction, which is hypothesized as a contributing factor to SUDEP. We review the evidence for increased SUDEP risk for patients with epilepsy due to pathogenic variants in these genes, and also discuss what is known about autonomic dysfunction in these contexts.
Collapse
Affiliation(s)
- Ahmed N Sahly
- Division of Neurology, Department of Pediatrics, Montreal Children's Hospital, McGill University Health Centre, Montreal, Quebec, Canada; Department of Neurosciences, King Faisal Specialist Hospital & Research Centre, Jeddah, Saudi Arabia
| | - Michael Shevell
- Division of Neurology, Department of Pediatrics, Montreal Children's Hospital, McGill University Health Centre, Montreal, Quebec, Canada; Department of Neurology and Neurosurgery, Montreal Children's Hospital, McGill University Health Centre, Montreal, Quebec, Canada; Research Institute of the McGill University Medical Centre, Montreal, Quebec, Canada
| | - Lynette G Sadleir
- Department of Paediatrics and Child Health, University of Otago, Wellington, New Zealand
| | - Kenneth A Myers
- Division of Neurology, Department of Pediatrics, Montreal Children's Hospital, McGill University Health Centre, Montreal, Quebec, Canada; Department of Neurology and Neurosurgery, Montreal Children's Hospital, McGill University Health Centre, Montreal, Quebec, Canada; Research Institute of the McGill University Medical Centre, Montreal, Quebec, Canada.
| |
Collapse
|
38
|
Tibery DV, de Souza ACB, Mourão CBF, do Nascimento JM, Schwartz EF. Purification and characterization of peptides Ap2, Ap3 and Ap5 (ω-toxins) from the venom of the Brazilian tarantula Acanthoscurria paulensis. Peptides 2021; 145:170622. [PMID: 34363923 DOI: 10.1016/j.peptides.2021.170622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 07/30/2021] [Accepted: 08/01/2021] [Indexed: 11/21/2022]
Abstract
Peptides isolated from spider venoms are of pharmacological interest due to their neurotoxic activity, acting on voltage-dependent ion channels present in different types of human body tissues. Three peptide toxins titled as Ap2, Ap3 and Ap5 were purified by RP-HPLC from Acanthoscurria paulensis venom. They were partially sequenced by MALDI In-source Decay method and their sequences were completed and confirmed by transcriptome analysis of the venom gland. The Ap2, Ap3 and Ap5 peptides have, respectively, 42, 41 and 46 amino acid residues, and experimental molecular masses of 4886.3, 4883.7 and 5454.7 Da, with the Ap2 peptide presenting an amidated C-terminus. Amongst the assayed channels - NaV1.1, NaV1.5, NaV1.7, CaV1.2, CaV2.1 and CaV2.2 - Ap2, Ap3 and Ap5 inhibited 20-30 % of CaV2.1 current at 1 μM concentration. Ap3 also inhibited sodium current in NaV1.1, Nav1.5 and Nav1.7 channels by 6.6 ± 1.91 % (p = 0.0276), 4.2 ± 1.09 % (p = 0.0185) and 16.05 ± 2.75 % (p = 0.0282), respectively. Considering that Ap2, Ap3 and Ap5 belong to the 'U'-unknown family of spider toxins, which has few descriptions of biological activity, the present work contributes to the knowledge of these peptides and demonstrates this potential as channel modulators.
Collapse
Affiliation(s)
- Diogo Vieira Tibery
- Laboratório de Neurofarmacologia, Departamento de Ciências Fisiológicas, Universidade de Brasília, Brasília, Brazil
| | | | - Caroline Barbosa Farias Mourão
- Laboratório de Neurofarmacologia, Departamento de Ciências Fisiológicas, Universidade de Brasília, Brasília, Brazil; Instituto Federal de Educação, Ciência e Tecnologia de Brasília, Campus Ceilândia, Brasília, DF, Brazil
| | | | - Elisabeth Ferroni Schwartz
- Laboratório de Neurofarmacologia, Departamento de Ciências Fisiológicas, Universidade de Brasília, Brasília, Brazil.
| |
Collapse
|
39
|
Bagheri S, Haddadi R, Saki S, Kourosh-Arami M, Komaki A. The effect of sodium channels on neurological/neuronal disorders: A systematic review. Int J Dev Neurosci 2021; 81:669-685. [PMID: 34687079 DOI: 10.1002/jdn.10153] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 10/06/2021] [Accepted: 10/19/2021] [Indexed: 12/19/2022] Open
Abstract
Neurological and neuronal disorders are associated with structural, biochemical, or electrical abnormalities in the nervous system. Many neurological diseases have not yet been discovered. Interventions used for the treatment of these disorders include avoidance measures, lifestyle changes, physiotherapy, neurorehabilitation, pain management, medication, and surgery. In the sodium channelopathies, alterations in the structure, expression, and function of voltage-gated sodium channels (VGSCs) are considered as the causes of neurological and neuronal diseases. Online databases, including Scopus, Science Direct, Google Scholar, and PubMed were assessed for studies published between 1977 and 2020 using the keywords of review, sodium channels blocker, neurological diseases, and neuronal diseases. VGSCs consist of one α subunit and two β subunits. These subunits are known to regulate the gating kinetics, functional characteristics, and localization of the ion channel. These channels are involved in cell migration, cellular connections, neuronal pathfinding, and neurite outgrowth. Through the VGSC, the action potential is triggered and propagated in the neurons. Action potentials are physiological functions and passage of impermeable ions. The electrophysiological properties of these channels and their relationship with neurological and neuronal disorders have been identified. Subunit mutations are involved in the development of diseases, such as epilepsy, multiple sclerosis, autism, and Alzheimer's disease. Accordingly, we conducted a review of the link between VGSCs and neurological and neuronal diseases. Also, novel therapeutic targets were introduced for future drug discoveries.
Collapse
Affiliation(s)
- Shokufeh Bagheri
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran.,Department of Neuroscience, School of Science and Advanced Technologies in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Rasool Haddadi
- Department of Pharmacology, School of Pharmacy, Hamadan University of Medical Science, Hamadan, Iran
| | - Sahar Saki
- Vice-Chancellor for Research and Technology, Hamadan University of Medical Science, Hamadan, Iran
| | - Masoumeh Kourosh-Arami
- Department of Neuroscience, School of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Alireza Komaki
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran.,Department of Neuroscience, School of Science and Advanced Technologies in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
40
|
Fonseca-Barriendos D, Frías-Soria CL, Pérez-Pérez D, Gómez-López R, Borroto Escuela DO, Rocha L. Drug-resistant epilepsy: Drug target hypothesis and beyond the receptors. Epilepsia Open 2021; 7 Suppl 1:S23-S33. [PMID: 34542940 PMCID: PMC9340308 DOI: 10.1002/epi4.12539] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 08/21/2021] [Accepted: 08/27/2021] [Indexed: 12/28/2022] Open
Abstract
Epilepsy is a chronic neurological disorder that affects more than 50 million people worldwide. Despite a recent introduction of antiseizure drugs for the treatment of epileptic seizures, one-third of these patients suffer from drug-resistant epilepsy (DRE). The therapeutic target hypothesis is a cited theory to explain DRE. According to the target hypothesis, the failure to achieve seizure freedom leads to alteration of the structure and/or function of the antiseizure medication (ASM) target. However, this hypothesis fails to explain why patients with DRE do not respond to antiseizure medications of different targets. This review presents different conditions, such as epigenetic mechanisms and protein-protein interactions that may result in alterations of diverse drug targets using different mechanisms. These novel conditions represent new targets to control DRE.
Collapse
Affiliation(s)
| | | | - Daniel Pérez-Pérez
- Plan of Combined Studies in Medicine (PECEM), Faculty of Medicine, UNAM, México City, Mexico
| | - Rosenda Gómez-López
- Escuela Nacional de Medicina y Homeopatía, Instituto Politécnico Nacional, Mexico City, México
| | | | - Luisa Rocha
- Pharmacobiology Department, Center for Research and Advanced Studies, México City, México
| |
Collapse
|
41
|
Rivaud MR, Delmar M, Remme CA. Heritable arrhythmia syndromes associated with abnormal cardiac sodium channel function: ionic and non-ionic mechanisms. Cardiovasc Res 2021; 116:1557-1570. [PMID: 32251506 PMCID: PMC7341171 DOI: 10.1093/cvr/cvaa082] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 03/01/2020] [Accepted: 04/01/2020] [Indexed: 12/19/2022] Open
Abstract
The cardiac sodium channel NaV1.5, encoded by the SCN5A gene, is responsible for the fast upstroke of the action potential. Mutations in SCN5A may cause sodium channel dysfunction by decreasing peak sodium current, which slows conduction and facilitates reentry-based arrhythmias, and by enhancing late sodium current, which prolongs the action potential and sets the stage for early afterdepolarization and arrhythmias. Yet, some NaV1.5-related disorders, in particular structural abnormalities, cannot be directly or solely explained on the basis of defective NaV1.5 expression or biophysics. An emerging concept that may explain the large disease spectrum associated with SCN5A mutations centres around the multifunctionality of the NaV1.5 complex. In this alternative view, alterations in NaV1.5 affect processes that are independent of its canonical ion-conducting role. We here propose a novel classification of NaV1.5 (dys)function, categorized into (i) direct ionic effects of sodium influx through NaV1.5 on membrane potential and consequent action potential generation, (ii) indirect ionic effects of sodium influx on intracellular homeostasis and signalling, and (iii) non-ionic effects of NaV1.5, independent of sodium influx, through interactions with macromolecular complexes within the different microdomains of the cardiomyocyte. These indirect ionic and non-ionic processes may, acting alone or in concert, contribute significantly to arrhythmogenesis. Hence, further exploration of these multifunctional effects of NaV1.5 is essential for the development of novel preventive and therapeutic strategies.
Collapse
Affiliation(s)
- Mathilde R Rivaud
- Department of Clinical and Experimental Cardiology, Heart Center, Amsterdam UMC (location AMC), University of Amsterdam, Amsterdam Cardiovascular Sciences, Meigberdreef 15, 1105AZ Amsterdam, The Netherlands
| | - Mario Delmar
- The Leon H. Charney Division of Cardiology, New York University School of Medicine, 435 E 30th St, NSB 707, New York, NY 10016, USA
| | - Carol Ann Remme
- Department of Clinical and Experimental Cardiology, Heart Center, Amsterdam UMC (location AMC), University of Amsterdam, Amsterdam Cardiovascular Sciences, Meigberdreef 15, 1105AZ Amsterdam, The Netherlands
| |
Collapse
|
42
|
Alsaloum M, Labau JIR, Sosniak D, Zhao P, Almomani R, Gerrits M, Hoeijmakers JGJ, Lauria G, Faber CG, Waxman SG, Dib-Hajj S. A novel gain-of-function sodium channel β2 subunit mutation in idiopathic small fiber neuropathy. J Neurophysiol 2021; 126:827-839. [PMID: 34320850 DOI: 10.1152/jn.00184.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Small fiber neuropathy (SFN) is a common condition affecting thinly myelinated Aδ and unmyelinated C fibers, often resulting in excruciating pain and dysautonomia. SFN has been associated with several conditions, but a significant number of cases have no discernible cause. Recent genetic studies have identified potentially pathogenic gain-of-function mutations in several the pore-forming voltage-gated sodium channel α subunits (NaVs) in a subset of patients with SFN, but the auxiliary sodium channel β subunits have been less implicated in the development of the disease. β subunits modulate NaV trafficking and gating, and several mutations have been linked to epilepsy and cardiac dysfunction. Recently, we provided the first evidence for the contribution of a mutation in the β2-subunit to pain in human painful diabetic neuropathy. Here, we provide the first evidence for the involvement of a sodium channel β subunit mutation in the pathogenesis of SFN with no other known causes. We show, through current-clamp analysis, that the newly-identified Y69H variant of the β2 subunit induces neuronal hyperexcitability in dorsal root ganglion neurons, lowering the threshold for action potential firing and allowing for increased repetitive action potential spiking. Underlying the hyperexcitability induced by the β2-Y69H variant, we demonstrate an upregulation in tetrodotoxin-sensitive, but not tetrodotoxin-resistant sodium currents. This provides the first evidence for the involvement of β2 subunits in SFN and strengthens the link between sodium channel β subunits and the development of neuropathic pain in humans.
Collapse
Affiliation(s)
- Matthew Alsaloum
- Department of Neurology, Yale University School of Medicine, New Haven, CT, United States.,Center for Neuroscience and Regeneration Research, Yale University, West Haven, CT, United States.,Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT, United States.,Yale Medical Scientist Training Program, Yale School of Medicine, New Haven, CT, United States.,Interdepartmental Neuroscience Program, Yale School of Medicine, New Haven, CT, United States
| | - Julie I R Labau
- Department of Neurology, Yale University School of Medicine, New Haven, CT, United States.,Center for Neuroscience and Regeneration Research, Yale University, West Haven, CT, United States.,Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT, United States.,Department of Genetics and Cell Biology, Clinical Genomics Unit, Maastricht University, Maastricht, the Netherlands.,Department of Neurology, Maastricht University Medical Centre+, Maastricht, the Netherlands
| | - Daniel Sosniak
- Department of Neurology, Yale University School of Medicine, New Haven, CT, United States.,Center for Neuroscience and Regeneration Research, Yale University, West Haven, CT, United States.,Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT, United States
| | - Peng Zhao
- Department of Neurology, Yale University School of Medicine, New Haven, CT, United States.,Center for Neuroscience and Regeneration Research, Yale University, West Haven, CT, United States.,Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT, United States
| | - Rowida Almomani
- Department of Genetics and Cell Biology, Clinical Genomics Unit, Maastricht University, Maastricht, the Netherlands.,Department of Medical Laboratory Sciences, Jordan University of Science and Technology, Irbid, Jordan
| | - Monique Gerrits
- Department of Clinical Genetics, Maastricht University Medical Centre+, Maastricht, the Netherlands
| | | | - Giuseppe Lauria
- Neuroalgology Unit, IRCCS Foundation "Carlo Besta" Neurological Institute, Milan, Italy.,Department of Biomedical and Clinical Sciences "Luigi Sacco," University of Milan, Milan, Italy
| | - Catherina G Faber
- Department of Neurology, Maastricht University Medical Centre+, Maastricht, the Netherlands
| | - Stephen G Waxman
- Department of Neurology, Yale University School of Medicine, New Haven, CT, United States.,Center for Neuroscience and Regeneration Research, Yale University, West Haven, CT, United States.,Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT, United States
| | - Sulayman Dib-Hajj
- Department of Neurology, Yale University School of Medicine, New Haven, CT, United States.,Center for Neuroscience and Regeneration Research, Yale University, West Haven, CT, United States.,Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT, United States
| |
Collapse
|
43
|
Beasley HK, Rodman TA, Collins GV, Hinton A, Exil V. TMEM135 is a Novel Regulator of Mitochondrial Dynamics and Physiology with Implications for Human Health Conditions. Cells 2021; 10:cells10071750. [PMID: 34359920 PMCID: PMC8303332 DOI: 10.3390/cells10071750] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 07/05/2021] [Accepted: 07/06/2021] [Indexed: 12/16/2022] Open
Abstract
Transmembrane proteins (TMEMs) are integral proteins that span biological membranes. TMEMs function as cellular membrane gates by modifying their conformation to control the influx and efflux of signals and molecules. TMEMs also reside in and interact with the membranes of various intracellular organelles. Despite much knowledge about the biological importance of TMEMs, their role in metabolic regulation is poorly understood. This review highlights the role of a single TMEM, transmembrane protein 135 (TMEM135). TMEM135 is thought to regulate the balance between mitochondrial fusion and fission and plays a role in regulating lipid droplet formation/tethering, fatty acid metabolism, and peroxisomal function. This review highlights our current understanding of the various roles of TMEM135 in cellular processes, organelle function, calcium dynamics, and metabolism.
Collapse
Affiliation(s)
- Heather K. Beasley
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA; (H.K.B.); (T.A.R.)
| | - Taylor A. Rodman
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA; (H.K.B.); (T.A.R.)
| | - Greg V. Collins
- Fraternal Order of Eagles Diabetes Research Center, Iowa City, IA 52242, USA;
- Department of Pediatrics-Cardiology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Antentor Hinton
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA; (H.K.B.); (T.A.R.)
- Correspondence: (A.H.J.); (V.E.)
| | - Vernat Exil
- Fraternal Order of Eagles Diabetes Research Center, Iowa City, IA 52242, USA;
- Department of Pediatrics-Cardiology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
- Correspondence: (A.H.J.); (V.E.)
| |
Collapse
|
44
|
Al-Ward H, Liu CY, Liu N, Shaher F, Al-Nusaif M, Mao J, Xu H. Voltage-Gated Sodium Channel β1 Gene: An Overview. Hum Hered 2021; 85:101-109. [PMID: 34038903 DOI: 10.1159/000516388] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 04/01/2021] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Voltage-gated sodium channels are protein complexes composed of 2 subunits, namely, pore-forming α- and regulatory β-subunits. A β-subunit consists of 5 proteins encoded by 4 genes (i.e., SCN1B-SCN4B). SUMMARY β1-Subunits regulate sodium ion channel functions, including gating properties, subcellular localization, and kinetics. Key Message: Sodium channel β1- and its variant β1B-subunits are encoded by SCN1B. These variants are associated with many human diseases, such as epilepsy, Brugada syndrome, Dravet syndrome, and cancers. On the basis of previous research, we aimed to provide an overview of the structure, expression, and involvement of SCN1B in physiological processes and focused on its role in diseases.
Collapse
Affiliation(s)
- Hisham Al-Ward
- Department of Biochemistry and Molecular Biology, Jiamusi University School of Basic Medical Sciences, Jiamusi, China
| | - Chun-Yang Liu
- Department of Biochemistry and Molecular Biology, Ankang University School of Medicine, Ankang, China
| | - Ning Liu
- Department of Biochemistry and Molecular Biology, Jiamusi University School of Basic Medical Sciences, Jiamusi, China
| | - Fahmi Shaher
- Department of Pathophysiology, Jiamusi University School of Basic Medical Sciences, Jiamusi, China
| | - Murad Al-Nusaif
- Department of Neurology, Dalian Medical University, Dalian, China
| | - Jing Mao
- Department of Biochemistry and Molecular Biology, Jiamusi University School of Basic Medical Sciences, Jiamusi, China
| | - Hui Xu
- Department of Biochemistry and Molecular Biology, Jiamusi University School of Basic Medical Sciences, Jiamusi, China
| |
Collapse
|
45
|
Wen Z, Chen J, Zhu B, Lu Y, Chen L. Effects of SCN1A and SCN2A polymorphisms on responsiveness to valproic acid monotherapy in epileptic children: A protocol for systematic review and meta-analysis. Medicine (Baltimore) 2021; 100:e25831. [PMID: 34011048 PMCID: PMC8136989 DOI: 10.1097/md.0000000000025831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 04/16/2021] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND : The gene mutation of coding sodium channel is one of the most important mechanisms in the pathogenesis of epilepsy. There exists a large inter-individual variation in the efficacy of valproic acid (VPA) against epilepsy. What are the genetic polymorphism influences of sodium channels on VPA response is still under discussion. In this study, a meta-analysis was used to further explore the effects of SCN1A and SCN2A gene polymorphism on VPA response in children with epilepsy. METHODS : The PubMed, EMBASE, Web of Science, Chinese National Knowledge Infrastructure, Chinese Science and Technique Journals Database, China Biology Medicine disc, and Wan Fang Database were searched up to April 2021 for appropriate studies regarding the association between SCN1A and SCN2A gene polymorphism on VPA response in children suffering from epilepsy. The meta-analysis was conducted by Review Manager 5.3 software. RESULTS : The results of this meta-analysis will be submitted to a peer-reviewed journal for publication. CONCLUSION : This meta-analysis will summarize the effects of SCN1A and SCN2A gene polymorphisms on VPA response in children with epilepsy. OSF REGISTRATION NUMBER DOI 10.17605/OSF.IO/N2786.
Collapse
Affiliation(s)
| | - Jiang Chen
- Department of Pediatrics, Haikou Hospital of the Maternal and Child Health, Haikou, Hainan Province, China
| | - Bin Zhu
- Department of Child Rehabilitation
| | - Yan Lu
- Department of Child Rehabilitation
| | | |
Collapse
|
46
|
Lopez-Charcas O, Pukkanasut P, Velu SE, Brackenbury WJ, Hales TG, Besson P, Gomora JC, Roger S. Pharmacological and nutritional targeting of voltage-gated sodium channels in the treatment of cancers. iScience 2021; 24:102270. [PMID: 33817575 PMCID: PMC8010468 DOI: 10.1016/j.isci.2021.102270] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Voltage-gated sodium (NaV) channels, initially characterized in excitable cells, have been shown to be aberrantly expressed in non-excitable cancer tissues and cells from epithelial origins such as in breast, lung, prostate, colon, and cervix, whereas they are not expressed in cognate non-cancer tissues. Their activity was demonstrated to promote aggressive and invasive potencies of cancer cells, both in vitro and in vivo, whereas their deregulated expression in cancer tissues has been associated with metastatic progression and cancer-related death. This review proposes NaV channels as pharmacological targets for anticancer treatments providing opportunities for repurposing existing NaV-inhibitors or developing new pharmacological and nutritional interventions.
Collapse
Affiliation(s)
- Osbaldo Lopez-Charcas
- Université de Tours, EA4245 Transplantation, Immunologie, Inflammation, Faculté de Médecine de Tours, 10 Boulevard Tonnellé, 37032 Tours, France
| | - Piyasuda Pukkanasut
- Department of Chemistry, The University of Alabama at Birmingham, CHEM 280. 901, 14th Street S, Birmingham, AL 35294, USA
| | - Sadanandan E. Velu
- Department of Chemistry, The University of Alabama at Birmingham, CHEM 280. 901, 14th Street S, Birmingham, AL 35294, USA
| | - William J. Brackenbury
- Department of Biology, York Biomedical Research Institute, University of York, Heslington, York YO10 5DD, UK
| | - Tim G. Hales
- Institute of Academic Anaesthesia, Division of Systems Medicine, School of Medicine, the University of Dundee, DD1 9SY, Dundee, UK
| | - Pierre Besson
- Université de Tours, EA4245 Transplantation, Immunologie, Inflammation, Faculté de Médecine de Tours, 10 Boulevard Tonnellé, 37032 Tours, France
| | - Juan Carlos Gomora
- Instituto de Fisiología Celular, Circuito Exterior s/n Ciudad Universitaria, Universidad Nacional Autónoma de México, Mexico City, 04510 México
| | - Sébastien Roger
- Université de Tours, EA4245 Transplantation, Immunologie, Inflammation, Faculté de Médecine de Tours, 10 Boulevard Tonnellé, 37032 Tours, France
- Institut Universitaire de France, 75005 Paris, France
| |
Collapse
|
47
|
Pasquetti E, Lo Bianco M, Sullo F, Patanè F, Sciuto L, Polizzi A, Praticò AD, Zanghì A, Falsaperla R. SCN1B Gene: A Close Relative to SCN1A. JOURNAL OF PEDIATRIC NEUROLOGY 2021. [DOI: 10.1055/s-0041-1727268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
AbstractOne of the first reported genes associated with epilepsy was SCN1B, which encodes for β-subunit of voltage-gated sodium channel of excitable cells and it is critical for neuronal function in both central and peripheral nervous system. β-subunits modulate the expression levels and functional properties of sodium channels and though their immunoglobulin domains may mediate interactions between channels and other proteins. Traditionally, SCN1B mutations were associated with generalized epilepsy with febrile seizures plus, a familial epilepsy syndrome characterized by heterogeneous phenotypes including febrile seizures (FS), febrile seizures plus (FS + ), mild generalized epilepsies, and severe epileptic encephalopathies. Throughout the years, SCN1B mutations have been also associated with Dravet syndrome and, more recently, with developmental and epileptic encephalopathies, expanding the spectrum associated with this gene mutations to more severe phenotypes.
Collapse
Affiliation(s)
- Elisa Pasquetti
- Pediatrics Postgraduate Residency Program, Section of Pediatrics and Child Neuropsychiatry, Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Manuela Lo Bianco
- Pediatrics Postgraduate Residency Program, Section of Pediatrics and Child Neuropsychiatry, Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Federica Sullo
- Pediatrics Postgraduate Residency Program, Section of Pediatrics and Child Neuropsychiatry, Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Francesca Patanè
- Pediatrics Postgraduate Residency Program, Section of Pediatrics and Child Neuropsychiatry, Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Laura Sciuto
- Pediatrics Postgraduate Residency Program, Section of Pediatrics and Child Neuropsychiatry, Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Agata Polizzi
- Chair of Pediatrics, Department of Educational Sciences, University of Catania, Catania, Italy
| | - Andrea D. Praticò
- Unit of Rare Diseases of the Nervous System in Childhood, Department of Clinical and Experimental Medicine, Section of Pediatrics and Child Neuropsychiatry, University of Catania, Catania, Italy
| | - Antonio Zanghì
- Department of Medical and Surgical Sciences and Advanced Technology “G.F. Ingrassia,” University of Catania, Catania, Italy
| | - Raffaele Falsaperla
- Unit of Pediatrics and Pediatric Emergency, University Hospital “Policlinico Rodolico-San Marco,” Catania, Italy
- Unit of Neonatal Intensive Care and Neonatology, University Hospital “Policlinico Rodolico-San Marco,” Catania, Italy
| |
Collapse
|
48
|
Structural Pharmacology of Voltage-Gated Sodium Channels. J Mol Biol 2021; 433:166967. [PMID: 33794261 DOI: 10.1016/j.jmb.2021.166967] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 03/22/2021] [Accepted: 03/22/2021] [Indexed: 12/19/2022]
Abstract
Voltage-gated sodium (NaV) channels initiate and propagate action potentials in excitable tissues to mediate key physiological processes including heart contraction and nervous system function. Accordingly, NaV channels are major targets for drugs, toxins and disease-causing mutations. Recent breakthroughs in cryo-electron microscopy have led to the visualization of human NaV1.1, NaV1.2, NaV1.4, NaV1.5 and NaV1.7 channel subtypes at high-resolution. These landmark studies have greatly advanced our structural understanding of channel architecture, ion selectivity, voltage-sensing, electromechanical coupling, fast inactivation, and the molecular basis underlying NaV channelopathies. NaV channel structures have also been increasingly determined in complex with toxin and small molecule modulators that target either the pore module or voltage sensor domains. These structural studies have provided new insights into the mechanisms of pharmacological action and opportunities for subtype-selective NaV channel drug design. This review will highlight the structural pharmacology of human NaV channels as well as the potential use of engineered and chimeric channels in future drug discovery efforts.
Collapse
|
49
|
Mantegazza M, Cestèle S, Catterall WA. Sodium channelopathies of skeletal muscle and brain. Physiol Rev 2021; 101:1633-1689. [PMID: 33769100 DOI: 10.1152/physrev.00025.2020] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Voltage-gated sodium channels initiate action potentials in nerve, skeletal muscle, and other electrically excitable cells. Mutations in them cause a wide range of diseases. These channelopathy mutations affect every aspect of sodium channel function, including voltage sensing, voltage-dependent activation, ion conductance, fast and slow inactivation, and both biosynthesis and assembly. Mutations that cause different forms of periodic paralysis in skeletal muscle were discovered first and have provided a template for understanding structure, function, and pathophysiology at the molecular level. More recent work has revealed multiple sodium channelopathies in the brain. Here we review the well-characterized genetics and pathophysiology of the periodic paralyses of skeletal muscle and then use this information as a foundation for advancing our understanding of mutations in the structurally homologous α-subunits of brain sodium channels that cause epilepsy, migraine, autism, and related comorbidities. We include studies based on molecular and structural biology, cell biology and physiology, pharmacology, and mouse genetics. Our review reveals unexpected connections among these different types of sodium channelopathies.
Collapse
Affiliation(s)
- Massimo Mantegazza
- Université Cote d'Azur, Valbonne-Sophia Antipolis, France.,CNRS UMR7275, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne-Sophia Antipolis, France.,INSERM, Valbonne-Sophia Antipolis, France
| | - Sandrine Cestèle
- Université Cote d'Azur, Valbonne-Sophia Antipolis, France.,CNRS UMR7275, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne-Sophia Antipolis, France
| | | |
Collapse
|
50
|
Abstract
The voltage-gated sodium channel α-subunit genes comprise a highly conserved gene family. Mutations of three of these genes, SCN1A, SCN2A and SCN8A, are responsible for a significant burden of neurological disease. Recent progress in identification and functional characterization of patient variants is generating new insights and novel approaches to therapy for these devastating disorders. Here we review the basic elements of sodium channel function that are used to characterize patient variants. We summarize a large body of work using global and conditional mouse mutants to characterize the in vivo roles of these channels. We provide an overview of the neurological disorders associated with mutations of the human genes and examples of the effects of patient mutations on channel function. Finally, we highlight therapeutic interventions that are emerging from new insights into mechanisms of sodium channelopathies.
Collapse
|