1
|
Vecchio D, Macchiaiolo M, Gonfiantini MV, Panfili FM, Petrizzelli F, Liorni N, Cortellessa F, Sinibaldi L, Rana I, Agolini E, Cocciadiferro D, Colantoni N, Semeraro M, Rizzo C, Deodati A, Cotugno N, Caggiano S, Verrillo E, Nucci CG, Alkan S, Saraiva JM, De Sá J, Almeida PM, Krishna J, Buonuomo PS, Martinelli D, Dionisi Vici C, Caputo V, Bartuli A, Novelli A, Mazza T. Widening the infantile hypotonia with psychomotor retardation and characteristic Facies-1 Syndrome's clinical and molecular spectrum through NALCN in-silico structural analysis. Front Genet 2024; 15:1477940. [PMID: 39722796 PMCID: PMC11668739 DOI: 10.3389/fgene.2024.1477940] [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: 08/08/2024] [Accepted: 11/27/2024] [Indexed: 12/28/2024] Open
Abstract
Introduction Infantile hypotonia with psychomotor retardation and characteristic facies-1 (IHPRF1, MIM#615419) is a rare, birth onset, autosomal recessive disorder caused by homozygous or compound heterozygous truncating variants in NALCN gene (MIM#611549) resulting in a loss-of-function effect. Methods We enrolled a new IHPRF1 patients' cohort in the framework of an international multicentric collaboration study. Using specialized in silico pathogenicity predictors and ad hoc structural analyses, we assessed the mechanistic consequences of the deleterious variants retrieved on NALCN structure and function. Results To date 38 different NALCN variants have been retrieved from 33 different families, 26 from unrelated and 22 from related patients. We report on five new IHPRF1 patients from four different families, harboring four newly identified and one previously retrieved variant that exhibited a markedly significant functional impact, thereby compromising the functionality of the protein complex. Discussion By widening the functional spectrum of biallelic variants affecting the NALCN gene, this article broadens the IHPRF1 syndrome's genotype-phenotype correlation and gives new insight into its pathogenic mechanism, diagnosis, and clinical management.
Collapse
Affiliation(s)
- Davide Vecchio
- Rare Diseases and Medical Genetics Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Marina Macchiaiolo
- Rare Diseases and Medical Genetics Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Michaela V. Gonfiantini
- Rare Diseases and Medical Genetics Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Filippo M. Panfili
- Rare Diseases and Medical Genetics Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Francesco Petrizzelli
- Bioinformatics Unit, Fondazione IRCCS Casa Sollievo Della Sofferenza, San Giovanni Rotondo, Italy
| | - Niccolò Liorni
- Bioinformatics Unit, Fondazione IRCCS Casa Sollievo Della Sofferenza, San Giovanni Rotondo, Italy
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Fabiana Cortellessa
- Rare Diseases and Medical Genetics Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Lorenzo Sinibaldi
- Rare Diseases and Medical Genetics Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Ippolita Rana
- Rare Diseases and Medical Genetics Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Emanuele Agolini
- Translational Cytogenomics Research Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Dario Cocciadiferro
- Translational Cytogenomics Research Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Nicole Colantoni
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Michela Semeraro
- Division of Metabolic Diseases, Bambino Gesù Children’s Hospital IRCCS, Rome, Italy
| | - Cristiano Rizzo
- Division of Metabolic Diseases, Bambino Gesù Children’s Hospital IRCCS, Rome, Italy
| | - Annalisa Deodati
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
- Diabetology and Growth Disorders Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Nicola Cotugno
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
- Research Unit of Clinical Immunology and Vaccinology, IRCCS Bambino Gesù Children’s Hospital, Rome, Italy
| | - Serena Caggiano
- Pediatric Pulmonology and Cystic Fibrosis Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Elisabetta Verrillo
- Pediatric Pulmonology and Cystic Fibrosis Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Carlotta G. Nucci
- Neurosurgery Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Serpil Alkan
- Department of Pediatrics, Centre Hospitalier Universitaire, CHU, Liège, Belgium
| | - Jorge M. Saraiva
- Medical Genetics Department, Hospital Pediátrico de Coimbra, Unidade Local de Saúde de Coimbra, Coimbra, Portugal
- University Clinic of Pediatrics, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Clinical Academic Center of Coimbra, Hospital Pediátrico de Coimbra, Unidade Local de Saúde de Coimbra, Coimbra, Portugal
| | - Joaquim De Sá
- Medical Genetics Department, Hospital Pediátrico de Coimbra, Unidade Local de Saúde de Coimbra, Coimbra, Portugal
| | - Pedro M. Almeida
- Medical Genetics Department, Hospital Pediátrico de Coimbra, Unidade Local de Saúde de Coimbra, Coimbra, Portugal
| | - Jayanth Krishna
- Krishna Institute of Medical Sciences (KIMS Hospital), Hyderabad, India
| | - Paola S. Buonuomo
- Rare Diseases and Medical Genetics Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Diego Martinelli
- Division of Metabolic Diseases, Bambino Gesù Children’s Hospital IRCCS, Rome, Italy
| | - Carlo Dionisi Vici
- Division of Metabolic Diseases, Bambino Gesù Children’s Hospital IRCCS, Rome, Italy
| | - Viviana Caputo
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Andrea Bartuli
- Rare Diseases and Medical Genetics Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Antonio Novelli
- Translational Cytogenomics Research Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Tommaso Mazza
- Bioinformatics Unit, Fondazione IRCCS Casa Sollievo Della Sofferenza, San Giovanni Rotondo, Italy
- Translational Cytogenomics Research Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| |
Collapse
|
2
|
Yang Y, Qiu J, Liu J, Zhang D, Ou M, Huang H, Liang P, Zhu T, Zhou C. Sodium leak channels in the central amygdala modulate the analgesic potency of volatile anaesthetics in mice. Br J Anaesth 2024; 133:983-997. [PMID: 39322470 DOI: 10.1016/j.bja.2024.06.049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 06/12/2024] [Accepted: 06/23/2024] [Indexed: 09/27/2024] Open
Abstract
BACKGROUND Analgesia is an important effect of volatile anaesthetics, for which the spinal cord is a critical neural target. However, how supraspinal mechanisms modulate analgesic potency of volatile anaesthetics is not clear. We investigated the contribution of the central amygdala (CeA) to the analgesic effects of isoflurane and sevoflurane. METHODS Analgesic potencies of volatile anaesthetics were tested during optogenetic and chemogenetic inhibition of CeA neurones. In vivo calcium imaging was used to measure neuronal activities of CeA neuronal subtypes under volatile anaesthesia. Contributions of the sodium leak channel (NALCN) in GABAergic CeA (CeAGABA) neurones to analgesic effects of volatile anaesthetics were explored by specific NALCN knockdown. Electrophysiological recordings on acute brain slices were applied to measure volatile anaesthetic modulation of CeA neuronal activity by NALCN. RESULTS Optogenetic or chemogenetic silencing CeA neurones reduced the analgesic effects of isoflurane or sevoflurane in vivo. The calcium signals of CeAGABA neurones increased during exposure to isoflurane or sevoflurane at analgesic concentrations. Knockdown of NALCN in CeAGABA neurones attenuated antinociceptive effects of isoflurane, sevoflurane, or both. For example, mean concentrations of isoflurane, sevoflurane, or both that induced immobility to tail-flick stimuli were significantly increased (isoflurane: 1.17 [0.05] vol% vs 1.24 [0.04] vol%, P=0.01; sevoflurane: 2.65 [0.07] vol% vs 2.81 [0.07] vol%; P<0.001). In brain slices, isoflurane, sevoflurane, or both at clinical concentrations increased NALCN-mediated holding currents and conductance in CeAGABA neurones, which increased excitability of CeAGABA neurones in an NALCN-dependent manner. CONCLUSIONS The analgesic potencies of volatile anaesthetics are partially mediated by modulation of NALCN in CeAGABA neurones.
Collapse
Affiliation(s)
- Yaoxin Yang
- Department of Anaesthesiology, West China Hospital of Sichuan University, Chengdu, Sichuan Province, China; Research Center of Anaesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Jingxuan Qiu
- Department of Anaesthesiology, West China Hospital of Sichuan University, Chengdu, Sichuan Province, China; Research Center of Anaesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Jin Liu
- Department of Anaesthesiology, West China Hospital of Sichuan University, Chengdu, Sichuan Province, China; Research Center of Anaesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Donghang Zhang
- Department of Anaesthesiology, West China Hospital of Sichuan University, Chengdu, Sichuan Province, China; Research Center of Anaesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Mengchan Ou
- Department of Anaesthesiology, West China Hospital of Sichuan University, Chengdu, Sichuan Province, China
| | - Han Huang
- Department of Anaesthesiology & Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital of Sichuan University, Chengdu, Sichuan Province, China
| | - Peng Liang
- Department of Anaesthesiology, West China Hospital of Sichuan University, Chengdu, Sichuan Province, China
| | - Tao Zhu
- Department of Anaesthesiology, West China Hospital of Sichuan University, Chengdu, Sichuan Province, China.
| | - Cheng Zhou
- Research Center of Anaesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
| |
Collapse
|
3
|
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
|
4
|
Chen Y, Xia X, Zhang Y, Gao L, He C, Cao J. Case Report: New presentation of CLIFAHDD syndrome with a novel variant in the NALCN gene and a literature review. Front Pediatr 2024; 12:1370790. [PMID: 38873579 PMCID: PMC11169720 DOI: 10.3389/fped.2024.1370790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 05/08/2024] [Indexed: 06/15/2024] Open
Abstract
Background Congenital contractures of the limbs and face, hypotonia, and developmental delay (CLIFAHDD) syndrome (OMIM #616266) is an autosomal dominant hereditary disease that can lead to the congenital contracture of the limbs and face, hypotonia, and developmental delay. In addition, it may result in growth retardation and present various clinical symptoms, such as brain atrophy, a small pituitary gland, musculoskeletal abnormalities, abnormal breathing, abdominal hernia, and abnormal facial features. Herein, we describe a novel de novo missense genetic variant in the sodium leak channel, non-selective (NALCN) gene that is associated with CLIFAHDD syndrome. Case description This study describes a patient with varus deformities in both feet, deviation of the ulnar side of the fingers, and severe hypotonia. This patient was subsequently confirmed to have CLIFAHDD syndrome through genetic testing, which also revealed a novel missense de novo genetic variant in the NALCN gene (c.3553G > A, p.Ala1185Thr). Conclusions Our findings further enrich the known variant spectrum of the NALCN gene and may expand the range of clinical options for treating NALCN-related disorders.
Collapse
Affiliation(s)
| | | | | | | | | | - Jianguo Cao
- Department of Rehabilitation Medicine, Shenzhen Children’s Hospital, Shenzhen, China
| |
Collapse
|
5
|
Schott K, Usher SG, Serra O, Carnevale V, Pless SA, Chua HC. Unplugging lateral fenestrations of NALCN reveals a hidden drug binding site within the pore region. Proc Natl Acad Sci U S A 2024; 121:e2401591121. [PMID: 38787877 PMCID: PMC11145269 DOI: 10.1073/pnas.2401591121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 04/09/2024] [Indexed: 05/26/2024] Open
Abstract
The sodium (Na+) leak channel (NALCN) is a member of the four-domain voltage-gated cation channel family that includes the prototypical voltage-gated sodium and calcium channels (NaVs and CaVs, respectively). Unlike NaVs and CaVs, which have four lateral fenestrations that serve as routes for lipophilic compounds to enter the central cavity to modulate channel function, NALCN has bulky residues (W311, L588, M1145, and Y1436) that block these openings. Structural data suggest that occluded fenestrations underlie the pharmacological resistance of NALCN, but functional evidence is lacking. To test this hypothesis, we unplugged the fenestrations of NALCN by substituting the four aforementioned residues with alanine (AAAA) and compared the effects of NaV, CaV, and NALCN blockers on both wild-type (WT) and AAAA channels. Most compounds behaved in a similar manner on both channels, but phenytoin and 2-aminoethoxydiphenyl borate (2-APB) elicited additional, distinct responses on AAAA channels. Further experiments using single alanine mutants revealed that phenytoin and 2-APB enter the inner cavity through distinct fenestrations, implying structural specificity to their modes of access. Using a combination of computational and functional approaches, we identified amino acid residues critical for 2-APB activity, supporting the existence of drug binding site(s) within the pore region. Intrigued by the activity of 2-APB and its analogues, we tested compounds containing the diphenylmethane/amine moiety on WT channels. We identified clinically used drugs that exhibited diverse activity, thus expanding the pharmacological toolbox for NALCN. While the low potencies of active compounds reiterate the pharmacological resistance of NALCN, our findings lay the foundation for rational drug design to develop NALCN modulators with refined properties.
Collapse
Affiliation(s)
- Katharina Schott
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen2100, Denmark
| | - Samuel George Usher
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen2100, Denmark
| | - Oscar Serra
- Department of Biology, Temple University, Philadelphia, PA19122
- Institute for Genomics and Evolutionary Medicine, Temple University, Philadelphia, PA19122
- Institute of Computational Molecular Science, Temple University, Philadelphia, PA19122
| | - Vincenzo Carnevale
- Department of Biology, Temple University, Philadelphia, PA19122
- Institute for Genomics and Evolutionary Medicine, Temple University, Philadelphia, PA19122
- Institute of Computational Molecular Science, Temple University, Philadelphia, PA19122
| | - Stephan Alexander Pless
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen2100, Denmark
| | - Han Chow Chua
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen2100, Denmark
| |
Collapse
|
6
|
Du S, Sun S, Ju Z, Wang W, Su K, Qiu F, Yu X, Xu G, Yuan D. Hierarchical Self-Assembly of Capsule-Shaped Zirconium Coordination Cages with Quaternary Structure. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308445. [PMID: 38229156 PMCID: PMC10953209 DOI: 10.1002/advs.202308445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 01/07/2024] [Indexed: 01/18/2024]
Abstract
Biological macromolecules exhibit emergent functions through hierarchical self-assembly, a concept that is extended to design artificial supramolecular assemblies. Here, the first example of breaking the common parallel arrangement of capsule-shaped zirconium coordination cages is reported by constructing the hierarchical porous framework ZrR-1. ZrR-1 adopts a quaternary structure resembling protein and contains 12-connected chloride clusters, representing the highest connectivity for zirconium-based cages reported thus far. Compared to the parallel framework ZrR-2, ZrR-1 demonstrated enhanced stability in acidic aqueous solutions and a tenfold increase in BET surface area (879 m2 g-1 ). ZrR-1 also exhibits excellent proton conductivity, reaching 1.31 × 10-2 S·cm-1 at 353 K and 98% relative humidity, with a low activation energy of 0.143 eV. This finding provides insights into controlling the hierarchical self-assembly of metal-organic cages to discover superstructures with emergent properties.
Collapse
Affiliation(s)
- Shunfu Du
- State Key Laboratory of Structural ChemistryFujian Provincial Key Laboratory of Materials and Techniques toward Hydrogen EnergyFujian Institute of Research on the Structure of MatterThe Chinese Academy of SciencesFuzhouFujian350108P. R. China
- University of the Chinese Academy of SciencesBeijing100049P. R. China
| | - Shihao Sun
- State Key Laboratory of Structural ChemistryFujian Provincial Key Laboratory of Materials and Techniques toward Hydrogen EnergyFujian Institute of Research on the Structure of MatterThe Chinese Academy of SciencesFuzhouFujian350108P. R. China
| | - Zhanfeng Ju
- State Key Laboratory of Structural ChemistryFujian Provincial Key Laboratory of Materials and Techniques toward Hydrogen EnergyFujian Institute of Research on the Structure of MatterThe Chinese Academy of SciencesFuzhouFujian350108P. R. China
- University of the Chinese Academy of SciencesBeijing100049P. R. China
| | - Wenjing Wang
- State Key Laboratory of Structural ChemistryFujian Provincial Key Laboratory of Materials and Techniques toward Hydrogen EnergyFujian Institute of Research on the Structure of MatterThe Chinese Academy of SciencesFuzhouFujian350108P. R. China
- University of the Chinese Academy of SciencesBeijing100049P. R. China
| | - Kongzhao Su
- State Key Laboratory of Structural ChemistryFujian Provincial Key Laboratory of Materials and Techniques toward Hydrogen EnergyFujian Institute of Research on the Structure of MatterThe Chinese Academy of SciencesFuzhouFujian350108P. R. China
- University of the Chinese Academy of SciencesBeijing100049P. R. China
| | - Fenglei Qiu
- State Key Laboratory of Structural ChemistryFujian Provincial Key Laboratory of Materials and Techniques toward Hydrogen EnergyFujian Institute of Research on the Structure of MatterThe Chinese Academy of SciencesFuzhouFujian350108P. R. China
- College of ChemistryFuzhou UniversityFuzhou350108P. R. China
| | - Xuying Yu
- State Key Laboratory of Structural ChemistryFujian Provincial Key Laboratory of Materials and Techniques toward Hydrogen EnergyFujian Institute of Research on the Structure of MatterThe Chinese Academy of SciencesFuzhouFujian350108P. R. China
- University of the Chinese Academy of SciencesBeijing100049P. R. China
| | - Gang Xu
- State Key Laboratory of Structural ChemistryFujian Provincial Key Laboratory of Materials and Techniques toward Hydrogen EnergyFujian Institute of Research on the Structure of MatterThe Chinese Academy of SciencesFuzhouFujian350108P. R. China
- University of the Chinese Academy of SciencesBeijing100049P. R. China
| | - Daqiang Yuan
- State Key Laboratory of Structural ChemistryFujian Provincial Key Laboratory of Materials and Techniques toward Hydrogen EnergyFujian Institute of Research on the Structure of MatterThe Chinese Academy of SciencesFuzhouFujian350108P. R. China
- University of the Chinese Academy of SciencesBeijing100049P. R. China
| |
Collapse
|
7
|
Schott K, Usher SG, Serra O, Carnevale V, Pless SA, Chua HC. Unplugging lateral fenestrations of NALCN reveals a hidden drug binding site within the pore module. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.04.12.536537. [PMID: 38328210 PMCID: PMC10849497 DOI: 10.1101/2023.04.12.536537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
The sodium (Na + ) leak channel (NALCN) is a member of the four-domain voltage-gated cation channel family that includes the prototypical voltage-gated sodium and calcium channels (Na V s and Ca V s, respectively). Unlike Na V s and Ca V s, which have four lateral fenestrations that serve as routes for lipophilic compounds to enter the central cavity to modulate channel function, NALCN has bulky residues (W311, L588, M1145 and Y1436) that block these openings. Structural data suggest that oc-cluded lateral fenestrations underlie the pharmacological resistance of NALCN to lipophilic compounds, but functional evidence is lacking. To test this hypothesis, we unplugged the fenestrations of NALCN by substituting the four aforementioned resi-dues with alanine (AAAA) and compared the effects of Na V , Ca V and NALCN block-ers on both wild-type (WT) and AAAA channels. Most compounds behaved in a simi-lar manner on both channels, but phenytoin and 2-aminoethoxydiphenyl borate (2-APB) elicited additional, distinct responses on AAAA channels. Further experiments using single alanine mutants revealed that phenytoin and 2-APB enter the inner cav-ity through distinct fenestrations, implying structural specificity to their modes of ac-cess. Using a combination of computational and functional approaches, we identified amino acid residues critical for 2-APB activity, supporting the existence of drug bind-ing site(s) within the pore region. Intrigued by the activity of 2-APB and its ana-logues, we tested additional compounds containing the diphenylmethane/amine moiety on WT channels. We identified compounds from existing clinically used drugs that exhibited diverse activity, thus expanding the pharmacological toolbox for NALCN. While the low potencies of active compounds reiterate the resistance of NALCN to pharmacological targeting, our findings lay the foundation for rational drug design to develop NALCN modulators with refined properties. Significance statement The sodium leak channel (NALCN) is essential for survival: mutations cause life-threatening developmental disorders in humans. However, no treatment is currently available due to the resistance of NALCN to pharmacological targeting. One likely reason is that the lateral fenestrations, a common route for clinically used drugs to enter and block related ion channels, are occluded in NALCN. Using a combination of computational and functional approaches, we unplugged the fenestrations of NALCN which led us to the first molecularly defined drug binding site within the pore region. Besides that, we also identified additional NALCN modulators from existing clinically used therapeutics, thus expanding the pharmacological toolbox for this leak channel.
Collapse
|
8
|
Zhang D, Wei Y. Role of sodium leak channel (NALCN) in sensation and pain: an overview. Front Pharmacol 2024; 14:1349438. [PMID: 38273833 PMCID: PMC10808581 DOI: 10.3389/fphar.2023.1349438] [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: 12/04/2023] [Accepted: 12/29/2023] [Indexed: 01/27/2024] Open
Abstract
The sodium leak channel (NALCN) is widely expressed in the central nervous system and plays a pivotal role in regulating the resting membrane potential (RMP) by mediating the Na+ leak current. NALCN was first reported in 1999, and since then, increasing evidence has provided insights into the structure and functions of NALCN. As an essential component of neuronal background currents, NALCN has been shown to be involved in many important physiological functions, particularly in the respiratory rhythm, as NALCN mutant mice have a severely disrupted respiratory rhythm and die within 24 h of birth. Many patients with NALCN mutations also develop serious clinical syndromes, such as severe hypotonia, speech impairment, and cognitive delay. Recently, emerging studies have clarified the human NALCN structure and revealed additional properties and functions of NALCN. For instance, accumulating evidence highlights that the NALCN is involved in normal sensation and pain. Here, we review the current literature and summarize the role of the NALCN in sensation and pain.
Collapse
Affiliation(s)
- Donghang Zhang
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Yiyong Wei
- Department of Anesthesiology, Longgang District Maternity and Child Healthcare Hospital of Shenzhen City (Longgang Maternity and Child Institute of Shantou University Medical College), Shenzhen, China
| |
Collapse
|
9
|
Ngodup T, Irie T, Elkins SP, Trussell LO. The Na + leak channel NALCN controls spontaneous activity and mediates synaptic modulation by α2-adrenergic receptors in auditory neurons. eLife 2024; 12:RP89520. [PMID: 38197879 PMCID: PMC10945507 DOI: 10.7554/elife.89520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2024] Open
Abstract
Cartwheel interneurons of the dorsal cochlear nucleus (DCN) potently suppress multisensory signals that converge with primary auditory afferent input, and thus regulate auditory processing. Noradrenergic fibers from locus coeruleus project to the DCN, and α2-adrenergic receptors inhibit spontaneous spike activity but simultaneously enhance synaptic strength in cartwheel cells, a dual effect leading to enhanced signal-to-noise for inhibition. However, the ionic mechanism of this striking modulation is unknown. We generated a glycinergic neuron-specific knockout of the Na+ leak channel NALCN in mice and found that its presence was required for spontaneous firing in cartwheel cells. Activation of α2-adrenergic receptors inhibited both NALCN and spike generation, and this modulation was absent in the NALCN knockout. Moreover, α2-dependent enhancement of synaptic strength was also absent in the knockout. GABAB receptors mediated inhibition through NALCN as well, acting on the same population of channels as α2 receptors, suggesting close apposition of both receptor subtypes with NALCN. Thus, multiple neuromodulatory systems determine the impact of synaptic inhibition by suppressing the excitatory leak channel, NALCN.
Collapse
Affiliation(s)
- Tenzin Ngodup
- Oregon Hearing Research Center and Vollum Institute, Oregon Health & Science UniversityPortlandUnited States
| | - Tomohiko Irie
- Department of Physiology, Kitasato University School of MedicineSagamiharaJapan
| | - Seán P Elkins
- Oregon Hearing Research Center and Vollum Institute, Oregon Health & Science UniversityPortlandUnited States
| | - Laurence O Trussell
- Oregon Hearing Research Center and Vollum Institute, Oregon Health & Science UniversityPortlandUnited States
| |
Collapse
|
10
|
Monteil A, Guérineau NC, Gil-Nagel A, Parra-Diaz P, Lory P, Senatore A. New insights into the physiology and pathophysiology of the atypical sodium leak channel NALCN. Physiol Rev 2024; 104:399-472. [PMID: 37615954 DOI: 10.1152/physrev.00014.2022] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 07/13/2023] [Accepted: 08/15/2023] [Indexed: 08/25/2023] Open
Abstract
Cell excitability and its modulation by hormones and neurotransmitters involve the concerted action of a large repertoire of membrane proteins, especially ion channels. Unique complements of coexpressed ion channels are exquisitely balanced against each other in different excitable cell types, establishing distinct electrical properties that are tailored for diverse physiological contributions, and dysfunction of any component may induce a disease state. A crucial parameter controlling cell excitability is the resting membrane potential (RMP) set by extra- and intracellular concentrations of ions, mainly Na+, K+, and Cl-, and their passive permeation across the cell membrane through leak ion channels. Indeed, dysregulation of RMP causes significant effects on cellular excitability. This review describes the molecular and physiological properties of the Na+ leak channel NALCN, which associates with its accessory subunits UNC-79, UNC-80, and NLF-1/FAM155 to conduct depolarizing background Na+ currents in various excitable cell types, especially neurons. Studies of animal models clearly demonstrate that NALCN contributes to fundamental physiological processes in the nervous system including the control of respiratory rhythm, circadian rhythm, sleep, and locomotor behavior. Furthermore, dysfunction of NALCN and its subunits is associated with severe pathological states in humans. The critical involvement of NALCN in physiology is now well established, but its study has been hampered by the lack of specific drugs that can block or agonize NALCN currents in vitro and in vivo. Molecular tools and animal models are now available to accelerate our understanding of how NALCN contributes to key physiological functions and the development of novel therapies for NALCN channelopathies.
Collapse
Affiliation(s)
- Arnaud Monteil
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
- LabEx "Ion Channel Science and Therapeutics," Montpellier, France
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Nathalie C Guérineau
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
- LabEx "Ion Channel Science and Therapeutics," Montpellier, France
| | - Antonio Gil-Nagel
- Department of Neurology, Epilepsy Program, Hospital Ruber Internacional, Madrid, Spain
| | - Paloma Parra-Diaz
- Department of Neurology, Epilepsy Program, Hospital Ruber Internacional, Madrid, Spain
| | - Philippe Lory
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
- LabEx "Ion Channel Science and Therapeutics," Montpellier, France
| | - Adriano Senatore
- Department of Biology, University of Toronto Mississauga, Mississauga, Ontario, Canada
| |
Collapse
|
11
|
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
|
12
|
Tehrani Fateh S, Bagheri S, Sadeghi H, Salehpour S, Fazeli Bavandpour F, Sadeghi B, Jamshidi S, Tonekaboni SH, Mirfakhraie R, Miryounesi M, Ghasemi MR. Extending and outlining the genotypic and phenotypic spectrum of novel mutations of NALCN gene in IHPRF1 syndrome: identifying recurrent urinary tract infection. Neurol Sci 2023; 44:4491-4498. [PMID: 37452996 DOI: 10.1007/s10072-023-06960-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 07/10/2023] [Indexed: 07/18/2023]
Abstract
Infantile hypotonia with psychomotor retardation and characteristic facies 1 (IHPRF1) is caused by biallelic mutations in the NALCN gene, the major ion channel responsible for the background Na + conduction in neurons. Through whole-exome sequencing (WES), we report three novel homozygous variants in three families, including c.1434 + 1G > A, c.3269G > A, and c.2648G > T, which are confirmed and segregated by Sanger sequencing. Consequently, intron 12's highly conserved splice donor location is disrupted by the pathogenic c.1434 + 1G > A variation, most likely causing the protein to degrade through nonsense-mediated decay (NMD). Subsequently, a premature stop codon is thus generated at amino acid 1090 of the protein as a result of the pathogenic c.3269G > A; p.W1090* variation, resulting in NMD or truncated protein production. Lastly, the missense mutation c.2648G > T; p.G883V can play a critical role in the interplay of functional domains. This study introduces recurrent urinary tract infections for the first time, broadening the phenotypic range of IHPRF1 syndrome in addition to the genotypic spectrum. This trait may result from insufficient bladder emptying, which may be related to the NALCN channelosome's function in background Na + conduction. This work advances knowledge about the molecular genetic underpinnings of IHPRF1 and introduces a novel phenotype through the widespread use of whole exome sequencing.
Collapse
Affiliation(s)
- Sahand Tehrani Fateh
- School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
- Center for Comprehensive Genetic Services, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Saman Bagheri
- Center for Comprehensive Genetic Services, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hossein Sadeghi
- Genomic Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shadab Salehpour
- Department of Pediatrics, Clinical Research Development Unit, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Behnia Sadeghi
- Center for Comprehensive Genetic Services, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sanaz Jamshidi
- Center for Comprehensive Genetic Services, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed Hassan Tonekaboni
- Department of Pediatric Neurology, School of Medicine, Pediatric Neurology Research Center, Mofid Children's Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Reza Mirfakhraie
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Miryounesi
- Center for Comprehensive Genetic Services, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Mohammad-Reza Ghasemi
- Center for Comprehensive Genetic Services, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
13
|
Ngodup T, Irie T, Elkins S, Trussell LO. The Na + leak channel NALCN controls spontaneous activity and mediates synaptic modulation by α2-adrenergic receptors in auditory neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.23.546323. [PMID: 37987013 PMCID: PMC10659375 DOI: 10.1101/2023.06.23.546323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Cartwheel interneurons of the dorsal cochlear nucleus (DCN) potently suppress multisensory signals that converge with primary auditory afferent input, and thus regulate auditory processing. Noradrenergic fibers from locus coeruleus project to the DCN, and α2-adrenergic receptors inhibit spontaneous spike activity but simultaneously enhance synaptic strength in cartwheel cells, a dual effect leading to enhanced signal-to-noise for inhibition. However, the ionic mechanism of this striking modulation is unknown. We generated a glycinergic neuron-specific knockout of the Na+ leak channel NALCN, and found that its presence was required for spontaneous firing in cartwheel cells. Activation of α2-adrenergic receptors inhibited both NALCN and spike generation, and this modulation was absent in the NALCN knockout. Moreover, α2-dependent enhancement of synaptic strength was also absent in the knockout. GABAB receptors mediated inhibition through NALCN as well, acting on the same population of channels as α2 receptors, suggesting close apposition of both receptor subtypes with NALCN. Thus, multiple neuromodulatory systems determine the impact of synaptic inhibition by suppressing the excitatory leak channel, NALCN.
Collapse
Affiliation(s)
- Tenzin Ngodup
- Oregon Hearing Research Center and Vollum Institute, Oregon Health & Science University, Portland OR USA
| | - Tomohiko Irie
- Department of Physiology, Kitasato University School of Medicine, Sagamihara, Japan
| | - Sean Elkins
- Oregon Hearing Research Center and Vollum Institute, Oregon Health & Science University, Portland OR USA
| | - Laurence O Trussell
- Oregon Hearing Research Center and Vollum Institute, Oregon Health & Science University, Portland OR USA
| |
Collapse
|
14
|
Horimoto AR, Boyken LA, Blue EE, Grinde KE, Nafikov RA, Sohi HK, Nato AQ, Bis JC, Brusco LI, Morelli L, Ramirez A, Dalmasso MC, Temple S, Satizabal C, Browning SR, Seshadri S, Wijsman EM, Thornton TA. Admixture mapping implicates 13q33.3 as ancestry-of-origin locus for Alzheimer disease in Hispanic and Latino populations. HGG ADVANCES 2023; 4:100207. [PMID: 37333771 PMCID: PMC10276158 DOI: 10.1016/j.xhgg.2023.100207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 05/16/2023] [Indexed: 06/20/2023] Open
Abstract
Alzheimer disease (AD) is the most common form of senile dementia, with high incidence late in life in many populations including Caribbean Hispanic (CH) populations. Such admixed populations, descended from more than one ancestral population, can present challenges for genetic studies, including limited sample sizes and unique analytical constraints. Therefore, CH populations and other admixed populations have not been well represented in studies of AD, and much of the genetic variation contributing to AD risk in these populations remains unknown. Here, we conduct genome-wide analysis of AD in multiplex CH families from the Alzheimer Disease Sequencing Project (ADSP). We developed, validated, and applied an implementation of a logistic mixed model for admixture mapping with binary traits that leverages genetic ancestry to identify ancestry-of-origin loci contributing to AD. We identified three loci on chromosome 13q33.3 associated with reduced risk of AD, where associations were driven by Native American (NAM) ancestry. This AD admixture mapping signal spans the FAM155A, ABHD13, TNFSF13B, LIG4, and MYO16 genes and was supported by evidence for association in an independent sample from the Alzheimer's Genetics in Argentina-Alzheimer Argentina consortium (AGA-ALZAR) study with considerable NAM ancestry. We also provide evidence of NAM haplotypes and key variants within 13q33.3 that segregate with AD in the ADSP whole-genome sequencing data. Interestingly, the widely used genome-wide association study approach failed to identify associations in this region. Our findings underscore the potential of leveraging genetic ancestry diversity in recently admixed populations to improve genetic mapping, in this case for AD-relevant loci.
Collapse
Affiliation(s)
| | - Lisa A. Boyken
- Department of Biostatistics, University of Washington, Seattle, WA 98195, USA
| | - Elizabeth E. Blue
- Division of Medical Genetics/Department of Medicine, University of Washington, Seattle, WA 98195, USA
- Brotman Baty Institute for Precision Medicine, Seattle, WA 98195, USA
| | - Kelsey E. Grinde
- Department of Biostatistics, University of Washington, Seattle, WA 98195, USA
- Department of Mathematics, Statistics and Computer Science, Macalester College, Saint Paul, MN 55105, USA
| | - Rafael A. Nafikov
- Department of Biostatistics, University of Washington, Seattle, WA 98195, USA
- Division of Medical Genetics/Department of Medicine, University of Washington, Seattle, WA 98195, USA
| | - Harkirat K. Sohi
- Division of Medical Genetics/Department of Medicine, University of Washington, Seattle, WA 98195, USA
- Biomedical and Health Informatics Program, University of Washington, Seattle, WA 98195, USA
| | - Alejandro Q. Nato
- Division of Medical Genetics/Department of Medicine, University of Washington, Seattle, WA 98195, USA
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA
| | - Joshua C. Bis
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA 98101, USA
| | - Luis I. Brusco
- CENECON - Center of Behavioural Neurology and Neuropsychiatry, School of Medicine, University of Buenos Aires, C1121A6B Buenos Aires, Argentina
| | - Laura Morelli
- Laboratory of Brain Aging and Neurodegeneration-Fundación Instituto Leloir-IIBBA- National Scientific and Technical Research Council (CONICET), C1405BWE Ciudad Autónoma de Buenos Aires, Argentina
| | - Alfredo Ramirez
- Division of Neurogenetics and Molecular Psychiatry, Department of Psychiatry and Psychotherapy, University of Cologne, Medical Faculty, 50937 Cologne, Germany
- Department of Neurodegeneration and Gerontopsychiatry, University of Bonn, 53127 Bonn, Germany
- German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany
- Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) University of Cologne, 50674 Cologne, Germany
- Department of Psychiatry, UT Health San Antonio, San Antonio, TX 78229, USA
- Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, UT Health San Antonio, San Antonio, TX 78229, USA
| | - Maria Carolina Dalmasso
- Division of Neurogenetics and Molecular Psychiatry, Department of Psychiatry and Psychotherapy, University of Cologne, Medical Faculty, 50937 Cologne, Germany
- Neurosciences and Complex Systems Unit (EnyS), CONICET, Hospital El Cruce, National University A. Jauretche (UNAJ), B1888AAE Florencio Varela, Argentina
| | - Seth Temple
- Department of Statistics, University of Washington, Seattle, WA 98195, USA
| | - Claudia Satizabal
- Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, UT Health San Antonio, San Antonio, TX 78229, USA
- Department of Population Health Sciences, University of Texas, San Antonio, TX 78229, USA
- Department of Neurology, University of Texas, San Antonio, TX 78229, USA
| | - Sharon R. Browning
- Department of Biostatistics, University of Washington, Seattle, WA 98195, USA
| | - Sudha Seshadri
- Department of Neurology, University of Texas, San Antonio, TX 78229, USA
| | - Ellen M. Wijsman
- Department of Biostatistics, University of Washington, Seattle, WA 98195, USA
- Division of Medical Genetics/Department of Medicine, University of Washington, Seattle, WA 98195, USA
| | - Timothy A. Thornton
- Department of Biostatistics, University of Washington, Seattle, WA 98195, USA
- Department of Statistics, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
15
|
Hashemi B, Huntsman RJ, Li H, Zhang D, Xi Y. New presentation of CLIFAHDD syndrome with a novel variant in NALCN gene: A report of a rare case. Clin Case Rep 2023; 11:e7647. [PMID: 37469362 PMCID: PMC10352546 DOI: 10.1002/ccr3.7647] [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: 02/06/2023] [Revised: 06/09/2023] [Accepted: 06/17/2023] [Indexed: 07/21/2023] Open
Abstract
Key Clinical Message Congenital Contractures of Limbs and Face, Hypotonia, and Developmental Delay (CLIFAHDD) syndrome is a recently described type of distal arthrogryposis which unlike other subtypes is associated with developmental delay and various neurologic presentation. Epilepsy and ataxia have been reported. We add paroxysmal dyskinesia to the clinical spectrum. Understanding the molecular mechanism can help developing targeted therapy in future. Abstract This study resulted in identification of a novel variant in NALCN gene leading to autosomal dominant CLIFAHDD syndrome. Our patient presented with a form of nonepileptic paroxysmal dyskinesia. This is a new phenotype that has not been described previously.
Collapse
Affiliation(s)
- Bita Hashemi
- Department of Pediatrics, Division of Medical GeneticsUniversity of SaskatchewanSaskatoonSaskatchewanCanada
| | - Richard J. Huntsman
- Department of Pediatrics, Division of NeurologyUniversity of SaskatchewanSaskatoonSaskatchewanCanada
| | - Huan Li
- Department of Biology, College of Arts and SciencesSaint Louis UniversitySaint LouisMissouriUSA
| | - Dapeng Zhang
- Department of Biology, College of Arts and SciencesSaint Louis UniversitySaint LouisMissouriUSA
- Program of Bioinformatics and Computational Biology, College of Arts and SciencesSaint Louis UniversitySaint LouisMissouriUSA
| | - Yanwei Xi
- Department of Pathology and Laboratory Medicine, Genomics LaboratoryUniversity of SaskatchewanSaskatoonSaskatchewanCanada
| |
Collapse
|
16
|
He J, Xu J, Chang Z, Yan J, Zhang L, Qin Y. NALCN is a potential biomarker and therapeutic target in human cancers. Front Genet 2023; 14:1164707. [PMID: 37152978 PMCID: PMC10154523 DOI: 10.3389/fgene.2023.1164707] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 04/10/2023] [Indexed: 05/09/2023] Open
Abstract
Background: Sodium leak channel non-selective (NALCN), known as a voltage-independent Na+ channel, is increasingly considered to play vital roles in tumorigenesis and metastasis of human cancers. However, no comprehensive pan-cancer analysis of NALCN has been conducted. Our study aims to explore the potential diagnostic, prognostic and therapeutic value of NALCN in human cancers. Methods: Through comprehensive application of datasets from Human Protein Atlas (HPA), The Cancer Genome Atlas (TCGA), Cancer Cell Line Encyclopedia (CCLE), Enhanced Version of Tumor Immune Estimation Resource (TIMER2.0), Tumor and Immune System Interaction Database (TISIDB), The University of Alabama at Birmingham Cancer data analysis Portal (UALCAN), cBioPortal, GeneMANIA and Search Tool for the Retrieval of Interaction Gene/Proteins (STRING) databases, we explored the potential roles of NALCN in different cancers. The differential expression, prognostic implications, pathological stages and grades, molecular and immune subtypes, diagnostic accuracy, tumor mutation burden (TMB), microsatellite instability (MSI), mismatch repair (MMR) genes, immune checkpoint genes, chemokine genes, major histocompatibility complex (MHC)-related genes, tumor-infiltrating immune cells (TIICs), promoter methylation, mutations, copy number alteration (CNA), and functional enrichment related to NALCN were analyzed. Results: Most cancers lowly expressed NALCN. Upregulated NALCN expression was associated with poor or better prognosis in different cancers. Moreover, NALCN was correlated with clinicopathological features in multiple cancers. NALCN showed high diagnostic accuracy in 5 caner types. NALCN is highly linked with immune-related biomarkers, immune-related genes and TIICs. Significant methylation changes and genetic alteration of NALCN can be observed in many cancers. Enrichment analysis showed that NALCN is closely related to multiple tumor-related signaling pathways. Conclusion: Our study revealed the vital involvement of NALCN in cancer. NALCN can be used as a prognostic biomarker for immune infiltration and clinical outcomes, and has potential diagnostic and therapeutic implications.
Collapse
Affiliation(s)
- Jian He
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jie Xu
- School of Public Health, Xinxiang Medical University, Xinxiang, China
| | - Zhiwei Chang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jiaqin Yan
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Limin Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yanru Qin
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- *Correspondence: Yanru Qin,
| |
Collapse
|
17
|
Abstract
The CACNA1C gene encodes the pore-forming subunit of the CaV1.2 L-type Ca2+ channel, a critical component of membrane physiology in multiple tissues, including the heart, brain, and immune system. As such, mutations altering the function of these channels have the potential to impact a wide array of cellular functions. The first mutations identified within CACNA1C were shown to cause a severe, multisystem disorder known as Timothy syndrome (TS), which is characterized by neurodevelopmental deficits, long-QT syndrome, life-threatening cardiac arrhythmias, craniofacial abnormalities, and immune deficits. Since this initial description, the number and variety of disease-associated mutations identified in CACNA1C have grown tremendously, expanding the range of phenotypes observed in affected patients. CACNA1C channelopathies are now known to encompass multisystem phenotypes as described in TS, as well as more selective phenotypes where patients may exhibit predominantly cardiac or neurological symptoms. Here, we review the impact of genetic mutations on CaV1.2 function and the resultant physiological consequences.
Collapse
Affiliation(s)
- Kevin G Herold
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - John W Hussey
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Ivy E Dick
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
18
|
Kim K, Lee J, Lee JY, Yong SH, Kim EY, Jung JY, Kang YA, Park MS, Kim YS, Oh CM, Lee SH. Clinical features and molecular genetics associated with brain metastasis in suspected early-stage non-small cell lung cancer. Front Oncol 2023; 13:1148475. [PMID: 37139160 PMCID: PMC10150586 DOI: 10.3389/fonc.2023.1148475] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 04/03/2023] [Indexed: 05/05/2023] Open
Abstract
Introduction Regarding whether brain magnetic resonance imaging (MRI) should be routine in patients with suspected early-stage lung cancer, guideline recommendations are inconsistent. Therefore, we performed this study to evaluate the incidence of and risk factors for brain metastasis (BM) in patients with suspected early-stage non-small-cell lung cancer (NSCLC). Methods A review of the medical charts of consecutive NSCLC patients diagnosed between January 2006 and May 2020 was performed. We identified 1,382 NSCLC patients with clinical staging of T1/2aN0M0 (excluding BM), and investigated the incidence, clinical predictors, and prognosis of BM in the cohort. We also performed RNA-sequencing differential expression analysis using transcriptome of 8 patients, using DESeq2 package (version 1.32.0) with R (version 4.1.0). Results Among 1,382 patients, nine hundred forty-nine patients (68.7%) underwent brain MRI during staging, and 34 patients (3.6%) were shown to have BM. Firth's bias-reduced logistic regression showed that tumor size (OR 1.056; 95% CI 1.009-1.106, p=0.018) was the only predictor of BM, and pathologic type was not a predictor of BM in our cohort (p>0.05). The median overall survival for patients with brain metastasis was 5.5 years, which is better than previously reported in the literature. RNA-sequencing differential expression analysis revealed the top 10 significantly upregulated genes and top 10 significantly downregulated genes. Among the genes involved in BM, Unc-79 homolog, non-selective sodium leak channel (NALCN) channel complex subunit (UNC79) was the most highly expressed gene in the lung adenocarcinoma tissues from the BM group, and an in vitro assay using A549 cells revealed that the NALCN inhibitor suppressed lung cancer cell proliferation and migration. Conclusions Given the incidence and favorable outcome of BM in patients with suspected early-stage NSCLC, selective screening with brain MRI may be considered, especially in patients with high-risk features.
Collapse
Affiliation(s)
- Kangjoon Kim
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jibeom Lee
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Jeong-Yun Lee
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Seung Hyun Yong
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Eun Young Kim
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Ji Ye Jung
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Young Ae Kang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Moo Suk Park
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Young Sam Kim
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Chang-Myung Oh
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
- *Correspondence: Chang-Myung Oh, ; Sang Hoon Lee,
| | - Sang Hoon Lee
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
- *Correspondence: Chang-Myung Oh, ; Sang Hoon Lee,
| |
Collapse
|
19
|
Choudhury K, Howard RJ, Delemotte L. An α-π transition in S6 shapes the conformational cycle of the bacterial sodium channel NavAb. J Gen Physiol 2022; 155:213748. [PMID: 36515966 PMCID: PMC9754703 DOI: 10.1085/jgp.202213214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 10/17/2022] [Accepted: 11/28/2022] [Indexed: 12/15/2022] Open
Abstract
Voltage-gated sodium channels play an important role in electrical signaling in excitable cells. In response to changes in membrane potential, they cycle between nonconducting and conducting conformations. With recent advances in structural biology, structures of sodium channels have been captured in several distinct conformations, which are thought to represent different functional states. However, it has been difficult to capture the intrinsically transient open state. We recently showed that a proposed open state of the bacterial sodium channel NavMs was not conductive and that a conformational change involving a transition to a π-helix in the pore-lining S6 helix converted this structure into a conducting state. However, the relevance of this structural feature in other sodium channels, and its implications for the broader gating cycle, remained unclear. Here, we propose a comparable open state of another class of bacterial channel from Aliarcobacter butzleri (NavAb) with characteristic pore hydration, ion permeation, and drug binding properties. Furthermore, we show that a π-helix transition can lead to pore opening and that such a conformational change blocks fenestrations in the inner helix bundle. We also discover that a region in the C-terminal domain can undergo a disordering transition proposed to be important for pore opening. These results support a role for a π-helix transition in the opening of NavAb, enabling new proposals for the structural annotation and drug modulation mechanisms in this important sodium channel model.
Collapse
Affiliation(s)
- Koushik Choudhury
- Science for Life Laboratory, Department of Applied Physics, KTH Royal Institute of Technology, Solna, Sweden
| | - Rebecca J. Howard
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Lucie Delemotte
- Science for Life Laboratory, Department of Applied Physics, KTH Royal Institute of Technology, Solna, Sweden,Correspondence to Lucie Delemotte:
| |
Collapse
|
20
|
Rahrmann EP, Shorthouse D, Jassim A, Hu LP, Ortiz M, Mahler-Araujo B, Vogel P, Paez-Ribes M, Fatemi A, Hannon GJ, Iyer R, Blundon JA, Lourenço FC, Kay J, Nazarian RM, Hall BA, Zakharenko SS, Winton DJ, Zhu L, Gilbertson RJ. The NALCN channel regulates metastasis and nonmalignant cell dissemination. Nat Genet 2022; 54:1827-1838. [PMID: 36175792 PMCID: PMC9729110 DOI: 10.1038/s41588-022-01182-0] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 08/08/2022] [Indexed: 02/07/2023]
Abstract
We identify the sodium leak channel non-selective protein (NALCN) as a key regulator of cancer metastasis and nonmalignant cell dissemination. Among 10,022 human cancers, NALCN loss-of-function mutations were enriched in gastric and colorectal cancers. Deletion of Nalcn from gastric, intestinal or pancreatic adenocarcinomas in mice did not alter tumor incidence, but markedly increased the number of circulating tumor cells (CTCs) and metastases. Treatment of these mice with gadolinium-a NALCN channel blocker-similarly increased CTCs and metastases. Deletion of Nalcn from mice that lacked oncogenic mutations and never developed cancer caused shedding of epithelial cells into the blood at levels equivalent to those seen in tumor-bearing animals. These cells trafficked to distant organs to form normal structures including lung epithelium, and kidney glomeruli and tubules. Thus, NALCN regulates cell shedding from solid tissues independent of cancer, divorcing this process from tumorigenesis and unmasking a potential new target for antimetastatic therapies.
Collapse
Affiliation(s)
- Eric P Rahrmann
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - David Shorthouse
- Department of Medical Physics and Biomedical Engineering, University College London, London, UK
| | - Amir Jassim
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Linda P Hu
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Mariaestela Ortiz
- Molecular Pharmacology Lab, Organoid Models Research and Biology, National Cancer Institute, Leidos Biomedical Research, Frederick, MD, USA
| | - Betania Mahler-Araujo
- Wellcome-MRC Institute of Metabolic Science, Histopathology Core, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Peter Vogel
- Veterinary Pathology Core Laboratory, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Marta Paez-Ribes
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Atefeh Fatemi
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Gregory J Hannon
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Radhika Iyer
- Texas Children's Cancer and Hematology Centers, Houston, TX, USA
| | - Jay A Blundon
- Department of Developmental Neurobiology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Filipe C Lourenço
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Jonathan Kay
- Departments of Medicine and of Population and Quantitative Health Sciences, University of Massachusetts Medical School and UMass Memorial Medical Center, Worcester, MA, USA
| | - Rosalynn M Nazarian
- Massachusetts General Hospital, Pathology Service, Dermatopathology Unit, Boston, MA, USA
| | - Benjamin A Hall
- Department of Medical Physics and Biomedical Engineering, University College London, London, UK
| | - Stanislav S Zakharenko
- Department of Developmental Neurobiology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Douglas J Winton
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Liqin Zhu
- Department of Pharmaceutical Sciences, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Richard J Gilbertson
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK.
- Department of Oncology, University of Cambridge, Cambridge, UK.
| |
Collapse
|
21
|
Bamgboye MA, Herold KG, Vieira DC, Traficante MK, Rogers PJ, Ben-Johny M, Dick IE. CaV1.2 channelopathic mutations evoke diverse pathophysiological mechanisms. J Gen Physiol 2022; 154:e202213209. [PMID: 36167061 PMCID: PMC9524202 DOI: 10.1085/jgp.202213209] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 08/09/2022] [Accepted: 09/13/2022] [Indexed: 01/17/2023] Open
Abstract
The first pathogenic mutation in CaV1.2 was identified in 2004 and was shown to cause a severe multisystem disorder known as Timothy syndrome (TS). The mutation was localized to the distal S6 region of the channel, a region known to play a major role in channel activation. TS patients suffer from life-threatening cardiac symptoms as well as significant neurodevelopmental deficits, including autism spectrum disorder (ASD). Since this discovery, the number and variety of mutations identified in CaV1.2 have grown tremendously, and the distal S6 regions remain a frequent locus for many of these mutations. While the majority of patients harboring these mutations exhibit cardiac symptoms that can be well explained by known pathogenic mechanisms, the same cannot be said for the ASD or neurodevelopmental phenotypes seen in some patients, indicating a gap in our understanding of the pathogenesis of CaV1.2 channelopathies. Here, we use whole-cell patch clamp, quantitative Ca2+ imaging, and single channel recordings to expand the known mechanisms underlying the pathogenesis of CaV1.2 channelopathies. Specifically, we find that mutations within the S6 region can exert independent and separable effects on activation, voltage-dependent inactivation (VDI), and Ca2+-dependent inactivation (CDI). Moreover, the mechanisms underlying the CDI effects of these mutations are varied and include altered channel opening and possible disruption of CDI transduction. Overall, these results provide a structure-function framework to conceptualize the role of S6 mutations in pathophysiology and offer insight into the biophysical defects associated with distinct clinical manifestations.
Collapse
Affiliation(s)
- Moradeke A. Bamgboye
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD
| | - Kevin G. Herold
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD
| | - Daiana C.O. Vieira
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD
| | - Maria K. Traficante
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD
| | - Philippa J. Rogers
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD
| | - Manu Ben-Johny
- Department of Physiology and Biophysics, Columbia University, New York, NY
| | - Ivy E. Dick
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD
| |
Collapse
|
22
|
Meyer DJ, Díaz-García CM, Nathwani N, Rahman M, Yellen G. The Na +/K + pump dominates control of glycolysis in hippocampal dentate granule cells. eLife 2022; 11:e81645. [PMID: 36222651 PMCID: PMC9592084 DOI: 10.7554/elife.81645] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 10/11/2022] [Indexed: 11/13/2022] Open
Abstract
Cellular ATP that is consumed to perform energetically expensive tasks must be replenished by new ATP through the activation of metabolism. Neuronal stimulation, an energetically demanding process, transiently activates aerobic glycolysis, but the precise mechanism underlying this glycolysis activation has not been determined. We previously showed that neuronal glycolysis is correlated with Ca2+ influx, but is not activated by feedforward Ca2+ signaling (Díaz-García et al., 2021a). Since ATP-powered Na+ and Ca2+ pumping activities are increased following stimulation to restore ion gradients and are estimated to consume most neuronal ATP, we aimed to determine if they are coupled to neuronal glycolysis activation. By using two-photon imaging of fluorescent biosensors and dyes in dentate granule cell somas of acute mouse hippocampal slices, we observed that production of cytoplasmic NADH, a byproduct of glycolysis, is strongly coupled to changes in intracellular Na+, while intracellular Ca2+ could only increase NADH production if both forward Na+/Ca2+ exchange and Na+/K+ pump activity were intact. Additionally, antidromic stimulation-induced intracellular [Na+] increases were reduced >50% by blocking Ca2+ entry. These results indicate that neuronal glycolysis activation is predominantly a response to an increase in activity of the Na+/K+ pump, which is strongly potentiated by Na+ influx through the Na+/Ca2+ exchanger during extrusion of Ca2+ following stimulation.
Collapse
Affiliation(s)
- Dylan J Meyer
- Department of Neurobiology, Harvard Medical SchoolBostonUnited States
| | | | - Nidhi Nathwani
- Department of Neurobiology, Harvard Medical SchoolBostonUnited States
| | - Mahia Rahman
- Department of Neurobiology, Harvard Medical SchoolBostonUnited States
| | - Gary Yellen
- Department of Neurobiology, Harvard Medical SchoolBostonUnited States
| |
Collapse
|
23
|
Network-Based Data Analysis Reveals Ion Channel-Related Gene Features in COVID-19: A Bioinformatic Approach. Biochem Genet 2022; 61:471-505. [PMID: 36104591 PMCID: PMC9473477 DOI: 10.1007/s10528-022-10280-x] [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: 03/19/2022] [Accepted: 09/01/2022] [Indexed: 11/02/2022]
Abstract
Coronavirus disease 2019 (COVID-19) seriously threatens human health and has been disseminated worldwide. Although there are several treatments for COVID-19, its control is currently suboptimal. Therefore, the development of novel strategies to treat COVID-19 is necessary. Ion channels are located on the membranes of all excitable cells and many intracellular organelles and are key components involved in various biological processes. They are a target of interest when searching for drug targets. This study aimed to reveal the relevant molecular features of ion channel genes in COVID-19 based on bioinformatic analyses. The RNA-sequencing data of patients with COVID-19 and healthy subjects (GSE152418 and GSE171110 datasets) were obtained from the Gene Expression Omnibus (GEO) database. Ion channel genes were selected from the Hugo Gene Nomenclature Committee (HGNC) database. The RStudio software was used to process the data based on the corresponding R language package to identify ion channel-associated differentially expressed genes (DEGs). Based on the DEGs, Gene Ontology (GO) functional and pathway enrichment analyses were performed using the Enrichr web tool. The STRING database was used to generate a protein-protein interaction (PPI) network, and the Cytoscape software was used to screen for hub genes in the PPI network based on the cytoHubba plug-in. Transcription factors (TF)-DEG, DEG-microRNA (miRNA) and DEG-disease association networks were constructed using the NetworkAnalyst web tool. Finally, the screened hub genes as drug targets were subjected to enrichment analysis based on the DSigDB using the Enrichr web tool to identify potential therapeutic agents for COVID-19. A total of 29 ion channel-associated DEGs were identified. GO functional analysis showed that the DEGs were integral components of the plasma membrane and were mainly involved in inorganic cation transmembrane transport and ion channel activity functions. Pathway analysis showed that the DEGs were mainly involved in nicotine addiction, calcium regulation in the cardiac cell and neuronal system pathways. The top 10 hub genes screened based on the PPI network included KCNA2, KCNJ4, CACNA1A, CACNA1E, NALCN, KCNA5, CACNA2D1, TRPC1, TRPM3 and KCNN3. The TF-DEG and DEG-miRNA networks revealed significant TFs (FOXC1, GATA2, HINFP, USF2, JUN and NFKB1) and miRNAs (hsa-mir-146a-5p, hsa-mir-27a-3p, hsa-mir-335-5p, hsa-let-7b-5p and hsa-mir-129-2-3p). Gene-disease association network analysis revealed that the DEGs were closely associated with intellectual disability and cerebellar ataxia. Drug-target enrichment analysis showed that the relevant drugs targeting the hub genes CACNA2D1, CACNA1A, CACNA1E, KCNA2 and KCNA5 were gabapentin, gabapentin enacarbil, pregabalin, guanidine hydrochloride and 4-aminopyridine. The results of this study provide a valuable basis for exploring the mechanisms of ion channel genes in COVID-19 and clues for developing therapeutic strategies for COVID-19.
Collapse
|
24
|
Kang Y, Chen L. Structure and mechanism of NALCN-FAM155A-UNC79-UNC80 channel complex. Nat Commun 2022; 13:2639. [PMID: 35550517 PMCID: PMC9098444 DOI: 10.1038/s41467-022-30403-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 04/27/2022] [Indexed: 11/09/2022] Open
Abstract
NALCN channel mediates sodium leak currents and is important for maintaining proper resting membrane potential. NALCN and FAM155A form the core complex of the channel, the activity of which essentially depends on the presence of both UNC79 and UNC80, two auxiliary proteins. NALCN, FAM155A, UNC79, and UNC80 co-assemble into a large hetero-tetrameric channel complex. Genetic mutations of NALCN channel components lead to neurodevelopmental diseases. However, the structure and mechanism of the intact channel complex remain elusive. Here, we present the cryo-EM structure of the mammalian NALCN-FAM155A-UNC79-UNC80 quaternary complex. The structure shows that UNC79-UNC80 form a large piler-shaped heterodimer which was tethered to the intracellular side of the NALCN channel through tripartite interactions with the cytoplasmic loops of NALCN. Two interactions are essential for proper cell surface localization of NALCN. The other interaction relieves the self-inhibition of NALCN by pulling the auto-inhibitory CTD Interacting Helix (CIH) out of its binding site. Our work defines the structural mechanism of NALCN modulation by UNC79 and UNC80.
Collapse
Affiliation(s)
- Yunlu Kang
- State Key Laboratory of Membrane Biology, College of Future Technology, Institute of Molecular Medicine, Peking. University, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Beijing, 100871, China.,National Biomedical Imaging Center, Peking University, Beijing, 100871, China
| | - Lei Chen
- State Key Laboratory of Membrane Biology, College of Future Technology, Institute of Molecular Medicine, Peking. University, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Beijing, 100871, China. .,National Biomedical Imaging Center, Peking University, Beijing, 100871, China. .,Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China. .,Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China.
| |
Collapse
|
25
|
Abstract
NALCN regulates the resting membrane potential by mediating the Na+ leak current in neurons, and it functions as a channelosome in complex with FAM155A, UNC79, and UNC80. Dysfunction of the NALCN channelosome causes a broad range of neurological and developmental diseases called NALCN channelopathies in humans. How the auxiliary subunits, especially the two large components UNC79 and UNC80, assemble with NALCN and regulate its function remains unclear. Here we report an overall architecture of the human NALCN channelosome. UNC79 and UNC80 each adopt an S-shape super-helical structure consisting of HEAT and armadillo repeats, forming a super-coiled heterodimeric assembly in the cytoplasmic side, which may provide a scaffold for the binding of other potential modulators of the channelosome. The UNC79-UNC80 assembly specifically associates with the NALCN-FAM155A subcomplex through the intracellular II-III linker of NALCN. Disruptions of the interaction interfaces between UNC79 and UNC80, and between the II-III linker of NALCN and the UNC79-UNC80 assembly, significantly reduce the NALCN-mediated currents in HEK293T system, suggesting the importance of the UNC79-UNC80 assembly in regulating channelosome function. Cross-linking mass spectrometry analysis identified an additional calmodulin (CaM) bound in the carboxyl-terminal domain of NALCN. Our study thus provides a structural basis for understanding the unique assembly mechanism and functional regulation of the NALCN channelosome, and also provides an opportunity for the interpretation of many disease-related mutations in UNC80.
Collapse
|
26
|
Structural architecture of the human NALCN channelosome. Nature 2022; 603:180-186. [PMID: 34929720 DOI: 10.1038/s41586-021-04313-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 12/07/2021] [Indexed: 11/08/2022]
Abstract
Depolarizing sodium (Na+) leak currents carried by the NALCN channel regulate the resting membrane potential of many neurons to modulate respiration, circadian rhythm, locomotion and pain sensitivity1-8. NALCN requires FAM155A, UNC79 and UNC80 to function, but the role of these auxiliary subunits is not understood3,7,9-12. NALCN, UNC79 and UNC80 are essential in rodents2,9,13, and mutations in human NALCN and UNC80 cause severe developmental and neurological disease14,15. Here we determined the structure of the NALCN channelosome, an approximately 1-MDa complex, as fundamental aspects about the composition, assembly and gating of this channelosome remain obscure. UNC79 and UNC80 are massive HEAT-repeat proteins that form an intertwined anti-parallel superhelical assembly, which docks intracellularly onto the NALCN-FAM155A pore-forming subcomplex. Calmodulin copurifies bound to the carboxy-terminal domain of NALCN, identifying this region as a putative modulatory hub. Single-channel analyses uncovered a low open probability for the wild-type complex, highlighting the tightly closed S6 gate in the structure, and providing a basis to interpret the altered gating properties of disease-causing variants. Key constraints between the UNC79-UNC80 subcomplex and the NALCN DI-DII and DII-DIII linkers were identified, leading to a model of channelosome gating. Our results provide a structural blueprint to understand the physiology of the NALCN channelosome and a template for drug discovery to modulate the resting membrane potential.
Collapse
|
27
|
Winczewska-Wiktor A, Hirschfeld AS, Badura-Stronka M, Wojsyk-Banaszak I, Sobkowiak P, Bartkowska-Śniatkowska A, Babak V, Steinborn B. Central Apneas Due to the CLIFAHDD Syndrome Successfully Treated with Pyridostigmine. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph19020775. [PMID: 35055596 PMCID: PMC8776169 DOI: 10.3390/ijerph19020775] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 12/12/2021] [Accepted: 01/06/2022] [Indexed: 02/05/2023]
Abstract
NALCN mutations lead to complex neurodevelopmental syndromes, including infantile hypotonia with psychomotor retardation and characteristic facies (IHPRF) and congenital contractures of limbs and face, hypotonia, and developmental delay (CLIFAHDD), which are recessively and dominantly inherited, respectively. We present a patient in whom congenital myasthenic syndrome (CMS) was suspected due to the occurrence of hypotonia and apnea episodes requiring resuscitation. For this reason, treatment with pyridostigmine was introduced. After starting the treatment, a significant improvement was observed in reducing the apnea episodes and slight psychomotor progress. In the course of further diagnostics, CMS was excluded, and CLIFAHDD syndrome was confirmed. Thus, we try to explain a possible mechanism of clinical improvement after the introduction of treatment with pyridostigmine in a patient with a mutation in the NALCN gene.
Collapse
Affiliation(s)
- Anna Winczewska-Wiktor
- Chair and Department of Developmental Neurology, Poznan University of Medical Sciences, Przybyszewskiego 49, 60-355 Poznan, Poland;
- Correspondence:
| | - Adam Sebastian Hirschfeld
- Chair and Department of Medical Genetics, Poznan University of Medical Sciences, 60-352 Poznan, Poland; (A.S.H.); (M.B.-S.); (V.B.)
| | - Magdalena Badura-Stronka
- Chair and Department of Medical Genetics, Poznan University of Medical Sciences, 60-352 Poznan, Poland; (A.S.H.); (M.B.-S.); (V.B.)
- Centers of Medical Genetics GENESIS, 60-529 Poznan, Poland
| | - Irena Wojsyk-Banaszak
- Department of Pulmonology, Pediatric Allergy and Clinical Immunology, Poznan University of Medical Sciences, 60-572 Poznan, Poland; (I.W.-B.); (P.S.)
| | - Paulina Sobkowiak
- Department of Pulmonology, Pediatric Allergy and Clinical Immunology, Poznan University of Medical Sciences, 60-572 Poznan, Poland; (I.W.-B.); (P.S.)
| | - Alicja Bartkowska-Śniatkowska
- Department of Pediatric Anesthesiology and Intensive Therapy, Poznan University of Medical Sciences, 60-572 Poznan, Poland;
| | - Valeriia Babak
- Chair and Department of Medical Genetics, Poznan University of Medical Sciences, 60-352 Poznan, Poland; (A.S.H.); (M.B.-S.); (V.B.)
| | - Barbara Steinborn
- Chair and Department of Developmental Neurology, Poznan University of Medical Sciences, Przybyszewskiego 49, 60-355 Poznan, Poland;
| |
Collapse
|
28
|
Lees JA, Dias JM, Han S. Applications of Cryo-EM in small molecule and biologics drug design. Biochem Soc Trans 2021; 49:2627-2638. [PMID: 34812853 PMCID: PMC8786282 DOI: 10.1042/bst20210444] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 10/22/2021] [Accepted: 10/27/2021] [Indexed: 02/03/2023]
Abstract
Electron cryo-microscopy (cryo-EM) is a powerful technique for the structural characterization of biological macromolecules, enabling high-resolution analysis of targets once inaccessible to structural interrogation. In recent years, pharmaceutical companies have begun to utilize cryo-EM for structure-based drug design. Structural analysis of integral membrane proteins, which comprise a large proportion of druggable targets and pose particular challenges for X-ray crystallography, by cryo-EM has enabled insights into important drug target families such as G protein-coupled receptors (GPCRs), ion channels, and solute carrier (SLCs) proteins. Structural characterization of biologics, such as vaccines, viral vectors, and gene therapy agents, has also become significantly more tractable. As a result, cryo-EM has begun to make major impacts in bringing critical therapeutics to market. In this review, we discuss recent instructive examples of impacts from cryo-EM in therapeutics design, focusing largely on its implementation at Pfizer. We also discuss the opportunities afforded by emerging technological advances in cryo-EM, and the prospects for future development of the technique.
Collapse
Affiliation(s)
- Joshua A. Lees
- Discovery Sciences, Medicine Design, Pfizer Worldwide Research and Development, Groton, CT 06340, U.S.A
| | - Joao M. Dias
- Discovery Sciences, Medicine Design, Pfizer Worldwide Research and Development, Groton, CT 06340, U.S.A
| | - Seungil Han
- Discovery Sciences, Medicine Design, Pfizer Worldwide Research and Development, Groton, CT 06340, U.S.A
| |
Collapse
|
29
|
Structural basis for Ca 2+ activation of the heteromeric PKD1L3/PKD2L1 channel. Nat Commun 2021; 12:4871. [PMID: 34381056 PMCID: PMC8357825 DOI: 10.1038/s41467-021-25216-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 07/23/2021] [Indexed: 02/07/2023] Open
Abstract
The heteromeric complex between PKD1L3, a member of the polycystic kidney disease (PKD) protein family, and PKD2L1, also known as TRPP2 or TRPP3, has been a prototype for mechanistic characterization of heterotetrametric TRP-like channels. Here we show that a truncated PKD1L3/PKD2L1 complex with the C-terminal TRP-fold fragment of PKD1L3 retains both Ca2+ and acid-induced channel activities. Cryo-EM structures of this core heterocomplex with or without supplemented Ca2+ were determined at resolutions of 3.1 Å and 3.4 Å, respectively. The heterotetramer, with a pseudo-symmetric TRP architecture of 1:3 stoichiometry, has an asymmetric selectivity filter (SF) guarded by Lys2069 from PKD1L3 and Asp523 from the three PKD2L1 subunits. Ca2+-entrance to the SF vestibule is accompanied by a swing motion of Lys2069 on PKD1L3. The S6 of PKD1L3 is pushed inward by the S4-S5 linker of the nearby PKD2L1 (PKD2L1-III), resulting in an elongated intracellular gate which seals the pore domain. Comparison of the apo and Ca2+-loaded complexes unveils an unprecedented Ca2+ binding site in the extracellular cleft of the voltage-sensing domain (VSD) of PKD2L1-III, but not the other three VSDs. Structure-guided mutagenic studies support this unconventional site to be responsible for Ca2+-induced channel activation through an allosteric mechanism.
Collapse
|
30
|
Impheng H, Lemmers C, Bouasse M, Legros C, Pakaprot N, Guérineau NC, Lory P, Monteil A. The sodium leak channel NALCN regulates cell excitability of pituitary endocrine cells. FASEB J 2021; 35:e21400. [PMID: 33793981 DOI: 10.1096/fj.202000841rr] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 01/11/2021] [Accepted: 01/13/2021] [Indexed: 11/11/2022]
Abstract
Anterior pituitary endocrine cells that release hormones such as growth hormone and prolactin are excitable and fire action potentials. In these cells, several studies previously showed that extracellular sodium (Na+ ) removal resulted in a negative shift of the resting membrane potential (RMP) and a subsequent inhibition of the spontaneous firing of action potentials, suggesting the contribution of a Na+ background conductance. Here, we show that the Na+ leak channel NALCN conducts a Ca2+ - Gd3+ -sensitive and TTX-resistant Na+ background conductance in the GH3 cell line, a cell model of pituitary endocrine cells. NALCN knockdown hyperpolarized the RMP, altered GH3 cell electrical properties and inhibited prolactin secretion. Conversely, the overexpression of NALCN depolarized the RMP, also reshaping the electrical properties of GH3 cells. Overall, our results indicate that NALCN is functional in GH3 cells and involved in endocrine cell excitability as well as in hormone secretion. Indeed, the GH3 cell line suitably models native pituitary cells that display a similar Na+ background conductance and appears as a proper cellular model to study the role of NALCN in cellular excitability.
Collapse
Affiliation(s)
- Hathaichanok Impheng
- IGF, Université de Montpellier, CNRS, INSERM, Montpellier, France.,LabEx 'Ion Channel Science and Therapeutics', Montpellier, France
| | - Céline Lemmers
- IGF, Université de Montpellier, CNRS, INSERM, Montpellier, France.,PVM, BCM, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Malik Bouasse
- IGF, Université de Montpellier, CNRS, INSERM, Montpellier, France.,LabEx 'Ion Channel Science and Therapeutics', Montpellier, France
| | - Christian Legros
- MITOVASC Institute, UMR CNRS 6015 - UMR INSERM U1083, Université d'Angers, Angers, France
| | - Narawut Pakaprot
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Nathalie C Guérineau
- IGF, Université de Montpellier, CNRS, INSERM, Montpellier, France.,LabEx 'Ion Channel Science and Therapeutics', Montpellier, France
| | - Philippe Lory
- IGF, Université de Montpellier, CNRS, INSERM, Montpellier, France.,LabEx 'Ion Channel Science and Therapeutics', Montpellier, France
| | - Arnaud Monteil
- IGF, Université de Montpellier, CNRS, INSERM, Montpellier, France.,LabEx 'Ion Channel Science and Therapeutics', Montpellier, France.,PVM, BCM, Université de Montpellier, CNRS, INSERM, Montpellier, France
| |
Collapse
|
31
|
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: 8.8] [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
|
32
|
García-Hernández JL, Corchete LA, Marcos-Alcalde Í, Gómez-Puertas P, Fons C, Lazo PA. Pathogenic convergence of CNVs in genes functionally associated to a severe neuromotor developmental delay syndrome. Hum Genomics 2021; 15:11. [PMID: 33557955 PMCID: PMC7871650 DOI: 10.1186/s40246-021-00309-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 01/26/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Complex developmental encephalopathy syndromes might be the consequence of unknown genetic alterations that are likely to contribute to the full neurological phenotype as a consequence of pathogenic gene combinations. METHODS To identify the additional genetic contribution to the neurological phenotype, we studied as a test case a boy, with a KCNQ2 exon-7 partial duplication, by single-nucleotide polymorphism (SNP) microarray to detect copy-number variations (CNVs). RESULTS The proband presented a cerebral palsy like syndrome with a severe motor and developmental encephalopathy. The SNP array analysis detected in the proband several de novo CNVs, nine partial gene losses (LRRC55, PCDH9, NALCN, RYR3, ELAVL2, CDH13, ATP1A2, SLC17A5, ANO3), and two partial gene duplications (PCDH19, EFNA5). The biological functions of these genes are associated with ion channels such as calcium, chloride, sodium, and potassium with several membrane proteins implicated in neural cell-cell interactions, synaptic transmission, and axon guidance. Pathogenically, these functions can be associated to cerebral palsy, seizures, dystonia, epileptic crisis, and motor neuron dysfunction, all present in the patient. CONCLUSIONS Severe motor and developmental encephalopathy syndromes of unknown origin can be the result of a phenotypic convergence by combination of several genetic alterations in genes whose physiological function contributes to the neurological pathogenic mechanism.
Collapse
Affiliation(s)
- Juan L García-Hernández
- Molecular Mechanisms of Cancer Program, Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC), Universidad de Salamanca, Salamanca, Spain.,Instituto de Investigación Biomédica de Salamanca (IBSAL), Departamento de Hematología, Hospital Universitario de Salamanca, Salamanca, Spain
| | - Luis A Corchete
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Departamento de Hematología, Hospital Universitario de Salamanca, Salamanca, Spain.,Network Center for Biomedical Research in Cancer (CIBERONC), Salamanca, Spain
| | - Íñigo Marcos-Alcalde
- Centro de Biología Molecular Severo Ochoa, CSIC-Universidad Autónoma de Madrid, Cantoblanco, Madrid, Spain.,Biosciences Research Institute, School of Experimental Sciences, Universidad Francisco de Vitoria, Pozuelo de Alarcón, Madrid, Spain
| | - Paulino Gómez-Puertas
- Centro de Biología Molecular Severo Ochoa, CSIC-Universidad Autónoma de Madrid, Cantoblanco, Madrid, Spain
| | - Carmen Fons
- Neurology Department, Hospital Sant Joan de Déu, Sant Joan de Déu Research Institute, Esplugues de Llobregat, Barcelona and CIBERER, Instituto de Salud Carlos III, Barcelona, Spain.
| | - Pedro A Lazo
- Molecular Mechanisms of Cancer Program, Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC), Universidad de Salamanca, Salamanca, Spain. .,Instituto de Investigación Biomédica de Salamanca (IBSAL), Departamento de Hematología, Hospital Universitario de Salamanca, Salamanca, Spain.
| |
Collapse
|
33
|
The Somatosensory World of the African Naked Mole-Rat. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1319:197-220. [PMID: 34424517 DOI: 10.1007/978-3-030-65943-1_7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The naked mole-rat (Heterocephalus glaber) is famous for its longevity and unusual physiology. This eusocial species that lives in highly ordered and hierarchical colonies with a single breeding queen, also discovered secrets enabling somewhat pain-free living around 20 million years ago. Unlike most mammals, naked mole-rats do not feel the burn of chili pepper's active ingredient, capsaicin, nor the sting of acid. Indeed, by accumulating mutations in genes encoding proteins that are only now being exploited as targets for new pain therapies (the nerve growth factor receptor TrkA and voltage-gated sodium channel, NaV1.7), this species mastered the art of analgesia before humans evolved. Recently, we have identified pain-insensitivity as a trait shared by several closely related African mole-rat species. In this chapter we will show how African mole-rats have evolved pain insensitivity as well as discussing what the proximate factors may have been that led to the evolution of pain-free traits.
Collapse
|
34
|
Kang Y, Wu JX, Chen L. Structure of voltage-modulated sodium-selective NALCN-FAM155A channel complex. Nat Commun 2020; 11:6199. [PMID: 33273469 PMCID: PMC7712781 DOI: 10.1038/s41467-020-20002-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 11/10/2020] [Indexed: 12/18/2022] Open
Abstract
Resting membrane potential determines the excitability of the cell and is essential for the cellular electrical activities. The NALCN channel mediates sodium leak currents, which positively adjust resting membrane potential towards depolarization. The NALCN channel is involved in several neurological processes and has been implicated in a spectrum of neurodevelopmental diseases. Here, we report the cryo-EM structure of rat NALCN and mouse FAM155A complex to 2.7 Å resolution. The structure reveals detailed interactions between NALCN and the extracellular cysteine-rich domain of FAM155A. We find that the non-canonical architecture of NALCN selectivity filter dictates its sodium selectivity and calcium block, and that the asymmetric arrangement of two functional voltage sensors confers the modulation by membrane potential. Moreover, mutations associated with human diseases map to the domain-domain interfaces or the pore domain of NALCN, intuitively suggesting their pathological mechanisms. The NALCN channel mediates sodium leak currents, which in turn adjusts resting membrane potential and neuronal excitability. Here the authors describe a cryo-EM structure of mammalian NALCN-FAM155A channel complex, showing how selectivity filter contributes to sodium permeation and calcium block and how the voltage sensors contribute to current modulation.
Collapse
Affiliation(s)
- Yunlu Kang
- State Key Laboratory of Membrane Biology, College of Future Technology, Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, 100871, Beijing, China
| | - Jing-Xiang Wu
- State Key Laboratory of Membrane Biology, College of Future Technology, Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, 100871, Beijing, China.,Peking-Tsinghua Center for Life Sciences, Peking University, 100871, Beijing, China.,Academy for Advanced Interdisciplinary Studies, Peking University, 100871, Beijing, China
| | - Lei Chen
- State Key Laboratory of Membrane Biology, College of Future Technology, Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, 100871, Beijing, China. .,Peking-Tsinghua Center for Life Sciences, Peking University, 100871, Beijing, China. .,Academy for Advanced Interdisciplinary Studies, Peking University, 100871, Beijing, China.
| |
Collapse
|
35
|
Xie J, Ke M, Xu L, Lin S, Huang J, Zhang J, Yang F, Wu J, Yan Z. Structure of the human sodium leak channel NALCN in complex with FAM155A. Nat Commun 2020; 11:5831. [PMID: 33203861 PMCID: PMC7672056 DOI: 10.1038/s41467-020-19667-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 10/26/2020] [Indexed: 01/18/2023] Open
Abstract
NALCN, a sodium leak channel expressed mainly in the central nervous system, is responsible for the resting Na+ permeability that controls neuronal excitability. Dysfunctions of the NALCN channelosome, NALCN with several auxiliary subunits, are associated with a variety of human diseases. Here, we report the cryo-EM structure of human NALCN in complex with FAM155A at an overall resolution of 3.1 angstroms. FAM155A forms extensive interactions with the extracellular loops of NALCN that may help stabilize NALCN in the membrane. A Na+ ion-binding site, reminiscent of a Ca2+ binding site in Cav channels, is identified in the unique EEKE selectivity filter. Despite its 'leaky' nature, the channel is closed and the intracellular gate is sealed by S6I, II-III linker and III-IV linker. Our study establishes the molecular basis of Na+ permeation and voltage sensitivity, and provides important clues to the mechanistic understanding of NALCN regulation and NALCN channelosome-related diseases.
Collapse
Affiliation(s)
- Jiongfang Xie
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, 310024, Hangzhou, Zhejiang, China
- Westlake Laboratory of Life Sciences and Biomedicine, 310024, Hangzhou, Zhejiang, China
- Institute of Biology, Westlake Institute for Advanced Study, 310024, Hangzhou, Zhejiang, China
| | - Meng Ke
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, 310024, Hangzhou, Zhejiang, China
- Westlake Laboratory of Life Sciences and Biomedicine, 310024, Hangzhou, Zhejiang, China
- Institute of Biology, Westlake Institute for Advanced Study, 310024, Hangzhou, Zhejiang, China
| | - Lizhen Xu
- Department of Biophysics and Kidney Disease Center, First Affiliated Hospital, Zhejiang University School of Medicine, 310058, Hangzhou, China
| | - Shiyi Lin
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, 310024, Hangzhou, Zhejiang, China
- Westlake Laboratory of Life Sciences and Biomedicine, 310024, Hangzhou, Zhejiang, China
- Institute of Biology, Westlake Institute for Advanced Study, 310024, Hangzhou, Zhejiang, China
| | - Jin Huang
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, 310024, Hangzhou, Zhejiang, China
- Westlake Laboratory of Life Sciences and Biomedicine, 310024, Hangzhou, Zhejiang, China
- Institute of Biology, Westlake Institute for Advanced Study, 310024, Hangzhou, Zhejiang, China
| | - Jiabei Zhang
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, 310024, Hangzhou, Zhejiang, China
- Westlake Laboratory of Life Sciences and Biomedicine, 310024, Hangzhou, Zhejiang, China
- Institute of Biology, Westlake Institute for Advanced Study, 310024, Hangzhou, Zhejiang, China
| | - Fan Yang
- Department of Biophysics and Kidney Disease Center, First Affiliated Hospital, Zhejiang University School of Medicine, 310058, Hangzhou, China.
| | - Jianping Wu
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, 310024, Hangzhou, Zhejiang, China.
- Westlake Laboratory of Life Sciences and Biomedicine, 310024, Hangzhou, Zhejiang, China.
- Institute of Biology, Westlake Institute for Advanced Study, 310024, Hangzhou, Zhejiang, China.
| | - Zhen Yan
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, 310024, Hangzhou, Zhejiang, China.
- Westlake Laboratory of Life Sciences and Biomedicine, 310024, Hangzhou, Zhejiang, China.
- Institute of Biology, Westlake Institute for Advanced Study, 310024, Hangzhou, Zhejiang, China.
| |
Collapse
|