1
|
Chen P, Wang S, Zhang H, Li J. Recent advances in nanotherapy-based treatment of epilepsy. Colloids Surf B Biointerfaces 2025; 249:114499. [PMID: 39778465 DOI: 10.1016/j.colsurfb.2025.114499] [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: 10/22/2024] [Revised: 12/21/2024] [Accepted: 01/04/2025] [Indexed: 01/11/2025]
Abstract
Epilepsy is a complex neurological disorder characterized by recurrent seizures affecting millions of people worldwide. Despite advances in drug therapy, a significant proportion of patients remain resistant to conventional antiepileptic drugs (AEDs) due to challenges such as impermeability of the blood-brain barrier (BBB), multidrug resistance, and multifaceted epileptogenesis. Nanotechnology offers promising strategies to overcome these barriers by enhancing drug delivery across the BBB, improving target specificity and minimizing systemic side effects. This review explores recent advances in different innovative strategies of nanodelivery systems for epilepsy therapy, and we will discuss the design principles, mechanisms of action and therapeutic efficacy of these nanodelivery systems. In addition, we discuss the challenges and limitations that hinder the clinical translation of nanomedicine-based therapies for epilepsy. We emphasize the need for personalized and multidisciplinary approaches as well as the importance of continued research and interdisciplinary collaboration in order to translate these innovative strategies into effective therapies. Ultimately, the use of nanotechnology has the potential to enhance seizure control, reduce the burden of epilepsy, and improve the quality of life of patients affected by this complex neurological disorder. Nanotechnology-based drug delivery systems may usher in a new era of precision medicine for epilepsy treatment.
Collapse
Affiliation(s)
- Peng Chen
- General Hospital of Northern Theater Command, Liaoning 110016, China
| | - Shudong Wang
- Jinzhou Medical University, Liaoning 121001, China
| | - Heming Zhang
- Dalian Medical University, Liaoning 116044, China
| | - Jian Li
- General Hospital of Northern Theater Command, Liaoning 110016, China.
| |
Collapse
|
2
|
Kodirov SA. Roles of funny HCN. Comp Biochem Physiol C Toxicol Pharmacol 2025; 295:110205. [PMID: 40233889 DOI: 10.1016/j.cbpc.2025.110205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 03/27/2025] [Accepted: 04/10/2025] [Indexed: 04/17/2025]
Abstract
To some extent, the main role of hyperpolarization-activated cyclic nucleotide-gated non-selective cation channels (HCN, Ih, or If), pace-making, is dogmatized as a functional expression of one or another alpha subunit of HCN channels does not make every region of the brain or heart a pacemaker one. Recent research hints at the role of HCN in arrhythmias and seizures that are often caused by voltage-dependent K and Na channels (Kv and Nav) and neurotransmitters, respectively. There are many parallels between the HCN and K channels. Similar to Kv channels, an altered HCN function also leads to long QT interval. Moreover, a mutation in HCN is believed to trigger correlated arrhythmias and, e.g., epilepsy, among many other brain pathologies. Unlike Kv channels, although no dedicated ancillary beta subunit has been discovered for HCN, the Ih properties are also influenced by other elements and factors. A new interaction has been discovered between HCN and the vesicle-associated membrane protein (VAMP). The prevailing interaction occurs via the subtype VAMP-associated protein B (VAPB). However, this interaction is not unique but universal, since there is also a link between Kv2.1 and VAMP2 (vesicular SNARE). The most remarkable similitude is the fact that a selective antagonist of HCN and medication ivabradine prevents the IKr via the cloned human ether-à-go-go-related gene (HERG) channels, also known as KvLQT and Kv11.1 alpha subunit.
Collapse
Affiliation(s)
- Sodikdjon A Kodirov
- Pavlov Institute of Physiology, Russian Academy of Sciences, Saint Petersburg, Russia; Institute of Physiology and Pathophysiology, University of Mainz, Germany; University of Texas at Brownsville, Department of Biological Sciences, TX 78520, USA; Instituto de Medicina Molecular, Universidade de Lisboa, Lisbon, Portugal; Institute of Biophysics, Johannes Kepler University, Linz, Austria.
| |
Collapse
|
3
|
Han Z, Xie L, Liu X, Yang J, Luo H, Ding R, Chen H, Cheng L, Fang Z, Jiang L. Characterization of the phenotype and functional alternations of three HCN1 variants in Chinese epilepsy patients. Epilepsy Res 2025; 211:107521. [PMID: 39952090 DOI: 10.1016/j.eplepsyres.2025.107521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 01/27/2025] [Accepted: 02/01/2025] [Indexed: 02/17/2025]
Abstract
PURPOSE To evaluate the electrophysiological properties of three HCN1 variant sites found in Chinese epileptic patients and to explore the potential relationship between genotype and phenotype. METHODS We correlated clinical severity of three patients with HCN1 variants with whole-cell patch-clamp measurements of channel activity, channel expression detected by Western blot, and bioinformatics prediction of the damaging effects of each variant. RESULTS Three patients with the variants p.L400P, p.D534H and p.M243delinsTL, showed different phenotypes, ranging from mild epilepsy to severe epileptic encephalopathy. Variants L400P and D534H were classified as pathogenic by all bioinformatics tools, and variant M243delinsTL was classified as a polymorphism by MutationTaster. The L400P and D534H variants showed significantly reduced current compared with that of the wild-type (WT), while the current density of M243delinsTL was similar to WT. The half-activation voltage (V1/2) of M243delinsTL variant was shifted in the hyperpolarizing direction when compared to the WT, and the slope factor (k) of activation of the M243delinsTL variant was significantly lower than that of the WT. The L400P variant was associated with a significantly higher activation time constant compared with that of the WT. In addition, quantitative detection of the FLAG-tagged HCN1 channel revealed that the expression level of the L400P variant was significantly decreased compared to WT. CONCLUSIONS Elucidation of the type and location of variant sites combined with the use of bioinformatics tools and patch-clamp techniques can improve our understanding of the clinical phenotype of epilepsy associated with HCN1 variants.
Collapse
Affiliation(s)
- Ziyao Han
- Department of Neurology Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Chongqing 400014, China
| | - Lingling Xie
- Department of Neurology Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Chongqing 400014, China
| | - Xiaorui Liu
- Department of Neurology Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Chongqing 400014, China
| | - Jiaxin Yang
- Department of Neurology Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Chongqing 400014, China
| | - Hanyu Luo
- Department of Neurology Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Chongqing 400014, China
| | - Ran Ding
- Department of Neurology Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Chongqing 400014, China
| | - Hengsheng Chen
- Department of Neurology Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Chongqing 400014, China
| | - Li Cheng
- Department of Neurology Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Chongqing 400014, China
| | - Zhixu Fang
- Department of Neurology Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Chongqing 400014, China.
| | - Li Jiang
- Department of Neurology Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Chongqing 400014, China.
| |
Collapse
|
4
|
Bleakley LE, Reid CA. HCN1 epilepsy: From genetics and mechanisms to precision therapies. J Neurochem 2024; 168:3891-3910. [PMID: 37565989 DOI: 10.1111/jnc.15928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 07/12/2023] [Accepted: 07/22/2023] [Indexed: 08/12/2023]
Abstract
Pathogenic variation in HCN1 is now an established cause of epilepsy and intellectual disability. Variation in HCN1 causes a spectrum of disease with a genotype-phenotype relationship emerging. De novo pathogenic variants that occur in the transmembrane domains of the channel typically cause a cation 'leak' that associates with severe developmental and epileptic encephalopathy (DEE). Genotype-phenotype associations for variants that fall outside of the transmembrane domains are less well established but do include milder forms of epilepsy that can be either de novo or inherited. HCN1 DEE mouse models have been generated which recapitulate the seizures and learning difficulties seen in human patients. These mice have also acted as powerful preclinical models which share pharmacoresponsiveness with human HCN1 DEE patients. Data from these mouse models support the conclusion that anti-seizure medications with sodium channel block as their primary mechanism of action should be used with caution in HCN1 DEE. Other comorbidities of HCN1 DEE including retinal dysfunction have also been modelled in HCN1 DEE mice, suggesting HCN1 variants can cause a dramatically reduced sensitivity to light with limited ability to process temporal information. Our understanding of the genetics and pathophysiological mechanisms underlying HCN1 epilepsy has progressed significantly and is already influencing therapy. However, more research effort is needed to fully understand the natural histories of HCN1 epilepsies and to develop precision therapeutic approaches.
Collapse
Affiliation(s)
- Lauren E Bleakley
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - Christopher A Reid
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
5
|
Zhao P, Xiong H, Kuang G, Sun C, Zhang X, Huang Y, Luo S, Zhang L, Jiang J, He X. Analysis of epilepsy-associated variants in HCN3 - Functional implications and clinical observations. Epilepsia Open 2024; 9:2294-2305. [PMID: 39361439 PMCID: PMC11633725 DOI: 10.1002/epi4.13049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 08/19/2024] [Accepted: 09/02/2024] [Indexed: 10/05/2024] Open
Abstract
OBJECTIVE This case study investigates the role of hyperpolarization-activated, cyclic nucleotide-gated (HCN) ion channels, which are integral membrane proteins crucial for regulating neuronal excitability. HCN channels are composed of four subunits (HCN1-4), with HCN1, HCN2, and HCN4 previously linked to epilepsy. However, the role of the HCN3 in epileptogenesis remains underexplored. METHODS We recruited a cohort of 298 epilepsy patients to screen for genetic variants in the HCN3 (NM_020897.3) using Sanger sequencing. We identified rare variants and conducted functional assays to evaluate their pathogenicity. RESULTS We identified three rare heterozygous variants in HCN3: c.1370G > A (R457H), c.1982G > A (R661Q), and c.1982G > A(P630L). In vitro functional analyses demonstrated that these variants affected the expression level of HCN3 protein without altering its membrane localization. Whole-cell voltage-clamp experiments showed that two variants (R457H and R661Q) significantly reduced current density in cells, while P630L has no effect on ion channel current. SIGNIFICANCE Our findings suggest that the identified HCN3 genetic variants disrupt HCN ion channel function, highlighting HCN3 as a novel candidate gene involved in epileptic disorders. This expands the genetic landscape of epilepsy and provides new insights into its molecular underpinnings. PLAIN LANGUAGE SUMMARY Epilepsy is a brain disease that can be caused by mutations in specific genes. We found three rare variants in HCN3 gene in 298 patients with epilepsy, and two of the three mutations could be pathogenic and cause epilepsy and another one is single-nucleotide polymorphism, which could have no effect and no contribution to the development of epilepsy.
Collapse
Affiliation(s)
- Peiwei Zhao
- Precision Medical Center, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical CollegeHuazhong University of Science & TechnologyWuhanChina
| | - Hongbo Xiong
- Department of Cardiology, Zhongnan HospitalWuhan UniversityWuhanChina
| | - Gunagtao Kuang
- Department of Neuroelectrophysiology, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical CollegeHuazhong University of Science & TechnologyWuhanChina
| | - Chen Sun
- Maternal Health Care Department, Wuhan Children’s Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical CollegeHuazhong University of Science & TechnologyWuhanChina
| | - Xiankai Zhang
- Precision Medical Center, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical CollegeHuazhong University of Science & TechnologyWuhanChina
| | - Yufeng Huang
- Precision Medical Center, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical CollegeHuazhong University of Science & TechnologyWuhanChina
| | - Sukun Luo
- Precision Medical Center, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical CollegeHuazhong University of Science & TechnologyWuhanChina
| | - Lei Zhang
- Precision Medical Center, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical CollegeHuazhong University of Science & TechnologyWuhanChina
| | - Jun Jiang
- Department of Cardiology, Zhongnan HospitalWuhan UniversityWuhanChina
| | - Xuelian He
- Precision Medical Center, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical CollegeHuazhong University of Science & TechnologyWuhanChina
- Clinical Medical Research Center for Birth Defect Prevention and Treatmentin WuhanWuhan Children’s Hospital (Wuhan Maternal and Child Healthcare Hospital)Tongji Medical College, Huazhong University of Science & TechnologyWuhanChina
| |
Collapse
|
6
|
Cámara-Checa A, Perin F, Rubio-Alarcón M, Dago M, Crespo-García T, Rapún J, Marín M, Cebrián J, Gómez R, Bermúdez-Jiménez F, Monserrat L, Tamargo J, Caballero R, Jiménez-Jáimez J, Delpón E. A gain-of-function HCN4 mutant in the HCN domain is responsible for inappropriate sinus tachycardia in a Spanish family. Proc Natl Acad Sci U S A 2023; 120:e2305135120. [PMID: 38032931 PMCID: PMC10710060 DOI: 10.1073/pnas.2305135120] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 10/12/2023] [Indexed: 12/02/2023] Open
Abstract
In a family with inappropriate sinus tachycardia (IST), we identified a mutation (p.V240M) of the hyperpolarization-activated cyclic nucleotide-gated type 4 (HCN4) channel, which contributes to the pacemaker current (If) in human sinoatrial node cells. Here, we clinically study fifteen family members and functionally analyze the p.V240M variant. Macroscopic (IHCN4) and single-channel currents were recorded using patch-clamp in cells expressing human native (WT) and/or p.V240M HCN4 channels. All p.V240M mutation carriers exhibited IST that was accompanied by cardiomyopathy in adults. IHCN4 generated by p.V240M channels either alone or in combination with WT was significantly greater than that generated by WT channels alone. The variant, which lies in the N-terminal HCN domain, increased the single-channel conductance and opening frequency and probability of HCN4 channels. Conversely, it did not modify the channel sensitivity for cAMP and ivabradine or the level of expression at the membrane. Treatment with ivabradine based on functional data reversed the IST and the cardiomyopathy of the carriers. In computer simulations, the p.V240M gain-of-function variant increases If and beating rate and thus explains the IST of the carriers. The results demonstrate the importance of the unique HCN domain in HCN4, which stabilizes the channels in the closed state.
Collapse
Affiliation(s)
- Anabel Cámara-Checa
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, 28040Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid28029, Spain
| | - Francesca Perin
- Department of Pediatric Cardiology, Virgen de las Nieves University Hospital, Granada18014, Spain
- Instituto de Investigación Biosanitaria de Granada, Granada18014, Spain
| | - Marcos Rubio-Alarcón
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, 28040Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid28029, Spain
| | - María Dago
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, 28040Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid28029, Spain
| | - Teresa Crespo-García
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, 28040Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid28029, Spain
| | - Josu Rapún
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, 28040Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid28029, Spain
| | - María Marín
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid28029, Spain
| | - Jorge Cebrián
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, 28040Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid28029, Spain
| | - Ricardo Gómez
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, 28040Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid28029, Spain
| | - Francisco Bermúdez-Jiménez
- Department of Pediatric Cardiology, Virgen de las Nieves University Hospital, Granada18014, Spain
- Instituto de Investigación Biosanitaria de Granada, Granada18014, Spain
- Centro Nacional de Investigaciones Cardiovasculares, Madrid28029, Spain
| | - Lorenzo Monserrat
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid28029, Spain
- Health in Code Sociedad Limitada, A Coruña15008, Spain
| | - Juan Tamargo
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, 28040Madrid, Spain
| | - Ricardo Caballero
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, 28040Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid28029, Spain
| | - Juan Jiménez-Jáimez
- Department of Pediatric Cardiology, Virgen de las Nieves University Hospital, Granada18014, Spain
- Instituto de Investigación Biosanitaria de Granada, Granada18014, Spain
| | - Eva Delpón
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, 28040Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid28029, Spain
| |
Collapse
|
7
|
DiFrancesco JC, Ragona F, Murano C, Frosio A, Melgari D, Binda A, Calamaio S, Prevostini R, Mauri M, Canafoglia L, Castellotti B, Messina G, Gellera C, Previtali R, Veggiotti P, Milanesi R, Barbuti A, Solazzi R, Freri E, Granata T, Rivolta I. A novel de novo HCN2 loss-of-function variant causing developmental and epileptic encephalopathy treated with a ketogenic diet. Epilepsia 2023; 64:e222-e228. [PMID: 37746765 DOI: 10.1111/epi.17777] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 09/20/2023] [Accepted: 09/20/2023] [Indexed: 09/26/2023]
Abstract
Missense variants of hyperpolarization-activated, cyclic nucleotide-gated (HCN) ion channels cause variable phenotypes, ranging from mild generalized epilepsy to developmental and epileptic encephalopathy (DEE). Although variants of HCN1 are an established cause of DEE, those of HCN2 have been reported in generalized epilepsies. Here we describe the first case of DEE caused by the novel de novo heterozygous missense variant c.1379G>A (p.G460D) of HCN2. Functional characterization in transfected HEK293 cells and neonatal rat cortical neurons revealed that HCN2 p.G460D currents were strongly reduced compared to wild-type, consistent with a dominant negative loss-of-function effect. Immunofluorescence staining showed that mutant channels are retained within the cell and do not reach the membrane. Moreover, mutant HCN2 also affect HCN1 channels, by reducing the Ih current expressed by the HCN1-HCN2 heteromers. Due to the persistence of frequent seizures despite pharmacological polytherapy, the patient was treated with a ketogenic diet, with a significant and long-lasting reduction of episodes. In vitro experiments conducted in a ketogenic environment demonstrated that the clinical improvement observed with this dietary regimen was not mediated by a direct action on HCN2 activity. These results expand the clinical spectrum related to HCN2 channelopathies, further broadening our understanding of the pathogenesis of DEE.
Collapse
Affiliation(s)
| | - Francesca Ragona
- Department of Pediatric Neuroscience, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Carmen Murano
- School of Medicine and Surgery, University of Milano-Bicocca, Milan Center for Neuroscience (NeuroMI), Monza, Italy
| | - Anthony Frosio
- IMTC - Institute of Molecular and Translational Cardiology, San Donato Milanese, Italy
| | - Dario Melgari
- IMTC - Institute of Molecular and Translational Cardiology, San Donato Milanese, Italy
| | - Anna Binda
- School of Medicine and Surgery, University of Milano-Bicocca, Milan Center for Neuroscience (NeuroMI), Monza, Italy
| | - Serena Calamaio
- IMTC - Institute of Molecular and Translational Cardiology, San Donato Milanese, Italy
| | - Rachele Prevostini
- IMTC - Institute of Molecular and Translational Cardiology, San Donato Milanese, Italy
| | - Mario Mauri
- School of Medicine and Surgery, University of Milano-Bicocca, Milan Center for Neuroscience (NeuroMI), Monza, Italy
| | - Laura Canafoglia
- Integrated Diagnostics for Epilepsy, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Barbara Castellotti
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Giuliana Messina
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Cinzia Gellera
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Roberto Previtali
- Pediatric Neurology Unit, V. Buzzi Hospital, University of Milan, Milan, Italy
| | | | - Raffaella Milanesi
- Department of Veterinary Medicine and Animal Science, University of Milan, Lodi, Italy
| | - Andrea Barbuti
- The Cell Physiology MiLab, Department of Biosciences, University of Milano, Milan, Italy
| | - Roberta Solazzi
- Department of Pediatric Neuroscience, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Elena Freri
- Department of Pediatric Neuroscience, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Tiziana Granata
- Department of Pediatric Neuroscience, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Ilaria Rivolta
- School of Medicine and Surgery, University of Milano-Bicocca, Milan Center for Neuroscience (NeuroMI), Monza, Italy
- IMTC - Institute of Molecular and Translational Cardiology, San Donato Milanese, Italy
| |
Collapse
|
8
|
Wittenmayer N, Petkova-Tuffy A, Borgmeyer M, Lee C, Becker J, Böning A, Kügler S, Rhee J, Viotti JS, Dresbach T. S-SCAM is essential for synapse formation. Front Cell Neurosci 2023; 17:1182493. [PMID: 38045729 PMCID: PMC10690602 DOI: 10.3389/fncel.2023.1182493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 10/02/2023] [Indexed: 12/05/2023] Open
Abstract
Synapse formation is critical for the wiring of neural circuits in the developing brain. The synaptic scaffolding protein S-SCAM/MAGI-2 has important roles in the assembly of signaling complexes at post-synaptic densities. However, the role of S-SCAM in establishing the entire synapse is not known. Here, we report significant effects of RNAi-induced S-SCAM knockdown on the number of synapses in early stages of network development in vitro. In vivo knockdown during the first three postnatal weeks reduced the number of dendritic spines in the rat brain neocortex. Knockdown of S-SCAM in cultured hippocampal neurons severely reduced the clustering of both pre- and post-synaptic components. This included synaptic vesicle proteins, pre- and post-synaptic scaffolding proteins, and cell adhesion molecules, suggesting that entire synapses fail to form. Correspondingly, functional and morphological characteristics of developing neurons were affected by reducing S-SCAM protein levels; neurons displayed severely impaired synaptic transmission and reduced dendritic arborization. A next-generation sequencing approach showed normal expression of housekeeping genes but changes in expression levels in 39 synaptic signaling molecules in cultured neurons. These results indicate that S-SCAM mediates the recruitment of all key classes of synaptic molecules during synapse assembly and is critical for the development of neural circuits in the developing brain.
Collapse
Affiliation(s)
- Nina Wittenmayer
- Institute of Anatomy and Embryology, University Medical Center Göttingen, Göttingen, Germany
- Institute for Translational Medicine, MSH Medical School Hamburg, Hamburg, Germany
| | - Andonia Petkova-Tuffy
- Institute of Anatomy and Embryology, University Medical Center Göttingen, Göttingen, Germany
| | - Maximilian Borgmeyer
- Institute for Translational Medicine, MSH Medical School Hamburg, Hamburg, Germany
| | - Chungku Lee
- Department of Molecular Neurobiology, Synaptic Physiology Group, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Jürgen Becker
- Institute of Anatomy and Cell Biology, University Medical Center Göttingen, Göttingen, Germany
| | - Andreas Böning
- Institute of Anatomy and Embryology, University Medical Center Göttingen, Göttingen, Germany
| | - Sebastian Kügler
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - JeongSeop Rhee
- Department of Molecular Neurobiology, Synaptic Physiology Group, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Julio S. Viotti
- Institute of Anatomy and Embryology, University Medical Center Göttingen, Göttingen, Germany
- University of Bordeaux, CNRS, IINS, UMR 5297, Bordeaux, France
| | - Thomas Dresbach
- Institute of Anatomy and Embryology, University Medical Center Göttingen, Göttingen, Germany
| |
Collapse
|
9
|
Horovitz DDG, de Faria Domingues de Lima MA, Pires LDC, Campos Araujo ADQ, Vargas FR. Neurological Phenotypes of IRF2BPL Gene Variants: A Report of Four Novel Variants. J Cent Nerv Syst Dis 2023; 15:11795735231181467. [PMID: 37346291 PMCID: PMC10280516 DOI: 10.1177/11795735231181467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/23/2023] Open
Abstract
IRF2BPL gene variants have recently been associated to developmental disability and epilepsy in children and movement disorders in adults. So far, only few cases have been reported; here we present four novel cases identified by exome sequencing, while investigating developmental delay, adult-onset cerebellar ataxia or regression.
Collapse
Affiliation(s)
- Dafne Dain Gandelman Horovitz
- Medical Genetics Department, Instituto Nacional de Saúde da
Mulher, da Criança e do Adolescente Fernandes Figueira/FIOCRUZ, Rio de
Janeiro, Brazil
| | | | | | - Abelardo de Queiroz Campos Araujo
- Associated Professor Neurology, Universidade Federal do Rio de
Janeiro, Rio de Janeiro, Brazil
- Researcher, Instituto Nacional de Infectologia Evandro
Chagas, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Fernando Regla Vargas
- Medical Genetics, Gaffrée and Guinle University
Hospital, Rio de Janeiro, Brazil
- Genetics and Molecular Biology Department, Universidade Federal do Estado do Rio de
Janeiro, Rio de Janeiro, Brazil
- Epidemiology of congenital malformations
laboratory, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
- Instituto Nacional de Genética Médica
Populacional, Rio de Janeiro, Brazil
| |
Collapse
|
10
|
Yu C, Deng XJ, Xu D. Gene mutations in comorbidity of epilepsy and arrhythmia. J Neurol 2023; 270:1229-1248. [PMID: 36376730 DOI: 10.1007/s00415-022-11430-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 10/14/2022] [Accepted: 10/15/2022] [Indexed: 11/16/2022]
Abstract
Epilepsy is one of the most common neurological disorders, and sudden unexpected death in epilepsy (SUDEP) is the most severe outcome of refractory epilepsy. Arrhythmia is one of the heterogeneous factors in the pathophysiological mechanism of SUDEP with a high incidence in patients with refractory epilepsy, increasing the risk of premature death. The gene co-expressed in the brain and heart is supposed to be the genetic basis between epilepsy and arrhythmia, among which the gene encoding ion channel contributes to the prevalence of "cardiocerebral channelopathy" theory. Nevertheless, this theory could only explain the molecular mechanism of comorbid arrhythmia in part of patients with epilepsy (PWE). Therefore, we summarized the mutant genes that can induce comorbidity of epilepsy and arrhythmia and the possible corresponding treatments. These variants involved the genes encoding sodium, potassium, calcium and HCN channels, as well as some non-ion channel coding genes such as CHD4, PKP2, FHF1, GNB5, and mitochondrial genes. The relationship between genotype and clinical phenotype was not simple linear. Indeed, genes co-expressed in the brain and heart could independently induce epilepsy and/or arrhythmia. Mutant genes in brain could affect cardiac rhythm through central or peripheral regulation, while in the heart it could also affect cerebral electrical activity by changing the hemodynamics or internal environment. Analysis of mutations in comorbidity of epilepsy and arrhythmia could refine and expand the theory of "cardiocerebral channelopathy" and provide new insights for risk stratification of premature death and corresponding precision therapy in PWE.
Collapse
Affiliation(s)
- Cheng Yu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, Hubei Province, China
| | - Xue-Jun Deng
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, Hubei Province, China
| | - Da Xu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, Hubei Province, China.
| |
Collapse
|
11
|
Zhao K, Li Y, Yang X, Zhou L. The Impact of Altered HCN1 Expression on Brain Function and Its Relationship with Epileptogenesis. Curr Neuropharmacol 2023; 21:2070-2078. [PMID: 37366350 PMCID: PMC10556362 DOI: 10.2174/1570159x21666230214110333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/13/2022] [Accepted: 12/06/2022] [Indexed: 03/08/2023] Open
Abstract
Hyperpolarization-activated cyclic nucleotide-gated cation channel 1 (HCN1) is predominantly expressed in neurons from the neocortex and hippocampus, two important regions related to epilepsy. Both animal models for epilepsy and epileptic patients show decreased HCN1 expression and HCN1-mediated Ih current. It has been shown in neuroelectrophysiological experiments that a decreased Ih current can increase neuronal excitability. However, some studies have shown that blocking the Ih current in vivo can exert antiepileptic effects. This paradox raises an important question regarding the causal relationship between HCN1 alteration and epileptogenesis, which to date has not been elucidated. In this review, we summarize the literature related to HCN1 and epilepsy, aiming to find a possible explanation for this paradox, and explore the correlation between HCN1 and the mechanism of epileptogenesis. We analyze the alterations in the expression and distribution of HCN1 and the corresponding impact on brain function in epilepsy. In addition, we also discuss the effect of blocking Ih on epilepsy symptoms. Addressing these issues will help to inspire new strategies to explore the relationship between HCN1 and epileptogenesis, and ultimately promote the development of new targets for epilepsy therapy.
Collapse
Affiliation(s)
- Ke Zhao
- Department of Neurology, The Seventh Affliated Hospital of Sun Yet-sen University, No. 628, Zhenyuan Road, Xinhu Street, Guangming District, Shenzhen, China
| | - Yinchao Li
- Department of Neurology, The Seventh Affliated Hospital of Sun Yet-sen University, No. 628, Zhenyuan Road, Xinhu Street, Guangming District, Shenzhen, China
| | - Xiaofeng Yang
- Guangzhou Laboratory, Guangzhou, No. 9 XingDaoHuanBei Road, Guangzhou International Bio Island, Guangzhou 510005, Guangdong Province, China
| | - Liemin Zhou
- Department of Neurology, The Seventh Affliated Hospital of Sun Yet-sen University, No. 628, Zhenyuan Road, Xinhu Street, Guangming District, Shenzhen, China
| |
Collapse
|
12
|
Ozturk H, Basoglu H, Yorulmaz N, Aydin-Abidin S, Abidin I. Fisetin decreases the duration of ictal-like discharges in mouse hippocampal slices. J Biol Phys 2022; 48:355-368. [PMID: 35948819 PMCID: PMC9411310 DOI: 10.1007/s10867-022-09612-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 07/27/2022] [Indexed: 11/24/2022] Open
Abstract
There is an increasing interest in the biological and therapeutic effects of fisetin, a natural phenolic compound. Fisetin has affinity on some neuronal targets and may have the potential to modulate neuronal activity. In this study the effects of acute application of fisetin on synchronized events were evaluated electro-physiologically. Besides, interaction of fisetin with closely related channels were investigated in silico. Acute horizontal hippocampal slices were obtained from 32- to 36-day-old C57BL/6 mice. Extracellular field potentials were recorded from CA3 region of the hippocampus. Bath application of 4 aminopyridine (4AP, 100 µM) initiated ictal- and interictal-like synchronized epileptiform discharges in the brain slices. Fifty micromolar fisetin was applied to the recording chamber during the epileptiform activity. The duration and frequencies of both ictal-like and interictal-like activities were calculated from the electrophysiological records. Molecular docking was performed to reveal interaction of fisetin on GABA-A, NMDA, AMPA receptors, and HCN2 channel, which are neuronal structures directly involved in recorded activity. Although fisetin does not affect basal neuronal activity in brain slice, it reduced the duration of ictal-like discharges significantly. Molecular docking results indicated that fisetin has no effect on GABA-A, NMDA, and AMPA receptors. However, fisetin binds to the (5JON) HCN2 channel strongly with the binding energy of -7.66 kcal/mol. Reduction on the duration of 4AP-induced ictal-like discharges can be explained as HCN channels can cause an inhibitory effect via enhancing M-type K + channels which increase K outward currents.
Collapse
Affiliation(s)
- Hilal Ozturk
- Department of Biophysics, Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkey
- Department of Biophysics, Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Harun Basoglu
- Department of Biophysics, Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkey.
| | - Nuri Yorulmaz
- Department of Physics, Faculty of Science, Harran University, Sanliurfa, Turkey
| | - Selcen Aydin-Abidin
- Department of Biophysics, Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkey
| | - Ismail Abidin
- Department of Biophysics, Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkey
| |
Collapse
|
13
|
Zidoune H, Ladjouze A, Chellat-Rezgoune D, Boukri A, Dib SA, Nouri N, Tebibel M, Sifi K, Abadi N, Satta D, Benelmadani Y, Bignon-Topalovic J, El-Zaiat-Munsch M, Bashamboo A, McElreavey K. Novel Genomic Variants, Atypical Phenotypes and Evidence of a Digenic/Oligogenic Contribution to Disorders/Differences of Sex Development in a Large North African Cohort. Front Genet 2022; 13:900574. [PMID: 36110220 PMCID: PMC9468775 DOI: 10.3389/fgene.2022.900574] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Accepted: 05/23/2022] [Indexed: 11/13/2022] Open
Abstract
In a majority of individuals with disorders/differences of sex development (DSD) a genetic etiology is often elusive. However, new genes causing DSD are routinely reported and using the unbiased genomic approaches, such as whole exome sequencing (WES) should result in an increased diagnostic yield. Here, we performed WES on a large cohort of 125 individuals all of Algerian origin, who presented with a wide range of DSD phenotypes. The study excluded individuals with congenital adrenal hypoplasia (CAH) or chromosomal DSD. Parental consanguinity was reported in 36% of individuals. The genetic etiology was established in 49.6% (62/125) individuals of the total cohort, which includes 42.2% (35/83) of 46, XY non-syndromic DSD and 69.2% (27/39) of 46, XY syndromic DSD. No pathogenic variants were identified in the 46, XX DSD cases (0/3). Variants in the AR, HSD17B3, NR5A1 and SRD5A2 genes were the most common causes of DSD. Other variants were identified in genes associated with congenital hypogonadotropic hypogonadism (CHH), including the CHD7 and PROKR2. Previously unreported pathogenic/likely pathogenic variants (n = 30) involving 25 different genes were identified in 22.4% of the cohort. Remarkably 11.5% of the 46, XY DSD group carried variants classified as pathogenic/likely pathogenic variant in more than one gene known to cause DSD. The data indicates that variants in PLXNA3, a candidate CHH gene, is unlikely to be involved in CHH. The data also suggest that NR2F2 variants may cause 46, XY DSD.
Collapse
Affiliation(s)
- Housna Zidoune
- Human Developmental Genetics Unit, Institut Pasteur, CNRS, Paris, France
- Laboratory of Molecular and Cellular Biology, Department of Animal Biology, University Frères Mentouri Constantine 1, Constantine, Algeria
- Department of Medicine, Laboratory of Biology and Molecular Genetics, University Salah Boubnider Constantine 3, Constantine, Algeria
| | | | - Djalila Chellat-Rezgoune
- Laboratory of Molecular and Cellular Biology, Department of Animal Biology, University Frères Mentouri Constantine 1, Constantine, Algeria
- Department of Medicine, Laboratory of Biology and Molecular Genetics, University Salah Boubnider Constantine 3, Constantine, Algeria
| | - Asma Boukri
- Department of Endocrinology and Diabetology, CHU Ibn Badis Constantine, Constantine, Algeria
| | | | - Nassim Nouri
- Department of Endocrinology and Diabetology, CHU Ibn Badis Constantine, Constantine, Algeria
| | - Meryem Tebibel
- Department of Pediatric Surgery, CHU Beni Messous, Algiers, Algeria
| | - Karima Sifi
- Department of Medicine, Laboratory of Biology and Molecular Genetics, University Salah Boubnider Constantine 3, Constantine, Algeria
| | - Noureddine Abadi
- Department of Medicine, Laboratory of Biology and Molecular Genetics, University Salah Boubnider Constantine 3, Constantine, Algeria
| | - Dalila Satta
- Laboratory of Molecular and Cellular Biology, Department of Animal Biology, University Frères Mentouri Constantine 1, Constantine, Algeria
- Department of Medicine, Laboratory of Biology and Molecular Genetics, University Salah Boubnider Constantine 3, Constantine, Algeria
| | - Yasmina Benelmadani
- Department of Medicine, Laboratory of Biology and Molecular Genetics, University Salah Boubnider Constantine 3, Constantine, Algeria
| | | | | | - Anu Bashamboo
- Human Developmental Genetics Unit, Institut Pasteur, CNRS, Paris, France
| | - Ken McElreavey
- Human Developmental Genetics Unit, Institut Pasteur, CNRS, Paris, France
- *Correspondence: Ken McElreavey,
| |
Collapse
|
14
|
Majgaard J, Skov FG, Kim S, Hjortdal VE, Boedtkjer DMB. Positive chronotropic action of HCN channel antagonism in human collecting lymphatic vessels. Physiol Rep 2022; 10:e15401. [PMID: 35980021 PMCID: PMC9387113 DOI: 10.14814/phy2.15401] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 06/16/2022] [Accepted: 07/09/2022] [Indexed: 06/15/2023] Open
Abstract
Spontaneous action potentials precede phasic contractile activity in human collecting lymphatic vessels. In this study, we investigated the expression of hyperpolarization-activated cyclic nucleotide-gated (HCN) channels in human collecting lymphatics and by pharmacological inhibition ex vivo tested their potential role in controlling contractile function. Spontaneous and agonist-evoked tension changes of isolated thoracic duct and mesenteric lymphatic vessels-obtained from surgical patients with informed consent-were investigated by isometric myography, and ivabradine, ZD7288 or cesium were used to inhibit HCN. Analysis of HCN isoforms by RT-PCR and immunofluorescence revealed HCN2 to be the predominantly expressed mRNA isoform in human thoracic duct and mesenteric lymphatic vessels and HCN2-immunoreactivity confirmed protein expression in both vessel types. However, in functional experiments ex vivo the HCN inhibitors ivabradine, ZD7288, and cesium failed to lower contraction frequency: conversely, all three antagonists induced a positive chronotropic effect with concurrent negative inotropic action, though these effects first occurred at concentrations regarded as supramaximal for HCN inhibition. Based on these results, we conclude that human collecting vessels express HCN channel proteins but under the ex vivo experimental conditions described here HCN channels have little involvement in regulating contraction frequency in human collecting lymphatic vessels. Furthermore, HCN antagonists can produce concentration-dependent positive chronotropic and negative inotropic effects, which are apparently unrelated to HCN antagonism.
Collapse
Affiliation(s)
- Jens Majgaard
- Department of BiomedicineAarhus UniversityAarhusDenmark
| | | | - Sukhan Kim
- Department of BiomedicineAarhus UniversityAarhusDenmark
| | - Vibeke Elisabeth Hjortdal
- Department of Clinical MedicineAarhus UniversityAarhusDenmark
- Department of Cardiothoracic and Vascular SurgeryAarhus University HospitalAarhusDenmark
| | - Donna M. B. Boedtkjer
- Department of BiomedicineAarhus UniversityAarhusDenmark
- Department of Clinical MedicineAarhus UniversityAarhusDenmark
| |
Collapse
|
15
|
Lee S, Kim SY, Lim BC, Kim KJ, Chae JH, Cho A. Expanding the Clinical and Genetic Spectrum of Caveolinopathy in Korea. ANNALS OF CHILD NEUROLOGY 2022. [DOI: 10.26815/acn.2022.00136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Purpose: Caveolinopathy is a disease caused by caveolin-3 (CAV3) mutations that shows a wide clinical spectrum, including isolated hyperCKemia and limb-girdle muscular dystrophy. While recent advances in next-generation sequencing (NGS) have enabled earlier diagnosis of this disease, it remains difficult to predict the clinical course of each patient.Methods: This study summarizes the clinical presentations of 13 genetically confirmed caveolinopathy patients in four Korean families. Genetic diagnosis was performed using NGS technologies for probands and Sanger sequencing for the other family members.Results: Four coding mutations were found (p.Val103_Val104del, p.Asp28Glu, p.Pro105Leu, and p.Arg27Gln), and each family showed autosomal dominant inheritance. While all 13 cases had hyperCKemia, only five of them showed some myopathic features including ankle contracture, calf hypertrophy, exercise intolerance, and muscle cramping. This high proportion of asymptomatic cases suggests both that these mutations may be associated with a mild phenotype and that caveolinopathy may be an underdiagnosed disease.Conclusion: This study extends our understanding of caveolinopathy; in particular, the findings suggest the need to consider caveolinopathy in patients with incidental findings of CK elevation. NGS may be a useful method in the differential diagnosis of such cases.
Collapse
|
16
|
Peters CH, Singh RK, Bankston JR, Proenza C. Regulation of HCN Channels by Protein Interactions. Front Physiol 2022; 13:928507. [PMID: 35795651 PMCID: PMC9251338 DOI: 10.3389/fphys.2022.928507] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 06/01/2022] [Indexed: 11/30/2022] Open
Abstract
Hyperpolarization-activated, cyclic nucleotide-sensitive (HCN) channels are key regulators of subthreshold membrane potentials in excitable cells. The four mammalian HCN channel isoforms, HCN1-HCN4, are expressed throughout the body, where they contribute to diverse physiological processes including cardiac pacemaking, sleep-wakefulness cycles, memory, and somatic sensation. While all HCN channel isoforms produce currents when expressed by themselves, an emerging list of interacting proteins shape HCN channel excitability to influence the physiologically relevant output. The best studied of these regulatory proteins is the auxiliary subunit, TRIP8b, which binds to multiple sites in the C-terminus of the HCN channels to regulate expression and disrupt cAMP binding to fine-tune neuronal HCN channel excitability. Less is known about the mechanisms of action of other HCN channel interaction partners like filamin A, Src tyrosine kinase, and MinK-related peptides, which have a range of effects on HCN channel gating and expression. More recently, the inositol trisphosphate receptor-associated cGMP-kinase substrates IRAG1 and LRMP (also known as IRAG2), were discovered as specific regulators of the HCN4 isoform. This review summarizes the known protein interaction partners of HCN channels and their mechanisms of action and identifies gaps in our knowledge.
Collapse
Affiliation(s)
- Colin H. Peters
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Rohit K. Singh
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - John R. Bankston
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Catherine Proenza
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Department of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- *Correspondence: Catherine Proenza,
| |
Collapse
|
17
|
Inflammation in pediatric epilepsies: Update on clinical features and treatment options. Epilepsy Behav 2022; 131:107959. [PMID: 33867302 DOI: 10.1016/j.yebeh.2021.107959] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 03/22/2021] [Accepted: 03/24/2021] [Indexed: 02/08/2023]
Abstract
The role of inflammation is increasingly recognized in triggering or sustaining epileptic activity. In the last decades, increasing research has provided definite evidence to support the link between immunity, inflammatory process, and epilepsy. Neuro- and systemic inflammation play a pivotal role in driving epileptogenesis through different pathogenetic mechanisms: the activation of innate immunity in glia, neurons, and microvasculature, the brain mediated by blood-brain barrier (BBB) impairment, and the imbalance of pro- and anti-inflammatory molecules produced by both arms of immunity. More recently, research has focused on the adverse effects of maternal or early-life immune activation and cytokine imbalance on fetal neurodevelopment and postnatal epilepsy. A complex crosstalk between the immune and nervous system, and a crucial interplay of genetic, epigenetic, and environmental factors may influence structures and functions of the developing brain. A better understanding of the inflammatory process in promoting epilepsy implies that targeting specific pathways may be effective in seizure control. Multiple targets have been identified so far, and several antiseizure interventions are obtained by inhibiting inflammatory signaling or protecting/restoring BBB. All this evidence has changed the field of epilepsy research and neuropharmacology. Further developments and new treatments will rapidly emerge to improve seizure management in inflammation-related epilepsies. This article is part of the Special Issue "Severe Infantile Epilepsies".
Collapse
|
18
|
Kessi M, Peng J, Duan H, He H, Chen B, Xiong J, Wang Y, Yang L, Wang G, Kiprotich K, Bamgbade OA, He F, Yin F. The Contribution of HCN Channelopathies in Different Epileptic Syndromes, Mechanisms, Modulators, and Potential Treatment Targets: A Systematic Review. Front Mol Neurosci 2022; 15:807202. [PMID: 35663267 PMCID: PMC9161305 DOI: 10.3389/fnmol.2022.807202] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 04/06/2022] [Indexed: 12/04/2022] Open
Abstract
Background Hyperpolarization-activated cyclic nucleotide-gated (HCN) current reduces dendritic summation, suppresses dendritic calcium spikes, and enables inhibitory GABA-mediated postsynaptic potentials, thereby suppressing epilepsy. However, it is unclear whether increased HCN current can produce epilepsy. We hypothesized that gain-of-function (GOF) and loss-of-function (LOF) variants of HCN channel genes may cause epilepsy. Objectives This systematic review aims to summarize the role of HCN channelopathies in epilepsy, update genetic findings in patients, create genotype–phenotype correlations, and discuss animal models, GOF and LOF mechanisms, and potential treatment targets. Methods The review was conducted in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analyses statement, for all years until August 2021. Results We identified pathogenic variants of HCN1 (n = 24), HCN2 (n = 8), HCN3 (n = 2), and HCN4 (n = 6) that were associated with epilepsy in 74 cases (43 HCN1, 20 HCN2, 2 HCN3, and 9 HCN4). Epilepsy was associated with GOF and LOF variants, and the mechanisms were indeterminate. Less than half of the cases became seizure-free and some developed drug-resistant epilepsy. Of the 74 cases, 12 (16.2%) died, comprising HCN1 (n = 4), HCN2 (n = 2), HCN3 (n = 2), and HCN4 (n = 4). Of the deceased cases, 10 (83%) had a sudden unexpected death in epilepsy (SUDEP) and 2 (16.7%) due to cardiopulmonary failure. SUDEP affected more adults (n = 10) than children (n = 2). HCN1 variants p.M234R, p.C329S, p.V414M, p.M153I, and p.M305L, as well as HCN2 variants p.S632W and delPPP (p.719–721), were associated with different phenotypes. HCN1 p.L157V and HCN4 p.R550C were associated with genetic generalized epilepsy. There are several HCN animal models, pharmacological targets, and modulators, but precise drugs have not been developed. Currently, there are no HCN channel openers. Conclusion We recommend clinicians to include HCN genes in epilepsy gene panels. Researchers should explore the possible underlying mechanisms for GOF and LOF variants by identifying the specific neuronal subtypes and neuroanatomical locations of each identified pathogenic variant. Researchers should identify specific HCN channel openers and blockers with high binding affinity. Such information will give clarity to the involvement of HCN channelopathies in epilepsy and provide the opportunity to develop targeted treatments.
Collapse
Affiliation(s)
- Miriam Kessi
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China
- Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
- Department of Pediatrics, Kilimanjaro Christian Medical University College, Moshi, Tanzania
| | - Jing Peng
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China
- Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
| | - Haolin Duan
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China
- Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
| | - Hailan He
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China
- Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
| | - Baiyu Chen
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China
- Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
| | - Juan Xiong
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China
- Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
| | - Ying Wang
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China
- Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
| | - Lifen Yang
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China
- Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
| | - Guoli Wang
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China
- Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
| | - Karlmax Kiprotich
- Department of Epidemiology and Medical Statistics, School of Public Health, Moi University, Eldoret, Kenya
| | - Olumuyiwa A. Bamgbade
- Department of Anesthesiology and Pharmacology, University of British Columbia, Vancouver, BC, Canada
| | - Fang He
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China
- Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
| | - Fei Yin
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China
- Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
- *Correspondence: Fei Yin
| |
Collapse
|
19
|
Porro A, Abbandonato G, Veronesi V, Russo A, Binda A, Antolini L, Granata T, Castellotti B, Marini C, Moroni A, DiFrancesco JC, Rivolta I. Do the functional properties of HCN1 mutants correlate with the clinical features in epileptic patients? PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2021; 166:147-155. [PMID: 34310985 DOI: 10.1016/j.pbiomolbio.2021.07.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 06/14/2021] [Accepted: 07/20/2021] [Indexed: 10/20/2022]
Abstract
The altered function of the Hyperpolarization-activated Cyclic-Nucleotide-gated (HCN) ion channels plays an important role in the pathogenesis of epilepsy in humans. In particular, HCN1 missense mutations have been recently identified in patients with different epileptic phenotypes, varying from mild to severe. Their electrophysiological characterization shows that mutated channels can act both with loss-of-function and gain-of-function mechanisms of action, without an evident correlation with the phenotype. In search for a correlation between clinical features and biophysical properties of the mutations, in this work we considered sixteen HCN1 mutations, found in eighteen Early Infantile Epileptic Encephalopathy (EIEE) patients. Statistical analysis did not establish any significant correlation between the clinical parameters and the current properties of the mutant channels. The lack of significance of our results could depend on the small number of mutations analyzed, epilepsy-associated with certainty. With the progressive increase of Next Generation Sequencing in patients with early-onset epilepsy, it is expected that the number of patients with HCN1 mutations will grow steadily. Functional characterization of epilepsy-associated HCN1 mutations remains a fundamental tool for a better understanding of the pathogenetic mechanisms leading to the disease in humans.
Collapse
Affiliation(s)
| | | | - Valentina Veronesi
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy.
| | - Alberto Russo
- Department of Biosciences, University of Milan, Milan, Italy.
| | - Anna Binda
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy.
| | - Laura Antolini
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy.
| | - Tiziana Granata
- Department of Pediatric Neuroscience, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy.
| | - Barbara Castellotti
- Unit of Genetics of Neurodegenerative and Metabolic Diseases, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy.
| | - Carla Marini
- Department of Child Neuropsychiatry, Children's Hospital, Ancona, Italy.
| | - Anna Moroni
- Department of Biosciences, University of Milan, Milan, Italy.
| | - Jacopo C DiFrancesco
- Department of Pediatric Neuroscience, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy; Department of Neurology, Epilepsy Center, ASST San Gerardo Hospital, University of Milano- Bicocca, Monza, Italy.
| | - Ilaria Rivolta
- School of Medicine and Surgery and Milan Center for Neuroscience (NeuroMI), University of Milano-Bicocca, Monza, Italy.
| |
Collapse
|
20
|
Dwivedi D, Bhalla US. Physiology and Therapeutic Potential of SK, H, and M Medium AfterHyperPolarization Ion Channels. Front Mol Neurosci 2021; 14:658435. [PMID: 34149352 PMCID: PMC8209339 DOI: 10.3389/fnmol.2021.658435] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 04/13/2021] [Indexed: 12/19/2022] Open
Abstract
SK, HCN, and M channels are medium afterhyperpolarization (mAHP)-mediating ion channels. The three channels co-express in various brain regions, and their collective action strongly influences cellular excitability. However, significant diversity exists in the expression of channel isoforms in distinct brain regions and various subcellular compartments, which contributes to an equally diverse set of specific neuronal functions. The current review emphasizes the collective behavior of the three classes of mAHP channels and discusses how these channels function together although they play specialized roles. We discuss the biophysical properties of these channels, signaling pathways that influence the activity of the three mAHP channels, various chemical modulators that alter channel activity and their therapeutic potential in treating various neurological anomalies. Additionally, we discuss the role of mAHP channels in the pathophysiology of various neurological diseases and how their modulation can alleviate some of the symptoms.
Collapse
Affiliation(s)
- Deepanjali Dwivedi
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, GKVK Campus, Bengaluru, India.,Department of Neurobiology, Harvard Medical School, Boston, MA, United States.,Stanley Center at the Broad, Cambridge, MA, United States
| | - Upinder S Bhalla
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, GKVK Campus, Bengaluru, India
| |
Collapse
|
21
|
John A, Ng-Cordell E, Hanna N, Brkic D, Baker K. The neurodevelopmental spectrum of synaptic vesicle cycling disorders. J Neurochem 2021; 157:208-228. [PMID: 32738165 DOI: 10.1111/jnc.15135] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 07/20/2020] [Accepted: 07/21/2020] [Indexed: 12/11/2022]
Abstract
In this review, we describe and discuss neurodevelopmental phenotypes arising from rare, high penetrance genomic variants which directly influence synaptic vesicle cycling (SVC disorders). Pathogenic variants in each SVC disorder gene lead to disturbance of at least one SVC subprocess, namely vesicle trafficking (e.g. KIF1A and GDI1), clustering (e.g. TRIO, NRXN1 and SYN1), docking and priming (e.g. STXBP1), fusion (e.g. SYT1 and PRRT2) or re-uptake (e.g. DNM1, AP1S2 and TBC1D24). We observe that SVC disorders share a common set of neurological symptoms (movement disorders, epilepsies), cognitive impairments (developmental delay, intellectual disabilities, cerebral visual impairment) and mental health difficulties (autism, ADHD, psychiatric symptoms). On the other hand, there is notable phenotypic variation between and within disorders, which may reflect selective disruption to SVC subprocesses, spatiotemporal and cell-specific gene expression profiles, mutation-specific effects, or modifying factors. Understanding the common cellular and systems mechanisms underlying neurodevelopmental phenotypes in SVC disorders, and the factors responsible for variation in clinical presentations and outcomes, may translate to personalized clinical management and improved quality of life for patients and families.
Collapse
Affiliation(s)
- Abinayah John
- MRC Cognition and Brain Sciences Unit, University of Cambridge, Cambridge, UK
| | - Elise Ng-Cordell
- MRC Cognition and Brain Sciences Unit, University of Cambridge, Cambridge, UK
| | - Nancy Hanna
- MRC Cognition and Brain Sciences Unit, University of Cambridge, Cambridge, UK
| | - Diandra Brkic
- MRC Cognition and Brain Sciences Unit, University of Cambridge, Cambridge, UK
| | - Kate Baker
- MRC Cognition and Brain Sciences Unit, University of Cambridge, Cambridge, UK
| |
Collapse
|
22
|
Concepcion FA, Khan MN, Ju Wang JD, Wei AD, Ojemann JG, Ko AL, Shi Y, Eng JK, Ramirez JM, Poolos NP. HCN Channel Phosphorylation Sites Mapped by Mass Spectrometry in Human Epilepsy Patients and in an Animal Model of Temporal Lobe Epilepsy. Neuroscience 2021; 460:13-30. [PMID: 33571596 DOI: 10.1016/j.neuroscience.2021.01.038] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/07/2021] [Accepted: 01/26/2021] [Indexed: 10/22/2022]
Abstract
Because hyperpolarization-activated cyclic nucleotide-gated (HCN) ion channels modulate the excitability of cortical and hippocampal principal neurons, these channels play a key role in the hyperexcitability that occurs during the development of epilepsy after a brain insult, or epileptogenesis. In epileptic rats generated by pilocarpine-induced status epilepticus, HCN channel activity is downregulated by two main mechanisms: a hyperpolarizing shift in gating and a decrease in amplitude of the current mediated by HCN channels, Ih. Because these mechanisms are modulated by various phosphorylation signaling pathways, we hypothesized that phosphorylation changes occur at individual HCN channel amino acid residues (phosphosites) during epileptogenesis. We collected CA1 hippocampal tissue from male Sprague Dawley rats made epileptic by pilocarpine-induced status epilepticus, and age-matched naïve controls. We also included resected human brain tissue containing epileptogenic zones (EZs) where seizures arise for comparison to our chronically epileptic rats. After enrichment for HCN1 and HCN2 isoforms by immunoprecipitation and trypsin in-gel digestion, the samples were analyzed by mass spectrometry. We identified numerous phosphosites from HCN1 and HCN2 channels, representing a novel survey of phosphorylation sites within HCN channels. We found high levels of HCN channel phosphosite homology between humans and rats. We also identified a novel HCN1 channel phosphosite S791, which underwent significantly increased phosphorylation during the chronic epilepsy stage. Heterologous expression of a phosphomimetic mutant, S791D, replicated a hyperpolarizing shift in Ih gating seen in neurons from chronically epileptic rats. These results show that HCN1 channel phosphorylation is altered in epilepsy and may be of pathogenic importance.
Collapse
Affiliation(s)
- F A Concepcion
- Department of Neurology and Regional Epilepsy Center, University of Washington, Seattle, WA, United States
| | - M N Khan
- Department of Neurology and Regional Epilepsy Center, University of Washington, Seattle, WA, United States
| | - J-D Ju Wang
- Seattle Children's Research Institute, Center for Integrative Brain Research, Seattle, WA, United States
| | - A D Wei
- Seattle Children's Research Institute, Center for Integrative Brain Research, Seattle, WA, United States
| | - J G Ojemann
- Seattle Children's Research Institute, Center for Integrative Brain Research, Seattle, WA, United States; Department of Neurological Surgery, University of Washington, Seattle, WA, United States
| | - A L Ko
- Department of Neurological Surgery, University of Washington, Seattle, WA, United States
| | - Y Shi
- Department of Electrical and Computer Engineering, University of Washington, Seattle, WA, United States
| | - J K Eng
- Proteomics Resource, University of Washington, Seattle, WA, United States
| | - J-M Ramirez
- Seattle Children's Research Institute, Center for Integrative Brain Research, Seattle, WA, United States; Department of Neurological Surgery, University of Washington, Seattle, WA, United States
| | - N P Poolos
- Department of Neurology and Regional Epilepsy Center, University of Washington, Seattle, WA, United States.
| |
Collapse
|
23
|
Hemizygous FLNA variant in West syndrome without periventricular nodular heterotopia. Hum Genome Var 2020; 7:43. [PMID: 33298907 PMCID: PMC7713383 DOI: 10.1038/s41439-020-00131-9] [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: 09/08/2020] [Revised: 10/29/2020] [Accepted: 10/29/2020] [Indexed: 11/21/2022] Open
Abstract
Pathogenic FLNA variants can be identified in patients with seizures accompanied by periventricular nodular heterotopia (PVNH). It is unusual to find FLNA aberrations in epileptic patients without PVNH on brain imaging. We report a boy with cryptogenic West syndrome followed by refractory seizures and psychomotor delay. We performed whole-exome sequencing and identified a de novo missense variant in FLNA. It is noteworthy that this patient showed no PVNH. As no other pathogenic variants were found in epilepsy-related genes, this FLNA variant likely caused West syndrome but with no PVNH.
Collapse
|
24
|
Han Y, Lyman KA, Foote KM, Chetkovich DM. The structure and function of TRIP8b, an auxiliary subunit of hyperpolarization-activated cyclic-nucleotide gated channels. Channels (Austin) 2020; 14:110-122. [PMID: 32189562 PMCID: PMC7153792 DOI: 10.1080/19336950.2020.1740501] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 02/18/2020] [Accepted: 02/21/2020] [Indexed: 02/08/2023] Open
Abstract
Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels are expressed throughout the mammalian central nervous system (CNS). These channels have been implicated in a wide range of diseases, including Major Depressive Disorder and multiple subtypes of epilepsy. The diversity of functions that HCN channels perform is in part attributable to differences in their subcellular localization. To facilitate a broad range of subcellular distributions, HCN channels are bound by auxiliary subunits that regulate surface trafficking and channel function. One of the best studied auxiliary subunits is tetratricopeptide-repeat containing, Rab8b-interacting protein (TRIP8b). TRIP8b is an extensively alternatively spliced protein whose only known function is to regulate HCN channels. TRIP8b binds to HCN pore-forming subunits at multiple interaction sites that differentially regulate HCN channel function and subcellular distribution. In this review, we summarize what is currently known about the structure and function of TRIP8b isoforms with an emphasis on the role of this auxiliary subunit in health and disease.
Collapse
Affiliation(s)
- Ye Han
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Kyle A. Lyman
- Department of Neurology, Stanford University, Palo Alto, CA, USA
| | - Kendall M. Foote
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Dane M. Chetkovich
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
25
|
Davydova YD, Kazantseva AV, Enikeeva RF, Mustafin RN, Lobaskova MM, Malykh SB, Gilyazova IR, Khusnutdinova EK. The Role of Oxytocin Receptor (OXTR) Gene Polymorphisms in the Development of Aggressive Behavior in Healthy Individuals. RUSS J GENET+ 2020. [DOI: 10.1134/s1022795420090057] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
26
|
D'Adamo MC, Liantonio A, Conte E, Pessia M, Imbrici P. Ion Channels Involvement in Neurodevelopmental Disorders. Neuroscience 2020; 440:337-359. [PMID: 32473276 DOI: 10.1016/j.neuroscience.2020.05.032] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 05/16/2020] [Accepted: 05/19/2020] [Indexed: 12/14/2022]
Abstract
Inherited and sporadic mutations in genes encoding for brain ion channels, affecting membrane expression or biophysical properties, have been associated with neurodevelopmental disorders characterized by epilepsy, cognitive and behavioral deficits with significant phenotypic and genetic heterogeneity. Over the years, the screening of a growing number of patients and the functional characterization of newly identified mutations in ion channels genes allowed to recognize new phenotypes and to widen the clinical spectrum of known diseases. Furthermore, advancements in understanding disease pathogenesis at atomic level or using patient-derived iPSCs and animal models have been pivotal to orient therapeutic intervention and to put the basis for the development of novel pharmacological options for drug-resistant disorders. In this review we will discuss major improvements and critical issues concerning neurodevelopmental disorders caused by dysfunctions in brain sodium, potassium, calcium, chloride and ligand-gated ion channels.
Collapse
Affiliation(s)
- Maria Cristina D'Adamo
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, Malta
| | | | - Elena Conte
- Department of Pharmacy-Drug Sciences, University of Bari "Aldo Moro", Italy
| | - Mauro Pessia
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, Malta; Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Paola Imbrici
- Department of Pharmacy-Drug Sciences, University of Bari "Aldo Moro", Italy.
| |
Collapse
|
27
|
Rivolta I, Binda A, Masi A, DiFrancesco JC. Cardiac and neuronal HCN channelopathies. Pflugers Arch 2020; 472:931-951. [PMID: 32424620 DOI: 10.1007/s00424-020-02384-3] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 04/21/2020] [Accepted: 04/24/2020] [Indexed: 12/31/2022]
Abstract
Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels are expressed as four different isoforms (HCN1-4) in the heart and in the central and peripheral nervous systems. In the voltage range of activation, HCN channels carry an inward current mediated by Na+ and K+, termed If in the heart and Ih in neurons. Altered function of HCN channels, mainly HCN4, is associated with sinus node dysfunction and other arrhythmias such as atrial fibrillation, ventricular tachycardia, and atrioventricular block. In recent years, several data have also shown that dysfunctional HCN channels, in particular HCN1, but also HCN2 and HCN4, can play a pathogenic role in epilepsy; these include experimental data from animal models, and data collected over genetic mutations of the channels identified and characterized in epileptic patients. In the central nervous system, alteration of the Ih current could predispose to the development of neurodegenerative diseases such as Parkinson's disease; since HCN channels are widely expressed in the peripheral nervous system, their dysfunctional behavior could also be associated with the pathogenesis of neuropathic pain. Given the fundamental role played by the HCN channels in the regulation of the discharge activity of cardiac and neuronal cells, the modulation of their function for therapeutic purposes is under study since it could be useful in various pathological conditions. Here we review the present knowledge of the HCN-related channelopathies in cardiac and neurological diseases, including clinical, genetic, therapeutic, and physiopathological aspects.
Collapse
Affiliation(s)
- Ilaria Rivolta
- School of Medicine and Surgery, Milan Center for Neuroscience (NeuroMI), University of Milano-Bicocca, Monza, Italy
| | - Anna Binda
- School of Medicine and Surgery, Milan Center for Neuroscience (NeuroMI), University of Milano-Bicocca, Monza, Italy
| | - Alessio Masi
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), section of Pharmacology and Toxicology, University of Florence, Florence, Italy
| | - Jacopo C DiFrancesco
- School of Medicine and Surgery, Milan Center for Neuroscience (NeuroMI), University of Milano-Bicocca, Monza, Italy. .,Department of Neurology, ASST San Gerardo Hospital, University of Milano-Bicocca, Via Pergolesi, 33, 20900, Monza, MB, Italy.
| |
Collapse
|
28
|
Bartolini E, Campostrini R, Kiferle L, Pradella S, Rosati E, Chinthapalli K, Palumbo P. Epilepsy and brain channelopathies from infancy to adulthood. Neurol Sci 2019; 41:749-761. [PMID: 31838630 DOI: 10.1007/s10072-019-04190-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 12/06/2019] [Indexed: 01/04/2023]
Abstract
Genetic brain channelopathies result from inherited or de novo mutations of genes encoding ion channel subunits within the central nervous system. Most neurological channelopathies arise in childhood with paroxysmal or episodic symptoms, likely because of a transient impairment of homeostatic mechanisms regulating membrane excitability, and the prototypical expression of this impairment is epilepsy. Migraine, episodic ataxia and alternating hemiplegia can also occur, as well as chronic phenotypes, such as spinocerebellar ataxias, intellectual disability and autism spectrum disorder. Voltage-gated and ligand-gated channels may be involved. In most cases, a single gene may be associated with a phenotypical spectrum that shows variable expressivity. Different clinical features may arise at different ages and the adult phenotype may be remarkably modified from the syndrome onset in childhood or adolescence. Recognizing the prominent phenotypical traits of brain channelopathies is essential to perform appropriate diagnostic investigations and to provide the better care not only in the paediatric setting but also for adult patients and their caregivers. Herein, we provide an overview of genetic brain channelopathies associated with epilepsy, highlight the different molecular mechanisms and describe the different clinical characteristics which may prompt the clinician to suspect specific syndromes and to possibly establish tailored treatments.
Collapse
Affiliation(s)
- Emanuele Bartolini
- USL Centro Toscana, Neurology Unit, Nuovo Ospedale Santo Stefano, Via Suor Niccolina Infermiera 20, 59100, Prato, Italy.
| | - Roberto Campostrini
- USL Centro Toscana, Neurology Unit, Nuovo Ospedale Santo Stefano, Via Suor Niccolina Infermiera 20, 59100, Prato, Italy
| | - Lorenzo Kiferle
- USL Centro Toscana, Neurology Unit, Nuovo Ospedale Santo Stefano, Via Suor Niccolina Infermiera 20, 59100, Prato, Italy
| | - Silvia Pradella
- USL Centro Toscana, Neurology Unit, Nuovo Ospedale Santo Stefano, Via Suor Niccolina Infermiera 20, 59100, Prato, Italy
| | - Eleonora Rosati
- USL Centro Toscana, Neurology Unit, Nuovo Ospedale Santo Stefano, Via Suor Niccolina Infermiera 20, 59100, Prato, Italy
| | | | - Pasquale Palumbo
- USL Centro Toscana, Neurology Unit, Nuovo Ospedale Santo Stefano, Via Suor Niccolina Infermiera 20, 59100, Prato, Italy
| |
Collapse
|
29
|
Foote KM, Lyman KA, Han Y, Michailidis IE, Heuermann RJ, Mandikian D, Trimmer JS, Swanson GT, Chetkovich DM. Phosphorylation of the HCN channel auxiliary subunit TRIP8b is altered in an animal model of temporal lobe epilepsy and modulates channel function. J Biol Chem 2019; 294:15743-15758. [PMID: 31492750 DOI: 10.1074/jbc.ra119.010027] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 08/30/2019] [Indexed: 12/14/2022] Open
Abstract
Temporal lobe epilepsy (TLE) is a prevalent neurological disorder with many patients experiencing poor seizure control with existing anti-epileptic drugs. Thus, novel insights into the mechanisms of epileptogenesis and identification of new drug targets can be transformative. Changes in ion channel function have been shown to play a role in generating the aberrant neuronal activity observed in TLE. Previous work demonstrates that hyperpolarization-activated cyclic nucleotide-gated (HCN) channels regulate neuronal excitability and are mislocalized within CA1 pyramidal cells in a rodent model of TLE. The subcellular distribution of HCN channels is regulated by an auxiliary subunit, tetratricopeptide repeat-containing Rab8b-interacting protein (TRIP8b), and disruption of this interaction correlates with channel mislocalization. However, the molecular mechanisms responsible for HCN channel dysregulation in TLE are unclear. Here we investigated whether changes in TRIP8b phosphorylation are sufficient to alter HCN channel function. We identified a phosphorylation site at residue Ser237 of TRIP8b that enhances binding to HCN channels and influences channel gating by altering the affinity of TRIP8b for the HCN cytoplasmic domain. Using a phosphospecific antibody, we demonstrate that TRIP8b phosphorylated at Ser237 is enriched in CA1 distal dendrites and that phosphorylation is reduced in the kainic acid model of TLE. Overall, our findings indicate that the TRIP8b-HCN interaction can be modulated by changes in phosphorylation and suggest that loss of TRIP8b phosphorylation may affect HCN channel properties during epileptogenesis. These results highlight the potential of drugs targeting posttranslational modifications to restore TRIP8b phosphorylation to reduce excitability in TLE.
Collapse
Affiliation(s)
- Kendall M Foote
- Davee Department of Neurology and Clinical Neurosciences, Northwestern University, Chicago, Illinois 60611.,Department of Pharmacology, Northwestern University, Chicago, Illinois 60611.,Vanderbilt University Medical Center Department of Neurology, Nashville, Tennessee 37232
| | - Kyle A Lyman
- Davee Department of Neurology and Clinical Neurosciences, Northwestern University, Chicago, Illinois 60611.,Vanderbilt University Medical Center Department of Neurology, Nashville, Tennessee 37232.,Department of Medicine, Stanford University, Palo Alto, California 94305
| | - Ye Han
- Davee Department of Neurology and Clinical Neurosciences, Northwestern University, Chicago, Illinois 60611.,Vanderbilt University Medical Center Department of Neurology, Nashville, Tennessee 37232
| | - Ioannis E Michailidis
- Vanderbilt University Medical Center Department of Neurology, Nashville, Tennessee 37232
| | - Robert J Heuermann
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Danielle Mandikian
- Department of Neurobiology, Physiology, and Behavior, University of California, Davis, California 95616
| | - James S Trimmer
- Department of Neurobiology, Physiology, and Behavior, University of California, Davis, California 95616.,Department of Physiology and Membrane Biology, University of California, Davis, California 95616
| | - Geoffrey T Swanson
- Department of Pharmacology, Northwestern University, Chicago, Illinois 60611.,Department of Neurobiology, Northwestern University, Evanston, Illinois 60208
| | - Dane M Chetkovich
- Vanderbilt University Medical Center Department of Neurology, Nashville, Tennessee 37232
| |
Collapse
|
30
|
Oyrer J, Bleakley LE, Richards KL, Maljevic S, Phillips AM, Petrou S, Nowell CJ, Reid CA. Using a Multiplex Nucleic Acid in situ Hybridization Technique to Determine HCN4 mRNA Expression in the Adult Rodent Brain. Front Mol Neurosci 2019; 12:211. [PMID: 31555092 PMCID: PMC6724756 DOI: 10.3389/fnmol.2019.00211] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 08/16/2019] [Indexed: 12/28/2022] Open
Abstract
Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels carry a non-selective cationic conductance, Ih, which is important for modulating neuron excitability. Four genes (HCN1-4) encode HCN channels, with each gene having distinct expression and biophysical profiles. Here we use multiplex nucleic acid in situ hybridization to determine HCN4 mRNA expression within the adult mouse brain. We take advantage of this approach to detect HCN4 mRNA simultaneously with either HCN1 or HCN2 mRNA and markers of excitatory (VGlut-positive) and inhibitory (VGat-positive) neurons, which was not previously reported. We have developed a Fiji-based analysis code that enables quantification of mRNA expression within identified cell bodies. The highest HCN4 mRNA expression was found in the habenula (medial and lateral) and the thalamus. HCN4 mRNA was particularly high in the medial habenula with essentially no co-expression of HCN1 or HCN2 mRNA. An absence of Ih-mediated “sag” in neurons recorded from the medial habenula of knockout mice confirmed that HCN4 channels are the predominant subtype in this region. Analysis in the thalamus revealed HCN4 mRNA in VGlut2-positive excitatory neurons that was always co-expressed with HCN2 mRNA. In contrast, HCN4 mRNA was undetectable in the nucleus reticularis. HCN4 mRNA expression was high in a subset of VGat-positive cells in the globus pallidus external. The majority of these neurons co-expressed HCN2 mRNA while a smaller subset also co-expressed HCN1 mRNA. In the striatum, a small subset of large cells which are likely to be giant cholinergic interneurons co-expressed high levels of HCN4 and HCN2 mRNA. The amygdala, cortex and hippocampus expressed low levels of HCN4 mRNA. This study highlights the heterogeneity of HCN4 mRNA expression in the brain and provides a morphological framework on which to better investigate the functional roles of HCN4 channels.
Collapse
Affiliation(s)
- Julia Oyrer
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Lauren E Bleakley
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Kay L Richards
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Snezana Maljevic
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, Australia
| | - A Marie Phillips
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, Australia.,School of Biosciences, The University of Melbourne, Parkville, VIC, Australia
| | - Steven Petrou
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Cameron J Nowell
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Christopher A Reid
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|