1
|
Zhao X, Ning H, Liu L, Zhu C, Zhang Y, Sun G, Ren H, Kong X. Genetic analysis of 37 cases with primary periodic paralysis in Chinese patients. Orphanet J Rare Dis 2024; 19:160. [PMID: 38609989 PMCID: PMC11015673 DOI: 10.1186/s13023-024-03170-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 03/30/2024] [Indexed: 04/14/2024] Open
Abstract
BACKGROUND Primary periodic paralysis (PPP) is an inherited disorders of ion channel dysfunction characterized by recurrent episodes of flaccid muscle weakness, which can classified as hypokalemic (HypoPP), normokalemic (NormoPP), or hyperkalemic (HyperPP) according to the potassium level during the paralytic attacks. However, PPP is charactered by remarkable clinical and genetic heterogeneity, and the diagnosis of suspected patients is based on the characteristic clinical presentation then confirmed by genetic testing. At present, there are only limited cohort studies on PPP in the Chinese population. RESULTS We included 37 patients with a clinical diagnosis of PPP. Eleven (29.7%) patients were tested using a specific gene panel and 26 (70.3%) by the whole-exome sequencing (WES). Twenty-two cases had a genetic variant identified, representing a diagnostic rate of 59.5% (22/37). All the identified mutations were either in the SCN4A or the CACNA1S gene. The overall detection rate was comparable between the panel (54.5%: 6/11) and WES (61.5%: 16/26). The remaining patients unresolved through panel sequencing were further analyzed by WES, without the detection of any mutation. The novel atypical splicing variant c.2020-5G > A affects the normal splicing of the SCN4A mRNA, which was confirmed by minigene splicing assay. Among 21 patients with HypoPP, 15 patients were classified as HypoPP-2 with SCN4A variants, and 6 HypoPP-1 patients had CACNA1S variants. CONCLUSIONS Our results suggest that SCN4A alleles are the main cause in our cohort, with the remainder caused by CACNA1S alleles, which are the predominant cause in Europe and the United States. Additionally, this study identified 3 novel SCN4A and 2 novel CACNA1S variants, broadening the mutation spectrum of genes associated with PPP.
Collapse
Affiliation(s)
- Xuechao Zhao
- The Genetics and Prenatal Diagnosis Center, The Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Jianshe Rd, Erqi District, 450052, Zhengzhou, Henan, China
| | - Haofeng Ning
- Obstetrics and Gynaecology, The Seventh Affiliated Hospital, Sun Yat-Sen University, No 628 Zhenyuan Road Guangming District, 518107, Shenzhen, PR China
| | - Lina Liu
- The Genetics and Prenatal Diagnosis Center, The Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Jianshe Rd, Erqi District, 450052, Zhengzhou, Henan, China
| | - Chaofeng Zhu
- The Genetics and Prenatal Diagnosis Center, The Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Jianshe Rd, Erqi District, 450052, Zhengzhou, Henan, China
| | - Yinghui Zhang
- The Department of Endocrinology, The First Affiliated Hospital of Zhengzhou University, Jianshe Rd, Erqi District, 450052, Zhengzhou, Henan, China
| | - Guifang Sun
- The Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Jianshe Rd, Erqi District, 450052, Zhengzhou, Henan, China
| | - Huanan Ren
- The Genetics and Prenatal Diagnosis Center, The Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Jianshe Rd, Erqi District, 450052, Zhengzhou, Henan, China
| | - Xiangdong Kong
- The Genetics and Prenatal Diagnosis Center, The Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Jianshe Rd, Erqi District, 450052, Zhengzhou, Henan, China.
| |
Collapse
|
2
|
Cannon SC. Periodic paralysis. HANDBOOK OF CLINICAL NEUROLOGY 2024; 203:39-58. [PMID: 39174253 DOI: 10.1016/b978-0-323-90820-7.00002-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2024]
Abstract
Periodic paralysis is a rare, dominantly inherited disorder of skeletal muscle in which episodic attacks of weakness are caused by a transient impairment of fiber excitability. Attacks of weakness are often elicited by characteristic environmental triggers, which were the basis for clinically delineating subtypes of periodic paralysis and are an important distinction for optimal disease management. All forms of familial periodic paralysis are caused by mutations of ion channels, often selectively expressed in skeletal muscle, that destabilize the resting potential. The missense mutations usually alter channel function through gain-of-function changes rather than producing a complete loss-of-function null. The knowledge of which channel gene harbors a variant, whether that variant is expected to (or known to) alter function, and how altered function impairs fiber excitability aides in the interpretation of patient signs and symptoms, the interpretation of gene test results, and how to optimize therapeutic intervention for symptom management and improve quality of life.
Collapse
Affiliation(s)
- Stephen C Cannon
- Departments of Physiology and of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States.
| |
Collapse
|
3
|
Liu Y, Bassetto CAZ, Pinto BI, Bezanilla F. A mechanistic reinterpretation of fast inactivation in voltage-gated Na + channels. Nat Commun 2023; 14:5072. [PMID: 37604801 PMCID: PMC10442390 DOI: 10.1038/s41467-023-40514-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 07/25/2023] [Indexed: 08/23/2023] Open
Abstract
The hinged-lid model was long accepted as the canonical model for fast inactivation in Nav channels. It predicts that the hydrophobic IFM motif acts intracellularly as the gating particle that binds and occludes the pore during fast inactivation. However, the observation in recent high-resolution structures that the bound IFM motif is located far from the pore, contradicts this preconception. Here, we provide a mechanistic reinterpretation of fast inactivation based on structural analysis and ionic/gating current measurements. We demonstrate that in Nav1.4 the final inactivation gate is comprised of two hydrophobic rings at the bottom of S6 helices. These rings function in series and close downstream of IFM binding. Reducing the volume of the sidechain in both rings leads to a partially conductive, leaky inactivated state and decreases the selectivity for Na+ ion. Altogether, we present an alternative molecular framework to describe fast inactivation.
Collapse
Affiliation(s)
- Yichen Liu
- Department of Neurobiology, University of Chicago, Chicago, IL, USA
| | - Carlos A Z Bassetto
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL, USA
| | - Bernardo I Pinto
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL, USA
| | - Francisco Bezanilla
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL, USA.
- Centro Interdisciplinario de Neurociencias de Valparaíso, Valparaíso, Chile.
| |
Collapse
|
4
|
Gada KD, Kamuene JM, Chandrashekar A, Kissell RC, Yauch AK, Plant LD. PI(4,5)P2 regulates the gating of NaV1.4 channels. J Gen Physiol 2023; 155:e202213255. [PMID: 37043561 PMCID: PMC10103707 DOI: 10.1085/jgp.202213255] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 02/22/2023] [Accepted: 03/22/2023] [Indexed: 04/14/2023] Open
Abstract
Voltage-gated sodium (NaV) channels are densely expressed in most excitable cells and activate in response to depolarization, causing a rapid influx of Na+ ions that initiates the action potential. The voltage-dependent activation of NaV channels is followed almost instantaneously by fast inactivation, setting the refractory period of excitable tissues. The gating cycle of NaV channels is subject to tight regulation, with perturbations leading to a range of pathophysiological states. The gating properties of most ion channels are regulated by the membrane phospholipid, phosphatidylinositol (4,5) bisphosphate (PI(4,5)P2). However, it is not known whether PI(4,5)P2 modulates the activity of NaV channels. Here, we utilize optogenetics to activate specific, membrane-associated phosphoinositide (PI)-phosphatases that dephosphorylate PI(4,5)P2 while simultaneously recording NaV1.4 channel currents. We show that dephosphorylating PI(4,5)P2 left-shifts the voltage-dependent gating of NaV1.4 to more hyperpolarized membrane potentials, augments the late current that persists after fast inactivation, and speeds the rate at which channels recover from fast inactivation. These effects are opposed by exogenous diC8PI(4,5)P2. We provide evidence that PI(4,5)P2 is a negative regulator that tunes the gating behavior of NaV1.4 channels.
Collapse
Affiliation(s)
- Kirin D. Gada
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Bouvé College of Health Sciences, Northeastern University, Boston, MA, USA
| | - Jordie M. Kamuene
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Bouvé College of Health Sciences, Northeastern University, Boston, MA, USA
| | - Aishwarya Chandrashekar
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Bouvé College of Health Sciences, Northeastern University, Boston, MA, USA
| | - R. Charles Kissell
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Bouvé College of Health Sciences, Northeastern University, Boston, MA, USA
| | - Anne K. Yauch
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Bouvé College of Health Sciences, Northeastern University, Boston, MA, USA
| | - Leigh D. Plant
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Bouvé College of Health Sciences, Northeastern University, Boston, MA, USA
- Center for Drug Discovery, Northeastern University, Boston, MA, USA
| |
Collapse
|
5
|
Liu Y, Bassetto CAZ, Pinto BI, Bezanilla F. A Mechanistic Reinterpretation of Fast Inactivation in Voltage-Gated Na + Channels. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.27.538555. [PMID: 37162849 PMCID: PMC10168311 DOI: 10.1101/2023.04.27.538555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Fast Inactivation in voltage-gated Na + channels plays essential roles in numerous physiological functions. The canonical hinged-lid model has long predicted that a hydrophobic motif in the DIII-DIV linker (IFM) acts as the gating particle that occludes the permeation pathway during fast inactivation. However, the fact that the IFM motif is located far from the pore in recent high-resolution structures of Nav + channels contradicts this status quo model. The precise molecular determinants of fast inactivation gate once again, become an open question. Here, we provide a mechanistic reinterpretation of fast inactivation based on ionic and gating current data. In Nav1.4 the actual inactivation gate is comprised of two hydrophobic rings at the bottom of S6. These function in series and closing once the IFM motif binds. Reducing the volume of the sidechain in both rings led to a partially conductive inactivated state. Our experiments also point to a previously overlooked coupling pathway between the bottom of S6 and the selectivity filter.
Collapse
Affiliation(s)
- Yichen Liu
- Department of Neurobiology, University of Chicago, Chicago, IL, USA
| | - Carlos A Z Bassetto
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL, USA
| | - Bernardo I Pinto
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL, USA
| | - Francisco Bezanilla
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL, USA
- Centro Interdisciplinario de Neurociencias de Valparaiso, Valparaiso, Chile
| |
Collapse
|
6
|
Yuan JH, Higuchi Y, Hashiguchi A, Ando M, Yoshimura A, Nakamura T, Hiramatsu Y, Sakiyama Y, Takashima H. Gene panel analysis of 119 index patients with suspected periodic paralysis in Japan. Front Neurol 2023; 14:1078195. [PMID: 36779057 PMCID: PMC9908745 DOI: 10.3389/fneur.2023.1078195] [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: 10/24/2022] [Accepted: 01/09/2023] [Indexed: 01/27/2023] Open
Abstract
Introduction Genetic factors are recognized as the major reason for patients with periodic paralysis. The goal of this study was to determine the genetic causes of periodic paralysis in Japan. Methods We obtained a Japanese nationwide case series of 119 index patients (108 men and 11 women) clinically suspected of periodic paralysis, and a gene panel analysis, targeting CACNA1S, SCN4A, and KCNJ2 genes, was conducted. Results From 34 cases, 25 pathogenic/likely pathogenic/unknown significance variants were detected in CACNA1S (nine cases), SCN4A (19 cases), or KCNJ2 (six cases), generating a molecular diagnostic rate of 28.6%. In total, seven variants have yet been found linked to periodic paralysis previously. The diagnostic yield of patients with hypokalemic and hyperkalemic periodic paralyzes was 26.2 (17/65) and 32.7% (17/52), respectively. A considerably higher yield was procured from patients with than without positive family history (18/25 vs. 16/94), onset age ≤20 years (24/57 vs. 9/59), or recurrent paralytic attacks (31/94 vs. 3/25). Discussion The low molecular diagnostic rate and specific genetic proportion of the present study highlight the etiological complexity of patients with periodic paralysis in Japan.
Collapse
|
7
|
Trivedi JR. Muscle Channelopathies. Continuum (Minneap Minn) 2022; 28:1778-1799. [PMID: 36537980 DOI: 10.1212/con.0000000000001183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
PURPOSE OF REVIEW This article describes the clinical features, diagnosis, pathophysiology, and management of nondystrophic myotonia and periodic paralysis. RECENT FINDINGS An increasing awareness exists about the genotype-phenotype overlap in skeletal muscle channelopathies, and thus genetic testing is needed to make a definitive diagnosis. Electrodiagnostic testing in channelopathies is highly specialized with significant overlap in various mutation subtypes. Randomized clinical trials have now been conducted in these disorders with expanded treatment options for patients with muscle channelopathies. SUMMARY Skeletal muscle channelopathies are rare heterogeneous conditions characterized by lifelong symptoms that require a comprehensive management plan that includes pharmacologic and nonpharmacologic interventions. The significant variability in biophysical features of various mutations, coupled with the difficulties of performing clinical trials in rare diseases, makes it challenging to design and implement treatment trials for muscle channelopathies.
Collapse
|
8
|
Cancer as a Channelopathy—Appreciation of Complimentary Pathways Provides a Different Perspective for Developing Treatments. Cancers (Basel) 2022; 14:cancers14194627. [PMID: 36230549 PMCID: PMC9562872 DOI: 10.3390/cancers14194627] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 09/17/2022] [Accepted: 09/21/2022] [Indexed: 12/15/2022] Open
Abstract
Simple Summary While improvements in technology have improved our ability to treat many forms of cancer when diagnosed at an early stage of the disease, the ability to improve survival and quality of life for patients with late stage disease has been limited, largely due to the ability of cancer cells to evade destruction when treatments block preferred paths for survival. Here, we review the role that ions and ion channels play in normal cell function, the development of disease and their role in the life and death of a cell. It is hoped that viewing cancer from the perspective of altered ion channel expression and ion balance may provide a novel approach for developing more effective treatments for this devastating disease. Abstract Life depends upon the ability of cells to evaluate and adapt to a constantly changing environment and to maintain internal stability to allow essential biochemical reactions to occur. Ions and ion channels play a crucial role in this process and are essential for survival. Alterations in the expression of the transmembrane proteins responsible for maintaining ion balance that occur as a result of mutations in the genetic code or in response to iatrogenically induced changes in the extracellular environment is a characteristic feature of oncogenesis and identifies cancer as one of a constellation of diseases known as channelopathies. The classification of cancer as a channelopathy provides a different perspective for viewing the disease. Potentially, it may expand opportunities for developing novel ways to affect or reverse the deleterious changes that underlie establishing and sustaining disease and developing tolerance to therapeutic attempts at treatment. The role of ions and ion channels and their interactions in the cell’s ability to maintain ionic balance, homeostasis, and survival are reviewed and possible approaches that mitigate gain or loss of ion channel function to contribute to new or enhance existing cancer therapies are discussed.
Collapse
|
9
|
Johnson JP, Focken T, Khakh K, Tari PK, Dube C, Goodchild SJ, Andrez JC, Bankar G, Bogucki D, Burford K, Chang E, Chowdhury S, Dean R, de Boer G, Decker S, Dehnhardt C, Feng M, Gong W, Grimwood M, Hasan A, Hussainkhel A, Jia Q, Lee S, Li J, Lin S, Lindgren A, Lofstrand V, Mezeyova J, Namdari R, Nelkenbrecher K, Shuart NG, Sojo L, Sun S, Taron M, Waldbrook M, Weeratunge D, Wesolowski S, Williams A, Wilson M, Xie Z, Yoo R, Young C, Zenova A, Zhang W, Cutts AJ, Sherrington RP, Pimstone SN, Winquist R, Cohen CJ, Empfield JR. NBI-921352, a first-in-class, Na V1.6 selective, sodium channel inhibitor that prevents seizures in Scn8a gain-of-function mice, and wild-type mice and rats. eLife 2022; 11:72468. [PMID: 35234610 PMCID: PMC8903829 DOI: 10.7554/elife.72468] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Accepted: 02/23/2022] [Indexed: 11/21/2022] Open
Abstract
NBI-921352 (formerly XEN901) is a novel sodium channel inhibitor designed to specifically target NaV1.6 channels. Such a molecule provides a precision-medicine approach to target SCN8A-related epilepsy syndromes (SCN8A-RES), where gain-of-function (GoF) mutations lead to excess NaV1.6 sodium current, or other indications where NaV1.6 mediated hyper-excitability contributes to disease (Gardella and Møller, 2019; Johannesen et al., 2019; Veeramah et al., 2012). NBI-921352 is a potent inhibitor of NaV1.6 (IC500.051 µM), with exquisite selectivity over other sodium channel isoforms (selectivity ratios of 756 X for NaV1.1, 134 X for NaV1.2, 276 X for NaV1.7, and >583 Xfor NaV1.3, NaV1.4, and NaV1.5). NBI-921352is a state-dependent inhibitor, preferentially inhibiting inactivatedchannels. The state dependence leads to potent stabilization of inactivation, inhibiting NaV1.6 currents, including resurgent and persistent NaV1.6 currents, while sparing the closed/rested channels. The isoform-selective profile of NBI-921352 led to a robust inhibition of action-potential firing in glutamatergic excitatory pyramidal neurons, while sparing fast-spiking inhibitory interneurons, where NaV1.1 predominates. Oral administration of NBI-921352 prevented electrically induced seizures in a Scn8a GoF mouse,as well as in wild-type mouse and ratseizure models. NBI-921352 was effective in preventing seizures at lower brain and plasma concentrations than commonly prescribed sodium channel inhibitor anti-seizure medicines (ASMs) carbamazepine, phenytoin, and lacosamide. NBI-921352 waswell tolerated at higher multiples of the effective plasma and brain concentrations than those ASMs. NBI-921352 is entering phase II proof-of-concept trials for the treatment of SCN8A-developmental epileptic encephalopathy (SCN8A-DEE) and adult focal-onset seizures.
Collapse
Affiliation(s)
- J P Johnson
- In Vitro Biology, Xenon Pharmaceuticals, Inc., Burnaby BC, Canada
| | - Thilo Focken
- Chemistry, Xenon Pharmaceuticals, Inc., Burnaby BC, Canada
| | - Kuldip Khakh
- In Vitro Biology, Xenon Pharmaceuticals, Inc., Burnaby BC, Canada
| | | | - Celine Dube
- In Vivo Biology, Xenon Pharmaceuticals, Inc., Burnaby BC, Canada
| | | | | | - Girish Bankar
- In Vivo Biology, Xenon Pharmaceuticals, Inc., Burnaby BC, Canada
| | - David Bogucki
- Chemistry, Medipure Pharmaceuticals, Burnaby BC, Canada
| | | | - Elaine Chang
- In Vitro Biology, Xenon Pharmaceuticals, Inc., Burnaby BC, Canada
| | | | - Richard Dean
- In Vitro Biology, Xenon Pharmaceuticals, Inc., Burnaby BC, Canada
| | - Gina de Boer
- Compound Properties, Xenon Pharmaceuticals, Inc., Burnaby BC, Canada
| | - Shannon Decker
- Chemistry, Xenon Pharmaceuticals, Inc., Burnaby BC, Canada
| | | | - Mandy Feng
- In Vitro Biology, Xenon Pharmaceuticals, Inc., Burnaby BC, Canada
| | - Wei Gong
- Chemistry, Xenon Pharmaceuticals, Inc., Burnaby BC, Canada
| | | | - Abid Hasan
- Chemistry, Xenon Pharmaceuticals, Inc., Burnaby BC, Canada
| | | | - Qi Jia
- Chemistry, Xenon Pharmaceuticals, Inc., Burnaby BC, Canada
| | - Stephanie Lee
- Compound Properties, Xenon Pharmaceuticals, Inc., Burnaby BC, Canada
| | - Jenny Li
- In Vitro Biology, Xenon Pharmaceuticals, Inc., Burnaby BC, Canada
| | - Sophia Lin
- In Vitro Biology, Xenon Pharmaceuticals, Inc., Burnaby BC, Canada
| | - Andrea Lindgren
- Compound Properties, Xenon Pharmaceuticals, Inc., Burnaby BC, Canada
| | | | - Janette Mezeyova
- In Vitro Biology, Xenon Pharmaceuticals, Inc., Burnaby BC, Canada
| | - Rostam Namdari
- Translational Drug Development, Xenon Pharmaceuticals, Inc., Burnaby BC, Canada
| | | | | | - Luis Sojo
- Compound Properties, Xenon Pharmaceuticals, Inc., Burnaby BC, Canada
| | - Shaoyi Sun
- Chemistry, Xenon Pharmaceuticals, Inc., Burnaby BC, Canada
| | - Matthew Taron
- Chemistry, Xenon Pharmaceuticals, Inc., Burnaby BC, Canada
| | | | - Diana Weeratunge
- In Vitro Biology, Xenon Pharmaceuticals, Inc., Burnaby BC, Canada
| | | | - Aaron Williams
- In Vitro Biology, Xenon Pharmaceuticals, Inc., Burnaby BC, Canada
| | - Michael Wilson
- Chemistry, Xenon Pharmaceuticals, Inc., Burnaby BC, Canada
| | - Zhiwei Xie
- In Vitro Biology, Xenon Pharmaceuticals, Inc., Burnaby BC, Canada
| | - Rhena Yoo
- In Vitro Biology, Xenon Pharmaceuticals, Inc., Burnaby BC, Canada
| | - Clint Young
- In Vitro Biology, Xenon Pharmaceuticals, Inc., Burnaby BC, Canada
| | - Alla Zenova
- Chemistry, Xenon Pharmaceuticals, Inc., Burnaby BC, Canada
| | - Wei Zhang
- Chemistry, Xenon Pharmaceuticals, Inc., Burnaby BC, Canada
| | - Alison J Cutts
- Scientific Affairs, Xenon Pharmaceuticals, Inc., Burnaby BC, Canada
| | | | | | | | - Charles J Cohen
- Executive Team, Xenon Pharmaceuticals, Inc., Burnaby BC, Canada
| | | |
Collapse
|
10
|
Nicole S, Lory P. New Challenges Resulting From the Loss of Function of Na v1.4 in Neuromuscular Diseases. Front Pharmacol 2021; 12:751095. [PMID: 34671263 PMCID: PMC8521073 DOI: 10.3389/fphar.2021.751095] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Accepted: 09/16/2021] [Indexed: 11/13/2022] Open
Abstract
The voltage-gated sodium channel Nav1.4 is a major actor in the excitability of skeletal myofibers, driving the muscle force in response to nerve stimulation. Supporting further this key role, mutations in SCN4A, the gene encoding the pore-forming α subunit of Nav1.4, are responsible for a clinical spectrum of human diseases ranging from muscle stiffness (sodium channel myotonia, SCM) to muscle weakness. For years, only dominantly-inherited diseases resulting from Nav1.4 gain of function (GoF) were known, i.e., non-dystrophic myotonia (delayed muscle relaxation due to myofiber hyperexcitability), paramyotonia congenita and hyperkalemic or hypokalemic periodic paralyses (episodic flaccid muscle weakness due to transient myofiber hypoexcitability). These last 5 years, SCN4A mutations inducing Nav1.4 loss of function (LoF) were identified as the cause of dominantly and recessively-inherited disorders with muscle weakness: periodic paralyses with hypokalemic attacks, congenital myasthenic syndromes and congenital myopathies. We propose to name this clinical spectrum sodium channel weakness (SCW) as the mirror of SCM. Nav1.4 LoF as a cause of permanent muscle weakness was quite unexpected as the Na+ current density in the sarcolemma is large, securing the ability to generate and propagate muscle action potentials. The properties of SCN4A LoF mutations are well documented at the channel level in cellular electrophysiological studies However, much less is known about the functional consequences of Nav1.4 LoF in skeletal myofibers with no available pertinent cell or animal models. Regarding the therapeutic issues for Nav1.4 channelopathies, former efforts were aimed at developing subtype-selective Nav channel antagonists to block myofiber hyperexcitability. Non-selective, Nav channel blockers are clinically efficient in SCM and paramyotonia congenita, whereas patient education and carbonic anhydrase inhibitors are helpful to prevent attacks in periodic paralyses. Developing therapeutic tools able to counteract Nav1.4 LoF in skeletal muscles is then a new challenge in the field of Nav channelopathies. Here, we review the current knowledge regarding Nav1.4 LoF and discuss the possible therapeutic strategies to be developed in order to improve muscle force in SCW.
Collapse
Affiliation(s)
- Sophie Nicole
- Institut de Génomique Fonctionnelle (IGF), Université de Montpellier, CNRS, INSERM, Montpellier, France.,LabEx 'Ion Channel Science and Therapeutics (ICST), Montpellier, France
| | - Philippe Lory
- Institut de Génomique Fonctionnelle (IGF), Université de Montpellier, CNRS, INSERM, Montpellier, France.,LabEx 'Ion Channel Science and Therapeutics (ICST), Montpellier, France
| |
Collapse
|
11
|
Maggi L, Bonanno S, Altamura C, Desaphy JF. Ion Channel Gene Mutations Causing Skeletal Muscle Disorders: Pathomechanisms and Opportunities for Therapy. Cells 2021; 10:cells10061521. [PMID: 34208776 PMCID: PMC8234207 DOI: 10.3390/cells10061521] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 06/03/2021] [Accepted: 06/10/2021] [Indexed: 02/06/2023] Open
Abstract
Skeletal muscle ion channelopathies (SMICs) are a large heterogeneous group of rare genetic disorders caused by mutations in genes encoding ion channel subunits in the skeletal muscle mainly characterized by myotonia or periodic paralysis, potentially resulting in long-term disabilities. However, with the development of new molecular technologies, new genes and new phenotypes, including progressive myopathies, have been recently discovered, markedly increasing the complexity in the field. In this regard, new advances in SMICs show a less conventional role of ion channels in muscle cell division, proliferation, differentiation, and survival. Hence, SMICs represent an expanding and exciting field. Here, we review current knowledge of SMICs, with a description of their clinical phenotypes, cellular and molecular pathomechanisms, and available treatments.
Collapse
Affiliation(s)
- Lorenzo Maggi
- Neuroimmunology and Neuromuscular Disorders Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy;
- Correspondence:
| | - Silvia Bonanno
- Neuroimmunology and Neuromuscular Disorders Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy;
| | - Concetta Altamura
- Department of Biomedical Sciences and Human Oncology, School of Medicine, University of Bari Aldo Moro, 70124 Bari, Italy; (C.A.); (J.-F.D.)
| | - Jean-François Desaphy
- Department of Biomedical Sciences and Human Oncology, School of Medicine, University of Bari Aldo Moro, 70124 Bari, Italy; (C.A.); (J.-F.D.)
| |
Collapse
|
12
|
Shen H, Yan N, Pan X. Structural determination of human Na v1.4 and Na v1.7 using single particle cryo-electron microscopy. Methods Enzymol 2021; 653:103-120. [PMID: 34099168 DOI: 10.1016/bs.mie.2021.03.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Voltage-gated sodium (Nav) channels are responsible for the initiation and propagation of action potentials. Their abnormal functions are associated with numerous diseases, such as epilepsy, cardiac arrhythmia, and pain syndromes. Therefore, these channels represent important drug targets. Even in the post-resolution revolution era, a lack of structural information continues to impede structure-based drug discovery. The limiting factor for the structural determination of Nav channels using single particle cryo-electron microscopy (cryo-EM) resides in the generation of sufficient high-quality recombinant proteins. After extensive trials, we have been successful in determining a series of high-resolution structures of Nav channels, including NavPaS from American cockroach, Nav1.4 from electric eel, and human Nav1.1, Nav1.2, Nav1.4, Nav1.5, and Nav1.7, with distinct strategies. These structures established the framework for understanding the electromechanical coupling and disease mechanism of Nav channels, and for facilitating drug discovery. Here, we exemplify these methods with two specific cases, human Nav1.4 and Nav1.7, which may shed light on the structural determination of other membrane proteins.
Collapse
Affiliation(s)
- Huaizong Shen
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China.
| | - Nieng Yan
- Department of Molecular Biology, Princeton University, Princeton, NJ, United States.
| | - Xiaojing Pan
- State Key Laboratory of Membrane Biology, Beijing Advanced Innovation Center for Structural Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China.
| |
Collapse
|
13
|
Mantegazza M, Cestèle S, Catterall WA. Sodium channelopathies of skeletal muscle and brain. Physiol Rev 2021; 101:1633-1689. [PMID: 33769100 DOI: 10.1152/physrev.00025.2020] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Voltage-gated sodium channels initiate action potentials in nerve, skeletal muscle, and other electrically excitable cells. Mutations in them cause a wide range of diseases. These channelopathy mutations affect every aspect of sodium channel function, including voltage sensing, voltage-dependent activation, ion conductance, fast and slow inactivation, and both biosynthesis and assembly. Mutations that cause different forms of periodic paralysis in skeletal muscle were discovered first and have provided a template for understanding structure, function, and pathophysiology at the molecular level. More recent work has revealed multiple sodium channelopathies in the brain. Here we review the well-characterized genetics and pathophysiology of the periodic paralyses of skeletal muscle and then use this information as a foundation for advancing our understanding of mutations in the structurally homologous α-subunits of brain sodium channels that cause epilepsy, migraine, autism, and related comorbidities. We include studies based on molecular and structural biology, cell biology and physiology, pharmacology, and mouse genetics. Our review reveals unexpected connections among these different types of sodium channelopathies.
Collapse
Affiliation(s)
- Massimo Mantegazza
- Université Cote d'Azur, Valbonne-Sophia Antipolis, France.,CNRS UMR7275, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne-Sophia Antipolis, France.,INSERM, Valbonne-Sophia Antipolis, France
| | - Sandrine Cestèle
- Université Cote d'Azur, Valbonne-Sophia Antipolis, France.,CNRS UMR7275, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne-Sophia Antipolis, France
| | | |
Collapse
|
14
|
Wang Z, Embaye KS, Yang Q, Qin L, Zhang C, Liu L, Zhan X, Zhang F, Wang X, Qin S. A Novel Metabolism-Related Signature as a Candidate Prognostic Biomarker for Hepatocellular Carcinoma. J Hepatocell Carcinoma 2021; 8:119-132. [PMID: 33758763 PMCID: PMC7981163 DOI: 10.2147/jhc.s294108] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 02/01/2021] [Indexed: 12/15/2022] Open
Abstract
PURPOSE Given that metabolic reprogramming has been recognized as an essential hallmark of cancer cells, this study sought to investigate the potential prognostic values of metabolism-related genes (MRGs) for the diagnosis and treatment of hepatocellular carcinoma (HCC). METHODS In total, 2752 metabolism-related gene sequencing data of HCC samples with clinical information were obtained from the International Cancer Genome Consortium (ICGC) and The Cancer Genome Atlas (TCGA). One hundred and seventy-eight the differentially expressed MRGs were identified from the ICGC cohort and TCGA cohort. Then, univariate Cox regression analysis was performed to identify these genes that were related to overall survival (OS). A novel metabolism-related prognostic signature was developed using the least absolute shrinkage and selection operator (Lasso) and multivariate Cox regression analyses in the ICGC dataset. The Broad Institute's Connectivity Map (CMap) was used in predicting which compounds on the basis of the prognostic MRGs. Furthermore, the signature was validated in the TCGA dataset. Finally, the expression levels of hub genes were validated in HCC cell lines by Western blotting (WB) and quantitative real-time PCR (qRT-PCR). RESULTS We found that 17 MRGs were most significantly associated with OS in HCC. Then, the Lasso and multivariate Cox regression analyses were applied to construct the novel metabolism-relevant prognostic signature, which consisted of six MRGs. The prognostic value of this prognostic model was further successfully validated in the TCGA dataset. Further analysis indicated that this particular signature could be an independent prognostic indicator after adjusting to other clinical factors. Six MRGs (FLVCR1, MOGAT2, SLC5A11, RRM2, COX7B2, and SCN4A) showed high prognostic performance in predicting HCC outcomes. Candidate drugs that aimed at hub ERGs were identified. Finally, hub genes were chosen for validation and the protein, mRNA expression of FLVCR1, SLC5A11, and RRM2 were significantly increased in human HCC cell lines compared to normal human hepatic cell lines, which were in agreement with the results of differential expression analysis. CONCLUSION Our data provided evidence that the metabolism-related signature could serve as a reliable prognostic and predictive tool for OS in patients with HCC.
Collapse
Affiliation(s)
- Zhihao Wang
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Kidane Siele Embaye
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Qing Yang
- Department of Pharmacy, Hiser Medical Center of Qingdao, Qingdao, 266033, People’s Republic of China
| | - Lingzhi Qin
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Chao Zhang
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Liwei Liu
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Xiaoqian Zhan
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Fengdi Zhang
- Department of Pathology, Wuhan Third Hospital, Wuhan, 430030, People’s Republic of China
| | - Xi Wang
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Shenghui Qin
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| |
Collapse
|
15
|
Ptáček LJ, Fu YH. The whole is greater than the sum of the parts. J Clin Invest 2021; 131:145965. [PMID: 33463539 PMCID: PMC7810464 DOI: 10.1172/jci145965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
16
|
Desaphy JF, Altamura C, Vicart S, Fontaine B. Targeted Therapies for Skeletal Muscle Ion Channelopathies: Systematic Review and Steps Towards Precision Medicine. J Neuromuscul Dis 2021; 8:357-381. [PMID: 33325393 PMCID: PMC8203248 DOI: 10.3233/jnd-200582] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Skeletal muscle ion channelopathies include non-dystrophic myotonias (NDM), periodic paralyses (PP), congenital myasthenic syndrome, and recently identified congenital myopathies. The treatment of these diseases is mainly symptomatic, aimed at reducing muscle excitability in NDM or modifying triggers of attacks in PP. OBJECTIVE This systematic review collected the evidences regarding effects of pharmacological treatment on muscle ion channelopathies, focusing on the possible link between treatments and genetic background. METHODS We searched databases for randomized clinical trials (RCT) and other human studies reporting pharmacological treatments. Preclinical studies were considered to gain further information regarding mutation-dependent drug effects. All steps were performed by two independent investigators, while two others critically reviewed the entire process. RESULTS For NMD, RCT showed therapeutic benefits of mexiletine and lamotrigine, while other human studies suggest some efficacy of various sodium channel blockers and of the carbonic anhydrase inhibitor (CAI) acetazolamide. Preclinical studies suggest that mutations may alter sensitivity of the channel to sodium channel blockers in vitro, which has been translated to humans in some cases. For hyperkalemic and hypokalemic PP, RCT showed efficacy of the CAI dichlorphenamide in preventing paralysis. However, hypokalemic PP patients carrying sodium channel mutations may have fewer benefits from CAI compared to those carrying calcium channel mutations. Few data are available for treatment of congenital myopathies. CONCLUSIONS These studies provided limited information about the response to treatments of individual mutations or groups of mutations. A major effort is needed to perform human studies for designing a mutation-driven precision medicine in muscle ion channelopathies.
Collapse
Affiliation(s)
- Jean-François Desaphy
- Department of Biomedical Sciences and Human Oncology, School of Medicine, University of Bari Aldo Moro, Bari, Italy
| | - Concetta Altamura
- Department of Biomedical Sciences and Human Oncology, School of Medicine, University of Bari Aldo Moro, Bari, Italy
| | - Savine Vicart
- Sorbonne Université, INSERM, Assistance Publique Hôpitaux de Paris, Centre de Recherche en Myologie-UMR 974, Reference center in neuro-muscular channelopathies, Institute of Myology, Hôpital Universitaire Pitié-Salpêtrière, Paris, France
| | - Bertrand Fontaine
- Sorbonne Université, INSERM, Assistance Publique Hôpitaux de Paris, Centre de Recherche en Myologie-UMR 974, Reference center in neuro-muscular channelopathies, Institute of Myology, Hôpital Universitaire Pitié-Salpêtrière, Paris, France
| |
Collapse
|
17
|
Stunnenberg BC, LoRusso S, Arnold WD, Barohn RJ, Cannon SC, Fontaine B, Griggs RC, Hanna MG, Matthews E, Meola G, Sansone VA, Trivedi JR, van Engelen BG, Vicart S, Statland JM. Guidelines on clinical presentation and management of nondystrophic myotonias. Muscle Nerve 2020; 62:430-444. [PMID: 32270509 PMCID: PMC8117169 DOI: 10.1002/mus.26887] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 04/01/2020] [Accepted: 04/04/2020] [Indexed: 12/26/2022]
Abstract
The nondystrophic myotonias are rare muscle hyperexcitability disorders caused by gain-of-function mutations in the SCN4A gene or loss-of-function mutations in the CLCN1 gene. Clinically, they are characterized by myotonia, defined as delayed muscle relaxation after voluntary contraction, which leads to symptoms of muscle stiffness, pain, fatigue, and weakness. Diagnosis is based on history and examination findings, the presence of electrical myotonia on electromyography, and genetic confirmation. In the absence of genetic confirmation, the diagnosis is supported by detailed electrophysiological testing, exclusion of other related disorders, and analysis of a variant of uncertain significance if present. Symptomatic treatment with a sodium channel blocker, such as mexiletine, is usually the first step in management, as well as educating patients about potential anesthetic complications.
Collapse
Affiliation(s)
- Bas C. Stunnenberg
- Department of Neurology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Samantha LoRusso
- Department of Neurology, Ohio State University Wexner Medical Center, Columbus, Ohio
| | - W. David Arnold
- Department of Neurology, Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Richard J. Barohn
- Department of Neurology, University of Kansas Medical Center, Kansas City, Kansas
| | - Stephen C. Cannon
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Bertrand Fontaine
- Assistance Publique-Hôpitaix de Paris, Sorbonne Université, INSERM, Service of Neuro-Myology and UMR 974, Institute of Myology, University Hospital Pitié-Salpêtrière, Paris, France
| | - Robert C. Griggs
- Department of Neurology, University of Rochester, Rochester, New York
| | - Michael G. Hanna
- MRC Centre for Neuromuscular Diseases, Department of Neuromuscular diseases, UCL Queen Square Institute of Neurology, United Kingdom
| | - Emma Matthews
- MRC Centre for Neuromuscular Diseases, Department of Neuromuscular diseases, UCL Queen Square Institute of Neurology, United Kingdom
| | - Giovanni Meola
- Department of Neurorehabilitation Sciences, Casa Cura Policlinico, Milan, Italy
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
| | - Valeria A. Sansone
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
- Neurorehabilitation Unit, University of Milan, NEuroMuscular Omnicentre (NEMO), Fondazione Serena Onlus, Milan, Italy
| | - Jaya R. Trivedi
- Department of Neurology and Neurotherapeutics, UT Southwestern Medical Center, Dallas, Texas
| | | | - Savine Vicart
- Assistance Publique-Hôpitaix de Paris, Sorbonne Université, INSERM, Service of Neuro-Myology and UMR 974, Institute of Myology, University Hospital Pitié-Salpêtrière, Paris, France
| | - Jeffrey M. Statland
- Department of Neurology, University of Kansas Medical Center, Kansas City, Kansas
| |
Collapse
|
18
|
Heyne HO, Baez-Nieto D, Iqbal S, Palmer DS, Brunklaus A, May P, Johannesen KM, Lauxmann S, Lemke JR, Møller RS, Pérez-Palma E, Scholl UI, Syrbe S, Lerche H, Lal D, Campbell AJ, Wang HR, Pan J, Daly MJ. Predicting functional effects of missense variants in voltage-gated sodium and calcium channels. Sci Transl Med 2020; 12:eaay6848. [PMID: 32801145 DOI: 10.1126/scitranslmed.aay6848] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 11/20/2019] [Accepted: 07/22/2020] [Indexed: 12/30/2022]
Abstract
Malfunctions of voltage-gated sodium and calcium channels (encoded by SCNxA and CACNA1x family genes, respectively) have been associated with severe neurologic, psychiatric, cardiac, and other diseases. Altered channel activity is frequently grouped into gain or loss of ion channel function (GOF or LOF, respectively) that often corresponds not only to clinical disease manifestations but also to differences in drug response. Experimental studies of channel function are therefore important, but laborious and usually focus only on a few variants at a time. On the basis of known gene-disease mechanisms of 19 different diseases, we inferred LOF (n = 518) and GOF (n = 309) likely pathogenic variants from the disease phenotypes of variant carriers. By training a machine learning model on sequence- and structure-based features, we predicted LOF or GOF effects [area under the receiver operating characteristics curve (ROC) = 0.85] of likely pathogenic missense variants. Our LOF versus GOF prediction corresponded to molecular LOF versus GOF effects for 87 functionally tested variants in SCN1/2/8A and CACNA1I (ROC = 0.73) and was validated in exome-wide data from 21,703 cases and 128,957 controls. We showed respective regional clustering of inferred LOF and GOF nucleotide variants across the alignment of the entire gene family, suggesting shared pathomechanisms in the SCNxA/CACNA1x family genes.
Collapse
Affiliation(s)
- Henrike O Heyne
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA 02114, USA.
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, 5WR36M Helsinki, Finland
| | - David Baez-Nieto
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Sumaiya Iqbal
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA 02114, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Center for Development of Therapeutics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Duncan S Palmer
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA 02114, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Andreas Brunklaus
- Paediatric Neurosciences Research Group, Royal Hospital for Sick Children, Glasgow G51 4TF, UK
- School of Medicine, University of Glasgow, Glasgow G12 8QQ, UK
| | - Patrick May
- Luxembourg Centre for Systems Biomedicine, Belvaux, University of Luxembourg, 4365 Esch-sur-Alzette, Luxembourg
| | - Katrine M Johannesen
- Department of Epilepsy Genetics and Personalized Treatment, Danish Epilepsy Centre, 4293 Dianalund, Denmark
- Department of Regional Health Research, University of Southern Denmark, 5230 Odense, Denmark
| | - Stephan Lauxmann
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tuebingen, 72076 Tuebingen, Germany
| | - Johannes R Lemke
- Institute of Human Genetics, University of Leipzig Medical Center, 04103 Leipzig, Germany
| | - Rikke S Møller
- Department of Epilepsy Genetics and Personalized Treatment, Danish Epilepsy Centre, 4293 Dianalund, Denmark
- Department of Regional Health Research, University of Southern Denmark, 5230 Odense, Denmark
| | - Eduardo Pérez-Palma
- Cologne Center for Genomics (CCG), University of Cologne, 50923, Germany
- Genomic Medicine Institute, Lemer Research Institute Cleveland Clinic, OH G92J47, USA
| | - Ute I Scholl
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Nephrology and Medical Intensive Care and BIH Center for Regenerative Therapies, 10178 Berlin, Germany
- Berlin Institute of Health (BIH), 10178 Berlin, Germany
| | - Steffen Syrbe
- Division of Pediatric Epileptology, Center for Paediatrics and Adolescent Medicine, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Holger Lerche
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tuebingen, 72076 Tuebingen, Germany
| | - Dennis Lal
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA 02114, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Cologne Center for Genomics (CCG), University of Cologne, 50923, Germany
- Genomic Medicine Institute, Lemer Research Institute Cleveland Clinic, OH G92J47, USA
- Epilepsy Center, Neurological Institute, Cleveland Clinic, Cleveland, OH G92J47, USA
| | - Arthur J Campbell
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Center for Development of Therapeutics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Hao-Ran Wang
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Jen Pan
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Mark J Daly
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA 02114, USA.
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, 5WR36M Helsinki, Finland
| |
Collapse
|
19
|
Genes containing hexanucleotide repeats resembling C9ORF72 and expressed in the central nervous system are frequent in the human genome. Neurobiol Aging 2020; 97:148.e1-148.e7. [PMID: 32843153 DOI: 10.1016/j.neurobiolaging.2020.07.027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Revised: 07/22/2020] [Accepted: 07/25/2020] [Indexed: 12/14/2022]
Abstract
More than 40 human diseases, mainly diseases affecting the central nervous system, are caused by the expansion of unstable nucleotide repeats. Repeats of sequences like (CAG)n present in different genes can be responsible for various diseases of the central nervous system. An expanded hexanucleotide repeat (GGGGCC)n in the C9ORF72 gene has been characterized as the most frequent genetic cause of amyotrophic lateral sclerosis and frontotemporal lobar dementia. In this study, we performed a genome-wide analysis in the human genome and identified 74 genes containing this precise hexanucleotide repeat, with a preference for a location in exon 1 or intron 1, similar to the C9ORF72 gene. A total of 36 of these 74 genes may be of interest as candidates in neurodevelopmental or neurodegenerative diseases, based on their function.
Collapse
|
20
|
Abstract
Skeletal muscle channelopathies are rare genetic neuromuscular conditions that include the nondystrophic myotonias and periodic paralyses. They cause disabling muscle symptoms and can limit educational potential, work opportunities, socialization, and quality of life. Effective therapy is available, making it essential to recognize and treat this group of disorders. Here, the authors highlight important aspects regarding diagnosis and management using illustrative case reports.
Collapse
Affiliation(s)
- Vinojini Vivekanandam
- Department of Neuromuscular Diseases, Queen Square Centre for Neuromuscular Diseases, UCL Queen Square Institute of Neurology and National Hospital for Neurology and Neurosurgery, London WC1N 3BG, UK
| | - Pinki Munot
- Dubowitz Neuromuscular Centre, Great Ormond Street Hospital for Children, London, UK
| | - Michael G Hanna
- Department of Neuromuscular Diseases, Queen Square Centre for Neuromuscular Diseases, UCL Queen Square Institute of Neurology and National Hospital for Neurology and Neurosurgery, London WC1N 3BG, UK
| | - Emma Matthews
- Department of Neuromuscular Diseases, Queen Square Centre for Neuromuscular Diseases, UCL Queen Square Institute of Neurology and National Hospital for Neurology and Neurosurgery, London WC1N 3BG, UK.
| |
Collapse
|
21
|
Matthews E, Balestrini S, Sisodiya SM, Hanna MG. Muscle and brain sodium channelopathies: genetic causes, clinical phenotypes, and management approaches. THE LANCET CHILD & ADOLESCENT HEALTH 2020; 4:536-547. [PMID: 32142633 DOI: 10.1016/s2352-4642(19)30425-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 10/29/2019] [Accepted: 12/12/2019] [Indexed: 01/26/2023]
Abstract
Voltage-gated sodium channels are essential for excitability of skeletal muscle fibres and neurons. An increasing number of disabling or fatal paediatric neurological disorders linked to mutations of voltage-gated sodium channel genes are recognised. Muscle phenotypes include episodic paralysis, myotonia, neonatal hypotonia, respiratory compromise, laryngospasm or stridor, congenital myasthenia, and myopathy. Evidence suggests a possible link between sodium channel dysfunction and sudden infant death. Increasingly recognised phenotypes of brain sodium channelopathies include several epilepsy disorders and complex encephalopathies. Together, these early-onset muscle and brain phenotypes have a substantial morbidity and a considerable mortality. Important advances in understanding the pathophysiological mechanisms underlying these channelopathies have helped to identify effective targeted therapies. The availability of effective treatments underlines the importance of increasing clinical awareness and the need to achieve a precise genetic diagnosis. In this Review, we describe the expanded range of phenotypes of muscle and brain sodium channelopathies and the underlying knowledge regarding mechanisms of sodium channel dysfunction. We also outline a diagnostic approach and review the available treatment options.
Collapse
Affiliation(s)
- Emma Matthews
- Department of Neuromuscular Diseases, Medical Research Council Centre for Neuromuscular Diseases, University College London Queen Square Institute of Neurology, London, UK; National Hospital for Neurology and Neurosurgery, University College London Hospitals National Health Service Foundation Trust, London, UK.
| | - Simona Balestrini
- Department of Clinical and Experimental Epilepsy, University College London Queen Square Institute of Neurology, London, UK; Chalfont Centre for Epilepsy, Buckinghamshire, UK; National Hospital for Neurology and Neurosurgery, University College London Hospitals National Health Service Foundation Trust, London, UK
| | - Sanjay M Sisodiya
- Department of Clinical and Experimental Epilepsy, University College London Queen Square Institute of Neurology, London, UK; Chalfont Centre for Epilepsy, Buckinghamshire, UK; National Hospital for Neurology and Neurosurgery, University College London Hospitals National Health Service Foundation Trust, London, UK
| | - Michael G Hanna
- Department of Neuromuscular Diseases, Medical Research Council Centre for Neuromuscular Diseases, University College London Queen Square Institute of Neurology, London, UK; National Hospital for Neurology and Neurosurgery, University College London Hospitals National Health Service Foundation Trust, London, UK
| |
Collapse
|
22
|
Morales F, Pusch M. An Up-to-Date Overview of the Complexity of Genotype-Phenotype Relationships in Myotonic Channelopathies. Front Neurol 2020; 10:1404. [PMID: 32010054 PMCID: PMC6978732 DOI: 10.3389/fneur.2019.01404] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 12/23/2019] [Indexed: 12/11/2022] Open
Abstract
Myotonic disorders are inherited neuromuscular diseases divided into dystrophic myotonias and non-dystrophic myotonias (NDM). The latter is a group of dominant or recessive diseases caused by mutations in genes encoding ion channels that participate in the generation and control of the skeletal muscle action potential. Their altered function causes hyperexcitability of the muscle membrane, thereby triggering myotonia, the main sign in NDM. Mutations in the genes encoding voltage-gated Cl− and Na+ channels (respectively, CLCN1 and SCN4A) produce a wide spectrum of phenotypes, which differ in age of onset, affected muscles, severity of myotonia, degree of hypertrophy, and muscle weakness, disease progression, among others. More than 200 CLCN1 and 65 SCN4A mutations have been identified and described, but just about half of them have been functionally characterized, an approach that is likely extremely helpful to contribute to improving the so-far rather poor clinical correlations present in NDM. The observed poor correlations may be due to: (1) the wide spectrum of symptoms and overlapping phenotypes present in both groups (Cl− and Na+ myotonic channelopathies) and (2) both genes present high genotypic variability. On the one hand, several mutations cause a unique and reproducible phenotype in most patients. On the other hand, some mutations can have different inheritance pattern and clinical phenotypes in different families. Conversely, different mutations can be translated into very similar phenotypes. For these reasons, the genotype-phenotype relationships in myotonic channelopathies are considered complex. Although the molecular bases for the clinical variability present in myotonic channelopathies remain obscure, several hypotheses have been put forward to explain the variability, which include: (a) differential allelic expression; (b) trans-acting genetic modifiers; (c) epigenetic, hormonal, or environmental factors; and (d) dominance with low penetrance. Improvements in clinical tests, the recognition of the different phenotypes that result from particular mutations and the understanding of how a mutation affects the structure and function of the ion channel, together with genetic screening, is expected to improve clinical correlation in NDMs.
Collapse
Affiliation(s)
- Fernando Morales
- Instituto de Investigaciones en Salud, Universidad de Costa, San José, Costa Rica
| | | |
Collapse
|
23
|
Cibulcik F, Spalek P, Martinka I, Zidkova J, Grofik M, Sivak S, Kurca E. Paramyotonia congenita in a Slovak population: Genetic and pedigree analysis of 3 families. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2019; 163:362-365. [DOI: 10.5507/bp.2018.078] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 12/09/2018] [Indexed: 11/23/2022] Open
|
24
|
Abstract
PURPOSE OF REVIEW This article reviews the episodic muscle disorders, including benign cramp-fasciculation syndrome, the periodic paralyses, and the nondystrophic myotonias. The core diagnostic criteria for a diagnosis of primary periodic paralysis, including clues to distinguish between the hypokalemic and hyperkalemic forms, and the distinctive elements that characterize Andersen-Tawil syndrome are discussed. Management of patients with these disorders is also discussed. RECENT FINDINGS Childhood presentations of periodic paralysis have recently been described, including atypical findings. Carbonic anhydrase inhibitors, such as dichlorphenamide, have recently been approved by the US Food and Drug Administration (FDA) for the treatment of both hypokalemic and hyperkalemic forms of periodic paralysis. Muscle MRI may be a useful outcome measure in pharmacologic trials in periodic paralysis. Genetic research continues to identify additional gene mutations responsible for periodic paralysis. SUMMARY This article will help neurologists diagnose and manage episodic muscle disorders and, in particular, the periodic paralyses and the nondystrophic myotonias.
Collapse
|
25
|
Metzger S, Dupont C, Voss AA, Rich MM. Central Role of Subthreshold Currents in Myotonia. Ann Neurol 2019; 87:175-183. [PMID: 31725924 DOI: 10.1002/ana.25646] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 11/12/2019] [Accepted: 11/12/2019] [Indexed: 01/11/2023]
Abstract
It is generally thought that muscle excitability is almost exclusively controlled by currents responsible for generation of action potentials. We propose that smaller ion channel currents that contribute to setting the resting potential and to subthreshold fluctuations in membrane potential can also modulate excitability in important ways. These channels open at voltages more negative than the action potential threshold and are thus termed subthreshold currents. As subthreshold currents are orders of magnitude smaller than the currents responsible for the action potential, they are hard to identify and easily overlooked. Discovery of their importance in regulation of excitability opens new avenues for improved therapy for muscle channelopathies and diseases of the neuromuscular junction. ANN NEUROL 2020;87:175-183.
Collapse
Affiliation(s)
- Sabrina Metzger
- Department of Neuroscience, Cell Biology, and Physiology, Wright State University, Dayton, OH
| | - Chris Dupont
- Department of Neuroscience, Cell Biology, and Physiology, Wright State University, Dayton, OH
| | - Andrew A Voss
- Department of Biology, Wright State University, Dayton, OH
| | - Mark M Rich
- Department of Neuroscience, Cell Biology, and Physiology, Wright State University, Dayton, OH
| |
Collapse
|
26
|
Characterization of a novel LQT3 variant with a selective efficacy of mexiletine treatment. Sci Rep 2019; 9:12997. [PMID: 31506521 PMCID: PMC6736863 DOI: 10.1038/s41598-019-49450-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 08/23/2019] [Indexed: 12/19/2022] Open
Abstract
Pathogenic variants in the human SCN5A gene encoding the a-subunit of the principle Na+ channel (Nav1.5) are associated with long QT syndrome (LQTS) 3. LQT3 patients display variable responses to Na+ channel blockers demanding for the development of variant-specific therapeutic strategies. Here we performed a combined electrophysiological analysis with in silico simulation of variant channel to elucidate mechanisms of therapeutic responsiveness. We identified a novel SCN5A variant (A1656D) in a LQTS patient with a distinct response to mexiletine resulting in suppression of non-sustained ventricular tachycardia and manifestation of premature atrial contraction. Patch clamp analysis revealed that A1656D variant exerted gain-of-function effects including hyperpolarizing shift of the voltage-dependence of activation, depolarizing shift in the voltage-dependence of inactivation, and slowing of fast inactivation. Among ranolazine, flecainide, and mexiletine, only mexiletine restored inactivation kinetics of A1656D currents. In silico simulation to assess the effect of A1656D variant on ventricular cardiac cell excitation predicted a prolonged action potential which is consistent with the prolonged QT and non-sustained ventricular tachycardia of the patient. It also predicted that only mexiletine suppressed the prolonged action potential of human ventricular myocytes expressing A1656D. These data elucidate the underlying mechanism of the distinct response to mexiletine in this patient.
Collapse
|
27
|
Johansson JK, Karema-Jokinen VI, Hakanen S, Jylhä A, Uusitalo H, Vihinen-Ranta M, Skottman H, Ihalainen TO, Nymark S. Sodium channels enable fast electrical signaling and regulate phagocytosis in the retinal pigment epithelium. BMC Biol 2019; 17:63. [PMID: 31412898 PMCID: PMC6694495 DOI: 10.1186/s12915-019-0681-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 07/11/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Voltage-gated sodium (Nav) channels have traditionally been considered a trademark of excitable cells. However, recent studies have shown the presence of Nav channels in several non-excitable cells, such as astrocytes and macrophages, demonstrating that the roles of these channels are more diverse than was previously thought. Despite the earlier discoveries, the presence of Nav channel-mediated currents in the cells of retinal pigment epithelium (RPE) has been dismissed as a cell culture artifact. We challenge this notion by investigating the presence and possible role of Nav channels in RPE both ex vivo and in vitro. RESULTS Our work demonstrates that several subtypes of Nav channels are found in human embryonic stem cell (hESC)-derived and mouse RPE, most prominently subtypes Nav1.4, Nav1.6, and Nav1.8. Whole cell patch clamp recordings from the hESC-derived RPE monolayers showed that the current was inhibited by TTX and QX-314 and was sensitive to the selective blockers of the main Nav subtypes. Importantly, we show that the Nav channels are involved in photoreceptor outer segment phagocytosis since blocking their activity significantly reduces the efficiency of particle internalization. Consistent with this role, our electron microscopy results and immunocytochemical analysis show that Nav1.4 and Nav1.8 accumulate on phagosomes and that pharmacological inhibition of Nav channels as well as silencing the expression of Nav1.4 with shRNA impairs the phagocytosis process. CONCLUSIONS Taken together, our study shows that Nav channels are present in RPE, giving this tissue the capacity of fast electrical signaling. The channels are critical for the physiology of RPE with an important role in photoreceptor outer segment phagocytosis.
Collapse
Affiliation(s)
- Julia K Johansson
- BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Viivi I Karema-Jokinen
- BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Satu Hakanen
- Department of Biological and Environmental Science and Nanoscience Center, University of Jyväskylä, Jyväskylä, Finland
| | - Antti Jylhä
- BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Hannu Uusitalo
- BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Tays Eye Centre, Tampere University Hospital, Tampere, Finland
| | - Maija Vihinen-Ranta
- Department of Biological and Environmental Science and Nanoscience Center, University of Jyväskylä, Jyväskylä, Finland
| | - Heli Skottman
- BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Teemu O Ihalainen
- BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Soile Nymark
- BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.
| |
Collapse
|
28
|
Li B, Silva JR, Lu X, Luo L, Wang Y, Xu L, Aierken A, Shynykul Z, Kamau PM, Luo A, Yang J, Su D, Yang F, Cui J, Yang S, Lai R. Molecular game theory for a toxin-dominant food chain model. Natl Sci Rev 2019; 6:1191-1200. [PMID: 34691998 PMCID: PMC8291550 DOI: 10.1093/nsr/nwz097] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 06/23/2019] [Accepted: 07/02/2019] [Indexed: 12/30/2022] Open
Abstract
Animal toxins that are used to subdue prey and deter predators act as the key drivers in natural food chains and ecosystems. However, the predators of venomous animals may exploit feeding adaptation strategies to overcome toxins their prey produce. Much remains unknown about the genetic and molecular game process in the toxin-dominant food chain model. Here, we show an evolutionary strategy in different trophic levels of scorpion-eating amphibians, scorpions and insects, representing each predation relationship in habitats dominated by the paralytic toxins of scorpions. For scorpions preying on insects, we found that the scorpion α-toxins irreversibly activate the skeletal muscle sodium channel of their prey (insect, BgNaV1) through a membrane delivery mechanism and an efficient binding with the Asp/Lys-Tyr motif of BgNaV1. However, in the predatory game between frogs and scorpions, with a single point mutation (Lys to Glu) in this motif of the frog's skeletal muscle sodium channel (fNaV1.4), fNaV1.4 breaks this interaction and diminishes muscular toxicity to the frog; thus, frogs can regularly prey on scorpions without showing paralysis. Interestingly, this molecular strategy also has been employed by some other scorpion-eating amphibians, especially anurans. In contrast to these amphibians, the Asp/Lys-Tyr motifs are structurally and functionally conserved in other animals that do not prey on scorpions. Together, our findings elucidate the protein-protein interacting mechanism of a toxin-dominant predator-prey system, implying the evolutionary game theory at a molecular level.
Collapse
Affiliation(s)
- Bowen Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Kunming 650223, China
| | - Jonathan R Silva
- Department of Biomedical Engineering, Washington University St. Louis, MO 63130, UK
| | - Xiancui Lu
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Kunming 650223, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lei Luo
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Kunming 650223, China
| | - Yunfei Wang
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Kunming 650223, China
| | - Lizhen Xu
- Key Laboratory of Medical Neurobiology, Department of Biophysics and Kidney Disease Center, First Affiliated Hospital, Institute of Neuroscience, National Health Commission and Chinese Academy of Medical Sciences, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Aerziguli Aierken
- Key Laboratory of Medical Neurobiology, Department of Biophysics and Kidney Disease Center, First Affiliated Hospital, Institute of Neuroscience, National Health Commission and Chinese Academy of Medical Sciences, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Zhanserik Shynykul
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Kunming 650223, China
| | - Peter Muiruri Kamau
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Kunming 650223, China
| | - Anna Luo
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Kunming 650223, China
| | - Jian Yang
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Kunming 650223, China
- Department of Biological Sciences, Columbia University, New York, NY 10027, UK
| | - Deyuan Su
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Kunming 650223, China
| | - Fan Yang
- Key Laboratory of Medical Neurobiology, Department of Biophysics and Kidney Disease Center, First Affiliated Hospital, Institute of Neuroscience, National Health Commission and Chinese Academy of Medical Sciences, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Jianmin Cui
- Department of Biomedical Engineering, Washington University St. Louis, MO 63130, UK
| | - Shilong Yang
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Kunming 650223, China
| | - Ren Lai
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Kunming 650223, China
- KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming 650223, China
| |
Collapse
|
29
|
Abstract
Skeletal muscle channelopathies are rare heterogeneous diseases with marked genotypic and phenotypic variability. These disorders cause lifetime disability and impact quality of life. Despite advances in understanding of the molecular pathology of these disorders, the diverse phenotypic manifestations remain a challenge in diagnosis, therapeutic, genetic counseling, and research planning. Electrodiagnostic testing is useful in directing the diagnosis, but has several limitations: patient discomfort, time consuming, and significant overlap of findings in muscle channelopathies. Although genetic testing is the gold standard in making a definitive diagnosis, a mutation might not be identified in many patients with a well-supported clinical diagnosis of periodic paralysis. In the recent past, there have been landmark clinical trials in non-dystrophic myotonia and periodic paralysis which are encouraging as they demonstrate the ability of robust clinical research consortia to conduct well-controlled trials of rare diseases.
Collapse
Affiliation(s)
- Lauren Phillips
- Department of Neurology and Neurotherapeutics, UT Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, Texas, 75390, USA
| | - Jaya R Trivedi
- Department of Neurology and Neurotherapeutics, UT Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, Texas, 75390, USA.
| |
Collapse
|
30
|
Lower-extremity magnetic resonance imaging in patients with hyperkalemic periodic paralysis carrying the SCN4A mutation T704M: 30-month follow-up of seven patients. Neuromuscul Disord 2018; 28:837-845. [DOI: 10.1016/j.nmd.2018.06.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 06/20/2018] [Accepted: 06/22/2018] [Indexed: 02/07/2023]
|
31
|
Marakhonov AV, Varenikov GG, Skoblov MY. Sodium Channelopathies: From Molecular Physiology towards Medical Genetics. RUSS J GENET+ 2018. [DOI: 10.1134/s102279541801009x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
32
|
Abstract
The periodic paralyses are a group of skeletal muscle channelopathies characterizeed by intermittent attacks of muscle weakness often associated with altered serum potassium levels. The underlying genetic defects include mutations in genes encoding the skeletal muscle calcium channel Cav1.1, sodium channel Nav1.4, and potassium channels Kir2.1, Kir3.4, and possibly Kir2.6. Our increasing knowledge of how mutant channels affect muscle excitability has resulted in better understanding of many clinical phenomena which have been known for decades and sheds light on some of the factors that trigger attacks. Insights into the pathophysiology are also leading to new therapeutic approaches.
Collapse
Affiliation(s)
- Doreen Fialho
- MRC Centre for Neuromuscular Diseases, National Hospital for Neurology and Neurosurgery, London, United Kingdom
| | - Robert C Griggs
- Department of Neurology, University of Rochester School of Medicine and Dentistry, Rochester, NY, United States.
| | - Emma Matthews
- MRC Centre for Neuromuscular Diseases, National Hospital for Neurology and Neurosurgery, London, United Kingdom
| |
Collapse
|
33
|
Na V Channels: Assaying Biosynthesis, Trafficking, Function. Methods Mol Biol 2017. [PMID: 29264805 DOI: 10.1007/978-1-4939-7553-2_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Integral to the cell surface is channels, pumps, and exchanger proteins that facilitate the movement of ions across the membrane. Ion channels facilitate the passive movement of ions down an electrochemical gradient. Ion pumps actively use energy to actively translocate ions, often against concentration or voltage gradients, while ion exchangers utilize energy to couple the transport of different ion species such that one ion moves down its gradient and the released free energy is used to drive the movement of a different ion against its electrochemical gradient. Some ion pumps and exchangers may be electrogenic, i.e., the ion transport they support is not electrically neutral and generates a current. Functions of these pore-forming membrane proteins include the establishment of membrane potentials, gating of ions flows across the cell membrane to elicit action potentials and other electrical signals, as well as the regulation of cell volumes. The major forms of ion channels include voltage-, ligand-, and signal-gated channels. In this review, we describe mammalian voltage dependent Na (NaV) channels.
Collapse
|
34
|
Symonds JD, Zuberi SM. Genetics update: Monogenetics, polygene disorders and the quest for modifying genes. Neuropharmacology 2017; 132:3-19. [PMID: 29037745 DOI: 10.1016/j.neuropharm.2017.10.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 10/09/2017] [Accepted: 10/11/2017] [Indexed: 12/19/2022]
Abstract
The genetic channelopathies are a broad collection of diseases. Many ion channel genes demonstrate wide phenotypic pleiotropy, but nonetheless concerted efforts have been made to characterise genotype-phenotype relationships. In this review we give an overview of the factors that influence genotype-phenotype relationships across this group of diseases as a whole, using specific individual channelopathies as examples. We suggest reasons for the limitations observed in these relationships. We discuss the role of ion channel variation in polygenic disease and highlight research that has contributed to unravelling the complex aetiological nature of these conditions. We focus specifically on the quest for modifying genes in inherited channelopathies, using the voltage-gated sodium channels as an example. Epilepsy related to genetic channelopathy is one area in which precision medicine is showing promise. We will discuss the successes and limitations of precision medicine in these conditions. This article is part of the Special Issue entitled 'Channelopathies.'
Collapse
Affiliation(s)
- Joseph D Symonds
- The Paediatric Neurosciences Research Group, Royal Hospital for Children, Queen Elizabeth University Hospitals, Glasgow, UK; School of Medicine, University of Glasgow, Glasgow, UK
| | - Sameer M Zuberi
- The Paediatric Neurosciences Research Group, Royal Hospital for Children, Queen Elizabeth University Hospitals, Glasgow, UK; School of Medicine, University of Glasgow, Glasgow, UK.
| |
Collapse
|
35
|
Abstract
Epileptic encephalopathies account for a large proportion of the intractable early-onset epilepsies and are characterized by frequent seizures and poor developmental outcome. The epileptic encephalopathies can be loosely divided into two related groups of named syndromes. The first comprises epilepsies where continuous EEG changes directly result in cognitive and developmental dysfunction. The second includes patients where cognitive impairment is present at seizure onset and is due to the underlying etiology but the epileptic activity may then worsen the cognitive abilities over time. Recent, large-scale exome studies have begun to establish the genetic architecture of the epileptic encephalopathies, resulting in a re-consideration of the boundaries of these named syndromes. The emergence of this genetic architecture has lead to three main pathophysiological concepts to provide a mechanistic framework for these disorders. In this article, we will review the classic syndromes, the most significant genetic findings, and relate both to the pathophysiological understanding of epileptic encephalopathies.
Collapse
|
36
|
Symonds JD, Zuberi SM. WITHDRAWN: Genetics update: Monogenetics, polygene disorders and the quest for modifying genes. Neuropharmacology 2017:S0028-3908(17)30347-7. [PMID: 28757052 DOI: 10.1016/j.neuropharm.2017.07.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 07/17/2017] [Indexed: 11/15/2022]
Abstract
The Publisher regrets that this article is an accidental duplication of an article that has already been published, https://doi.org/10.1016/j.neuropharm.2017.10.013. The duplicate article has therefore been withdrawn. The full Elsevier Policy on Article Withdrawal can be found at https://www.elsevier.com/about/our-business/policies/article-withdrawal.
Collapse
Affiliation(s)
- Joseph D Symonds
- The Paediatric Neurosciences Research Group, Royal Hospital for Children, Queen Elizabeth University Hospitals, Glasgow, UK; School of Medicine, University of Glasgow, Glasgow, UK
| | - Sameer M Zuberi
- The Paediatric Neurosciences Research Group, Royal Hospital for Children, Queen Elizabeth University Hospitals, Glasgow, UK; School of Medicine, University of Glasgow, Glasgow, UK
| |
Collapse
|
37
|
Abstract
The epileptic encephalopathies are severe and often treatment-resistant conditions that are associated with a progressive disturbance of brain function, resulting in a broad range of neurological and non-neurological comorbidities. The concept of epileptic encephalopathies entails that the encephalopathy aspect of the overall condition is primarily driven by the epileptic activity of the disease, which often manifests as specific and pathological features on the electroencephalogram. Genetic factors in epileptic encephalopathies are increasingly recognized. As of 2016, more than 30 genes have been securely implicated as causative genes for genetic epileptic encephalopathies. Even though the traditional concept of epileptic encephalopathies entails that the progressive disturbance of brain dysfunction is primarily due to the abnormal hypersynchronous activity that underlies the seizure disorders, this strict concept rarely holds true for patients with identified genetic etiologies. More commonly, an underlying genetic etiology is thought to predispose both to the neurodevelopmental comorbidities and to the seizure phenotype with a complex interaction between both. In this chapter, we will elucidate to what extent neurodegeneration rather than epilepsy-related regression is a feature of the common epileptic encephalopathies, drawing parallels between two relatively separate fields of neurogenetic research.
Collapse
|
38
|
Kullmann D, Houlden H, Lunn M. Mechanisms of Neurological Disease. Neurology 2016. [DOI: 10.1002/9781118486160.ch3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
|
39
|
Chung KW, Yoo DH, Lee SJ, Choi BO, Lee SS. A Large Dominant Myotonia Congenita Family with a V1293I Mutation in SCN4A. J Clin Neurol 2016; 12:509-511. [PMID: 27486940 PMCID: PMC5063882 DOI: 10.3988/jcn.2016.12.4.509] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 02/27/2016] [Accepted: 02/29/2016] [Indexed: 11/17/2022] Open
Affiliation(s)
- Ki Wha Chung
- Department of Biological Sciences, Kongju National University, Gongju, Korea
| | - Da Hye Yoo
- Department of Biological Sciences, Kongju National University, Gongju, Korea
| | - Soo Jung Lee
- Department of Biological Sciences, Kongju National University, Gongju, Korea
| | - Byung Ok Choi
- Department of Neurology, Samsung Medical Center, Samsung Advanced Institute for Health Science & Tech, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Sang Soo Lee
- Department of Neurology, Chungbuk National University College of Medicine, Cheongju, Korea.
| |
Collapse
|
40
|
Ohno K, Ohkawara B, Ito M. Recent advances in congenital myasthenic syndromes. ACTA ACUST UNITED AC 2016. [DOI: 10.1111/cen3.12316] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Affiliation(s)
- Kinji Ohno
- Division of Neurogenetics; Center for Neurological Diseases and Cancer; Nagoya University Graduate School of Medicine; Nagoya Japan
| | - Bisei Ohkawara
- Division of Neurogenetics; Center for Neurological Diseases and Cancer; Nagoya University Graduate School of Medicine; Nagoya Japan
| | - Mikako Ito
- Division of Neurogenetics; Center for Neurological Diseases and Cancer; Nagoya University Graduate School of Medicine; Nagoya Japan
| |
Collapse
|
41
|
Shimomura T, Irie K, Fujiyoshi Y. Molecular determinants of prokaryotic voltage-gated sodium channels for recognition of local anesthetics. FEBS J 2016; 283:2881-95. [DOI: 10.1111/febs.13776] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2016] [Revised: 05/22/2016] [Accepted: 06/06/2016] [Indexed: 11/28/2022]
Affiliation(s)
- Takushi Shimomura
- Cellular and Structural Physiology Institute (CeSPI); Nagoya University; Japan
| | - Katsumasa Irie
- Cellular and Structural Physiology Institute (CeSPI); Nagoya University; Japan
- Graduate School of Pharmaceutical Science; Nagoya University; Japan
| | - Yoshinori Fujiyoshi
- Cellular and Structural Physiology Institute (CeSPI); Nagoya University; Japan
- Graduate School of Pharmaceutical Science; Nagoya University; Japan
| |
Collapse
|
42
|
Abstract
PURPOSE OF REVIEW This is an update on skeletal muscle sodium channelopathies since knowledge in the field have dramatically increased in the past years. RECENT FINDING The relationship between two phenotypes and SCN4A has been confirmed with additional cases that remain extremely rare: severe neonatal episodic laryngospasm mimicking encephalopathy, which should be actively searched for since patients respond well to sodium channel blockers; congenital myasthenic syndromes, which have the particularity to be the first recessive Nav1.4 channelopathy. Deep DNA sequencing suggests the contribution of other ion channels in the clinical expressivity of sodium channelopathies, which may be one of the factors modulating the latter. The increased knowledge of channel molecular structure, the quantity of sodium channel blockers, and the availability of preclinical models would permit a most personalized choice of medication for patients suffering from these debilitating neuromuscular diseases. SUMMARY Advances in the understanding of the molecular structure of voltage-gated sodium channels, as well as availability of preclinical models, would lead to improved medical care of patients suffering from skeletal muscle, as well as other sodium channelopathies.
Collapse
|
43
|
Jen JC, Ashizawa T, Griggs RC, Waters MF. Rare neurological channelopathies--networks to study patients, pathogenesis and treatment. Nat Rev Neurol 2016; 12:195-203. [PMID: 26943780 DOI: 10.1038/nrneurol.2016.18] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Each of the thousands of rare neurological diseases requires a widely distributed network of centres, investigators and patients, so as to foster multidisciplinary investigations and involve sufficient numbers of patients in the discovery of disease pathogenesis and novel treatment. In this Review, we highlight the value of this collaborative approach in patient-oriented research into rare neurological channelopathies. Two networks, the Consortium for Clinical Investigations of Neurological Channelopathies (CINCH) and the Clinical Research Consortium for Studies of Cerebellar Ataxias (CRC-SCA), provide a link between patients with rare channelopathies and investigators who are studying disease pathogenesis and developing novel treatments. Interactions between patients, researchers and advocacy groups promote shared agendas that benefit patient education and recruitment, research collaboration and funding, and training and mentoring of junior investigators who are attracted to the study of the diseases that provide the focus for the two networks. Here, we discuss how linkage of national and international centres has enabled recruitment of study participants, provided opportunities for novel studies of pathogenesis, and facilitated successful clinical trials.
Collapse
Affiliation(s)
- Joanna C Jen
- Departments of Neurology and Neurobiology, David Geffen School of Medicine, University of California, 710 Westwood Plaza, Los Angeles, California 90095, USA
| | - Tetsuo Ashizawa
- McKnight Brain Institute &Department of Neurology, University of Florida, 1149 South Newell Drive, Gainsville, Florida 32611, USA
| | - Robert C Griggs
- Departments of Neurology, Medicine, Pathology and Laboratory Medicine, and Pediatrics, and Center for Human Experimental Therapeutics, University of Rochester School of Medicine and Dentistry, 601 Elmwood Avenue, Box CU 420669, Rochester, New York 14642, USA
| | - Michael F Waters
- Departments of Neurology, Neuroscience and Biomedical Engineering, College of Medicine, University of Florida, 100 Newell Road, Gainesville, Florida 32610, USA
| |
Collapse
|
44
|
Sansone VA, Burge J, McDermott MP, Smith PC, Herr B, Tawil R, Pandya S, Kissel J, Ciafaloni E, Shieh P, Ralph JW, Amato A, Cannon SC, Trivedi J, Barohn R, Crum B, Mitsumoto H, Pestronk A, Meola G, Conwit R, Hanna MG, Griggs RC. Randomized, placebo-controlled trials of dichlorphenamide in periodic paralysis. Neurology 2016; 86:1408-1416. [PMID: 26865514 DOI: 10.1212/wnl.0000000000002416] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 12/15/2015] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To determine the short-term and long-term effects of dichlorphenamide (DCP) on attack frequency and quality of life in hyperkalemic (HYP) and hypokalemic (HOP) periodic paralysis. METHODS Two multicenter randomized, double-blind, placebo-controlled trials lasted 9 weeks (Class I evidence), followed by a 1-year extension phase in which all participants received DCP. Forty-four HOP and 21 HYP participants participated. The primary outcome variable was the average number of attacks per week over the final 8 weeks of the double-blind phase. RESULTS The median attack rate was lower in HOP participants on DCP than in participants on placebo (0.3 vs 2.4, p = 0.02). The 9-week mean change in the Physical Component Summary score of the Short Form-36 was also better in HOP participants receiving DCP (treatment effect = 7.29 points, 95% confidence interval 2.26 to 12.32, p = 0.006). The median attack rate was also lower in HYP participants on DCP (0.9 vs 4.8) than in participants on placebo, but the difference in median attack rate was not significant (p = 0.10). There were no significant effects of DCP on muscle strength or muscle mass in either trial. The most common adverse events in both trials were paresthesia (47% DCP vs 14% placebo, both trials combined) and confusion (19% DCP vs 7% placebo, both trials combined). CONCLUSIONS DCP is effective in reducing the attack frequency, is safe, and improves quality of life in HOP periodic paralysis. CLASSIFICATION OF EVIDENCE These studies provide Class I evidence that DCP significantly reduces attack frequency in HOP but lacked the precision to support either efficacy or lack of efficacy of DCP in HYP.
Collapse
Affiliation(s)
- Valeria A Sansone
- From NEMO Clinical Center (V.A.S.) and IRCCS Policlinico San Donato (G.M.), University of Milan, Italy; MRC Centre for Neuromuscular Diseases (J.B., M.G.H.), UCL Institute of Neurology, Queen Square, London, UK; University of Rochester (M.P.M., P.C.S., B.H., R.T., S.P., E.C., R.C.G.), NY; Ohio State University (J.K.), Columbus; UCLA Medical Center (P.S.), Los Angeles, CA; University of California San Francisco School of Medicine (J.W.R.); Brigham and Women's Hospital (A.A.), Boston, MA; UT Southwestern Medical Center (S.C.C., J.T.), Dallas, TX; University of Kansas Medical Center (R.B.), Kansas City; Mayo Clinic (B.C.), Rochester MN; Columbia University (H.M.), New York, NY; Washington University (A.P.), St. Louis, MO; and the Office of Clinical Research (R.C.), NINDS, Bethesda, MD.
| | - James Burge
- From NEMO Clinical Center (V.A.S.) and IRCCS Policlinico San Donato (G.M.), University of Milan, Italy; MRC Centre for Neuromuscular Diseases (J.B., M.G.H.), UCL Institute of Neurology, Queen Square, London, UK; University of Rochester (M.P.M., P.C.S., B.H., R.T., S.P., E.C., R.C.G.), NY; Ohio State University (J.K.), Columbus; UCLA Medical Center (P.S.), Los Angeles, CA; University of California San Francisco School of Medicine (J.W.R.); Brigham and Women's Hospital (A.A.), Boston, MA; UT Southwestern Medical Center (S.C.C., J.T.), Dallas, TX; University of Kansas Medical Center (R.B.), Kansas City; Mayo Clinic (B.C.), Rochester MN; Columbia University (H.M.), New York, NY; Washington University (A.P.), St. Louis, MO; and the Office of Clinical Research (R.C.), NINDS, Bethesda, MD
| | - Michael P McDermott
- From NEMO Clinical Center (V.A.S.) and IRCCS Policlinico San Donato (G.M.), University of Milan, Italy; MRC Centre for Neuromuscular Diseases (J.B., M.G.H.), UCL Institute of Neurology, Queen Square, London, UK; University of Rochester (M.P.M., P.C.S., B.H., R.T., S.P., E.C., R.C.G.), NY; Ohio State University (J.K.), Columbus; UCLA Medical Center (P.S.), Los Angeles, CA; University of California San Francisco School of Medicine (J.W.R.); Brigham and Women's Hospital (A.A.), Boston, MA; UT Southwestern Medical Center (S.C.C., J.T.), Dallas, TX; University of Kansas Medical Center (R.B.), Kansas City; Mayo Clinic (B.C.), Rochester MN; Columbia University (H.M.), New York, NY; Washington University (A.P.), St. Louis, MO; and the Office of Clinical Research (R.C.), NINDS, Bethesda, MD
| | - Patty C Smith
- From NEMO Clinical Center (V.A.S.) and IRCCS Policlinico San Donato (G.M.), University of Milan, Italy; MRC Centre for Neuromuscular Diseases (J.B., M.G.H.), UCL Institute of Neurology, Queen Square, London, UK; University of Rochester (M.P.M., P.C.S., B.H., R.T., S.P., E.C., R.C.G.), NY; Ohio State University (J.K.), Columbus; UCLA Medical Center (P.S.), Los Angeles, CA; University of California San Francisco School of Medicine (J.W.R.); Brigham and Women's Hospital (A.A.), Boston, MA; UT Southwestern Medical Center (S.C.C., J.T.), Dallas, TX; University of Kansas Medical Center (R.B.), Kansas City; Mayo Clinic (B.C.), Rochester MN; Columbia University (H.M.), New York, NY; Washington University (A.P.), St. Louis, MO; and the Office of Clinical Research (R.C.), NINDS, Bethesda, MD
| | - Barbara Herr
- From NEMO Clinical Center (V.A.S.) and IRCCS Policlinico San Donato (G.M.), University of Milan, Italy; MRC Centre for Neuromuscular Diseases (J.B., M.G.H.), UCL Institute of Neurology, Queen Square, London, UK; University of Rochester (M.P.M., P.C.S., B.H., R.T., S.P., E.C., R.C.G.), NY; Ohio State University (J.K.), Columbus; UCLA Medical Center (P.S.), Los Angeles, CA; University of California San Francisco School of Medicine (J.W.R.); Brigham and Women's Hospital (A.A.), Boston, MA; UT Southwestern Medical Center (S.C.C., J.T.), Dallas, TX; University of Kansas Medical Center (R.B.), Kansas City; Mayo Clinic (B.C.), Rochester MN; Columbia University (H.M.), New York, NY; Washington University (A.P.), St. Louis, MO; and the Office of Clinical Research (R.C.), NINDS, Bethesda, MD
| | - Rabi Tawil
- From NEMO Clinical Center (V.A.S.) and IRCCS Policlinico San Donato (G.M.), University of Milan, Italy; MRC Centre for Neuromuscular Diseases (J.B., M.G.H.), UCL Institute of Neurology, Queen Square, London, UK; University of Rochester (M.P.M., P.C.S., B.H., R.T., S.P., E.C., R.C.G.), NY; Ohio State University (J.K.), Columbus; UCLA Medical Center (P.S.), Los Angeles, CA; University of California San Francisco School of Medicine (J.W.R.); Brigham and Women's Hospital (A.A.), Boston, MA; UT Southwestern Medical Center (S.C.C., J.T.), Dallas, TX; University of Kansas Medical Center (R.B.), Kansas City; Mayo Clinic (B.C.), Rochester MN; Columbia University (H.M.), New York, NY; Washington University (A.P.), St. Louis, MO; and the Office of Clinical Research (R.C.), NINDS, Bethesda, MD
| | - Shree Pandya
- From NEMO Clinical Center (V.A.S.) and IRCCS Policlinico San Donato (G.M.), University of Milan, Italy; MRC Centre for Neuromuscular Diseases (J.B., M.G.H.), UCL Institute of Neurology, Queen Square, London, UK; University of Rochester (M.P.M., P.C.S., B.H., R.T., S.P., E.C., R.C.G.), NY; Ohio State University (J.K.), Columbus; UCLA Medical Center (P.S.), Los Angeles, CA; University of California San Francisco School of Medicine (J.W.R.); Brigham and Women's Hospital (A.A.), Boston, MA; UT Southwestern Medical Center (S.C.C., J.T.), Dallas, TX; University of Kansas Medical Center (R.B.), Kansas City; Mayo Clinic (B.C.), Rochester MN; Columbia University (H.M.), New York, NY; Washington University (A.P.), St. Louis, MO; and the Office of Clinical Research (R.C.), NINDS, Bethesda, MD
| | - John Kissel
- From NEMO Clinical Center (V.A.S.) and IRCCS Policlinico San Donato (G.M.), University of Milan, Italy; MRC Centre for Neuromuscular Diseases (J.B., M.G.H.), UCL Institute of Neurology, Queen Square, London, UK; University of Rochester (M.P.M., P.C.S., B.H., R.T., S.P., E.C., R.C.G.), NY; Ohio State University (J.K.), Columbus; UCLA Medical Center (P.S.), Los Angeles, CA; University of California San Francisco School of Medicine (J.W.R.); Brigham and Women's Hospital (A.A.), Boston, MA; UT Southwestern Medical Center (S.C.C., J.T.), Dallas, TX; University of Kansas Medical Center (R.B.), Kansas City; Mayo Clinic (B.C.), Rochester MN; Columbia University (H.M.), New York, NY; Washington University (A.P.), St. Louis, MO; and the Office of Clinical Research (R.C.), NINDS, Bethesda, MD
| | - Emma Ciafaloni
- From NEMO Clinical Center (V.A.S.) and IRCCS Policlinico San Donato (G.M.), University of Milan, Italy; MRC Centre for Neuromuscular Diseases (J.B., M.G.H.), UCL Institute of Neurology, Queen Square, London, UK; University of Rochester (M.P.M., P.C.S., B.H., R.T., S.P., E.C., R.C.G.), NY; Ohio State University (J.K.), Columbus; UCLA Medical Center (P.S.), Los Angeles, CA; University of California San Francisco School of Medicine (J.W.R.); Brigham and Women's Hospital (A.A.), Boston, MA; UT Southwestern Medical Center (S.C.C., J.T.), Dallas, TX; University of Kansas Medical Center (R.B.), Kansas City; Mayo Clinic (B.C.), Rochester MN; Columbia University (H.M.), New York, NY; Washington University (A.P.), St. Louis, MO; and the Office of Clinical Research (R.C.), NINDS, Bethesda, MD
| | - Perry Shieh
- From NEMO Clinical Center (V.A.S.) and IRCCS Policlinico San Donato (G.M.), University of Milan, Italy; MRC Centre for Neuromuscular Diseases (J.B., M.G.H.), UCL Institute of Neurology, Queen Square, London, UK; University of Rochester (M.P.M., P.C.S., B.H., R.T., S.P., E.C., R.C.G.), NY; Ohio State University (J.K.), Columbus; UCLA Medical Center (P.S.), Los Angeles, CA; University of California San Francisco School of Medicine (J.W.R.); Brigham and Women's Hospital (A.A.), Boston, MA; UT Southwestern Medical Center (S.C.C., J.T.), Dallas, TX; University of Kansas Medical Center (R.B.), Kansas City; Mayo Clinic (B.C.), Rochester MN; Columbia University (H.M.), New York, NY; Washington University (A.P.), St. Louis, MO; and the Office of Clinical Research (R.C.), NINDS, Bethesda, MD
| | - Jeffrey W Ralph
- From NEMO Clinical Center (V.A.S.) and IRCCS Policlinico San Donato (G.M.), University of Milan, Italy; MRC Centre for Neuromuscular Diseases (J.B., M.G.H.), UCL Institute of Neurology, Queen Square, London, UK; University of Rochester (M.P.M., P.C.S., B.H., R.T., S.P., E.C., R.C.G.), NY; Ohio State University (J.K.), Columbus; UCLA Medical Center (P.S.), Los Angeles, CA; University of California San Francisco School of Medicine (J.W.R.); Brigham and Women's Hospital (A.A.), Boston, MA; UT Southwestern Medical Center (S.C.C., J.T.), Dallas, TX; University of Kansas Medical Center (R.B.), Kansas City; Mayo Clinic (B.C.), Rochester MN; Columbia University (H.M.), New York, NY; Washington University (A.P.), St. Louis, MO; and the Office of Clinical Research (R.C.), NINDS, Bethesda, MD
| | - Antony Amato
- From NEMO Clinical Center (V.A.S.) and IRCCS Policlinico San Donato (G.M.), University of Milan, Italy; MRC Centre for Neuromuscular Diseases (J.B., M.G.H.), UCL Institute of Neurology, Queen Square, London, UK; University of Rochester (M.P.M., P.C.S., B.H., R.T., S.P., E.C., R.C.G.), NY; Ohio State University (J.K.), Columbus; UCLA Medical Center (P.S.), Los Angeles, CA; University of California San Francisco School of Medicine (J.W.R.); Brigham and Women's Hospital (A.A.), Boston, MA; UT Southwestern Medical Center (S.C.C., J.T.), Dallas, TX; University of Kansas Medical Center (R.B.), Kansas City; Mayo Clinic (B.C.), Rochester MN; Columbia University (H.M.), New York, NY; Washington University (A.P.), St. Louis, MO; and the Office of Clinical Research (R.C.), NINDS, Bethesda, MD
| | - Steve C Cannon
- From NEMO Clinical Center (V.A.S.) and IRCCS Policlinico San Donato (G.M.), University of Milan, Italy; MRC Centre for Neuromuscular Diseases (J.B., M.G.H.), UCL Institute of Neurology, Queen Square, London, UK; University of Rochester (M.P.M., P.C.S., B.H., R.T., S.P., E.C., R.C.G.), NY; Ohio State University (J.K.), Columbus; UCLA Medical Center (P.S.), Los Angeles, CA; University of California San Francisco School of Medicine (J.W.R.); Brigham and Women's Hospital (A.A.), Boston, MA; UT Southwestern Medical Center (S.C.C., J.T.), Dallas, TX; University of Kansas Medical Center (R.B.), Kansas City; Mayo Clinic (B.C.), Rochester MN; Columbia University (H.M.), New York, NY; Washington University (A.P.), St. Louis, MO; and the Office of Clinical Research (R.C.), NINDS, Bethesda, MD
| | - Jaya Trivedi
- From NEMO Clinical Center (V.A.S.) and IRCCS Policlinico San Donato (G.M.), University of Milan, Italy; MRC Centre for Neuromuscular Diseases (J.B., M.G.H.), UCL Institute of Neurology, Queen Square, London, UK; University of Rochester (M.P.M., P.C.S., B.H., R.T., S.P., E.C., R.C.G.), NY; Ohio State University (J.K.), Columbus; UCLA Medical Center (P.S.), Los Angeles, CA; University of California San Francisco School of Medicine (J.W.R.); Brigham and Women's Hospital (A.A.), Boston, MA; UT Southwestern Medical Center (S.C.C., J.T.), Dallas, TX; University of Kansas Medical Center (R.B.), Kansas City; Mayo Clinic (B.C.), Rochester MN; Columbia University (H.M.), New York, NY; Washington University (A.P.), St. Louis, MO; and the Office of Clinical Research (R.C.), NINDS, Bethesda, MD
| | - Richard Barohn
- From NEMO Clinical Center (V.A.S.) and IRCCS Policlinico San Donato (G.M.), University of Milan, Italy; MRC Centre for Neuromuscular Diseases (J.B., M.G.H.), UCL Institute of Neurology, Queen Square, London, UK; University of Rochester (M.P.M., P.C.S., B.H., R.T., S.P., E.C., R.C.G.), NY; Ohio State University (J.K.), Columbus; UCLA Medical Center (P.S.), Los Angeles, CA; University of California San Francisco School of Medicine (J.W.R.); Brigham and Women's Hospital (A.A.), Boston, MA; UT Southwestern Medical Center (S.C.C., J.T.), Dallas, TX; University of Kansas Medical Center (R.B.), Kansas City; Mayo Clinic (B.C.), Rochester MN; Columbia University (H.M.), New York, NY; Washington University (A.P.), St. Louis, MO; and the Office of Clinical Research (R.C.), NINDS, Bethesda, MD
| | - Brian Crum
- From NEMO Clinical Center (V.A.S.) and IRCCS Policlinico San Donato (G.M.), University of Milan, Italy; MRC Centre for Neuromuscular Diseases (J.B., M.G.H.), UCL Institute of Neurology, Queen Square, London, UK; University of Rochester (M.P.M., P.C.S., B.H., R.T., S.P., E.C., R.C.G.), NY; Ohio State University (J.K.), Columbus; UCLA Medical Center (P.S.), Los Angeles, CA; University of California San Francisco School of Medicine (J.W.R.); Brigham and Women's Hospital (A.A.), Boston, MA; UT Southwestern Medical Center (S.C.C., J.T.), Dallas, TX; University of Kansas Medical Center (R.B.), Kansas City; Mayo Clinic (B.C.), Rochester MN; Columbia University (H.M.), New York, NY; Washington University (A.P.), St. Louis, MO; and the Office of Clinical Research (R.C.), NINDS, Bethesda, MD
| | - Hiroshi Mitsumoto
- From NEMO Clinical Center (V.A.S.) and IRCCS Policlinico San Donato (G.M.), University of Milan, Italy; MRC Centre for Neuromuscular Diseases (J.B., M.G.H.), UCL Institute of Neurology, Queen Square, London, UK; University of Rochester (M.P.M., P.C.S., B.H., R.T., S.P., E.C., R.C.G.), NY; Ohio State University (J.K.), Columbus; UCLA Medical Center (P.S.), Los Angeles, CA; University of California San Francisco School of Medicine (J.W.R.); Brigham and Women's Hospital (A.A.), Boston, MA; UT Southwestern Medical Center (S.C.C., J.T.), Dallas, TX; University of Kansas Medical Center (R.B.), Kansas City; Mayo Clinic (B.C.), Rochester MN; Columbia University (H.M.), New York, NY; Washington University (A.P.), St. Louis, MO; and the Office of Clinical Research (R.C.), NINDS, Bethesda, MD
| | - Alan Pestronk
- From NEMO Clinical Center (V.A.S.) and IRCCS Policlinico San Donato (G.M.), University of Milan, Italy; MRC Centre for Neuromuscular Diseases (J.B., M.G.H.), UCL Institute of Neurology, Queen Square, London, UK; University of Rochester (M.P.M., P.C.S., B.H., R.T., S.P., E.C., R.C.G.), NY; Ohio State University (J.K.), Columbus; UCLA Medical Center (P.S.), Los Angeles, CA; University of California San Francisco School of Medicine (J.W.R.); Brigham and Women's Hospital (A.A.), Boston, MA; UT Southwestern Medical Center (S.C.C., J.T.), Dallas, TX; University of Kansas Medical Center (R.B.), Kansas City; Mayo Clinic (B.C.), Rochester MN; Columbia University (H.M.), New York, NY; Washington University (A.P.), St. Louis, MO; and the Office of Clinical Research (R.C.), NINDS, Bethesda, MD
| | - Giovanni Meola
- From NEMO Clinical Center (V.A.S.) and IRCCS Policlinico San Donato (G.M.), University of Milan, Italy; MRC Centre for Neuromuscular Diseases (J.B., M.G.H.), UCL Institute of Neurology, Queen Square, London, UK; University of Rochester (M.P.M., P.C.S., B.H., R.T., S.P., E.C., R.C.G.), NY; Ohio State University (J.K.), Columbus; UCLA Medical Center (P.S.), Los Angeles, CA; University of California San Francisco School of Medicine (J.W.R.); Brigham and Women's Hospital (A.A.), Boston, MA; UT Southwestern Medical Center (S.C.C., J.T.), Dallas, TX; University of Kansas Medical Center (R.B.), Kansas City; Mayo Clinic (B.C.), Rochester MN; Columbia University (H.M.), New York, NY; Washington University (A.P.), St. Louis, MO; and the Office of Clinical Research (R.C.), NINDS, Bethesda, MD
| | - Robin Conwit
- From NEMO Clinical Center (V.A.S.) and IRCCS Policlinico San Donato (G.M.), University of Milan, Italy; MRC Centre for Neuromuscular Diseases (J.B., M.G.H.), UCL Institute of Neurology, Queen Square, London, UK; University of Rochester (M.P.M., P.C.S., B.H., R.T., S.P., E.C., R.C.G.), NY; Ohio State University (J.K.), Columbus; UCLA Medical Center (P.S.), Los Angeles, CA; University of California San Francisco School of Medicine (J.W.R.); Brigham and Women's Hospital (A.A.), Boston, MA; UT Southwestern Medical Center (S.C.C., J.T.), Dallas, TX; University of Kansas Medical Center (R.B.), Kansas City; Mayo Clinic (B.C.), Rochester MN; Columbia University (H.M.), New York, NY; Washington University (A.P.), St. Louis, MO; and the Office of Clinical Research (R.C.), NINDS, Bethesda, MD
| | - Michael G Hanna
- From NEMO Clinical Center (V.A.S.) and IRCCS Policlinico San Donato (G.M.), University of Milan, Italy; MRC Centre for Neuromuscular Diseases (J.B., M.G.H.), UCL Institute of Neurology, Queen Square, London, UK; University of Rochester (M.P.M., P.C.S., B.H., R.T., S.P., E.C., R.C.G.), NY; Ohio State University (J.K.), Columbus; UCLA Medical Center (P.S.), Los Angeles, CA; University of California San Francisco School of Medicine (J.W.R.); Brigham and Women's Hospital (A.A.), Boston, MA; UT Southwestern Medical Center (S.C.C., J.T.), Dallas, TX; University of Kansas Medical Center (R.B.), Kansas City; Mayo Clinic (B.C.), Rochester MN; Columbia University (H.M.), New York, NY; Washington University (A.P.), St. Louis, MO; and the Office of Clinical Research (R.C.), NINDS, Bethesda, MD
| | - Robert C Griggs
- From NEMO Clinical Center (V.A.S.) and IRCCS Policlinico San Donato (G.M.), University of Milan, Italy; MRC Centre for Neuromuscular Diseases (J.B., M.G.H.), UCL Institute of Neurology, Queen Square, London, UK; University of Rochester (M.P.M., P.C.S., B.H., R.T., S.P., E.C., R.C.G.), NY; Ohio State University (J.K.), Columbus; UCLA Medical Center (P.S.), Los Angeles, CA; University of California San Francisco School of Medicine (J.W.R.); Brigham and Women's Hospital (A.A.), Boston, MA; UT Southwestern Medical Center (S.C.C., J.T.), Dallas, TX; University of Kansas Medical Center (R.B.), Kansas City; Mayo Clinic (B.C.), Rochester MN; Columbia University (H.M.), New York, NY; Washington University (A.P.), St. Louis, MO; and the Office of Clinical Research (R.C.), NINDS, Bethesda, MD
| | | |
Collapse
|
45
|
|
46
|
Loussouarn G, Sternberg D, Nicole S, Marionneau C, Le Bouffant F, Toumaniantz G, Barc J, Malak OA, Fressart V, Péréon Y, Baró I, Charpentier F. Physiological and Pathophysiological Insights of Nav1.4 and Nav1.5 Comparison. Front Pharmacol 2016; 6:314. [PMID: 26834636 PMCID: PMC4712308 DOI: 10.3389/fphar.2015.00314] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 12/21/2015] [Indexed: 12/19/2022] Open
Abstract
Mutations in Nav1.4 and Nav1.5 α-subunits have been associated with muscular and cardiac channelopathies, respectively. Despite intense research on the structure and function of these channels, a lot of information is still missing to delineate the various physiological and pathophysiological processes underlying their activity at the molecular level. Nav1.4 and Nav1.5 sequences are similar, suggesting structural and functional homologies between the two orthologous channels. This also suggests that any characteristics described for one channel subunit may shed light on the properties of the counterpart channel subunit. In this review article, after a brief clinical description of the muscular and cardiac channelopathies related to Nav1.4 and Nav1.5 mutations, respectively, we compare the knowledge accumulated in different aspects of the expression and function of Nav1.4 and Nav1.5 α-subunits: the regulation of the two encoding genes (SCN4A and SCN5A), the associated/regulatory proteins and at last, the functional effect of the same missense mutations detected in Nav1.4 and Nav1.5. First, it appears that more is known on Nav1.5 expression and accessory proteins. Because of the high homologies of Nav1.5 binding sites and equivalent Nav1.4 sites, Nav1.5-related results may guide future investigations on Nav1.4. Second, the analysis of the same missense mutations in Nav1.4 and Nav1.5 revealed intriguing similarities regarding their effects on membrane excitability and alteration in channel biophysics. We believe that such comparison may bring new cues to the physiopathology of cardiac and muscular diseases.
Collapse
Affiliation(s)
- Gildas Loussouarn
- Institut National de la Santé et de la Recherche Médicale, UMR 1087, l'Institut du ThoraxNantes, France; Centre National de la Recherche Scientifique, UMR 6291Nantes, France; Université de NantesNantes, France
| | - Damien Sternberg
- Institut National de la Santé et de la Recherche Médicale, U1127Paris, France; Sorbonne Universités, Université Pierre-et-Marie-Curie, UMR S1127Paris, France; Centre National de la Recherche Scientifique, UMR 7225Paris, France; Institut du Cerveau et de la Moelle Épinière, ICMParis, France; Assistance Publique - Hôpitaux de Paris (AP-HP), Centres de Référence des Canalopathies Musculaires et des Maladies Neuro-musculaires Paris-EstParis, France; Assistance Publique - Hôpitaux de Paris (AP-HP), Hôpital de la Pitié Salpêtrière, Service de Biochimie Métabolique, Unité de Cardiogénétique et MyogénétiqueParis, France
| | - Sophie Nicole
- Institut National de la Santé et de la Recherche Médicale, U1127Paris, France; Sorbonne Universités, Université Pierre-et-Marie-Curie, UMR S1127Paris, France; Centre National de la Recherche Scientifique, UMR 7225Paris, France; Institut du Cerveau et de la Moelle Épinière, ICMParis, France
| | - Céline Marionneau
- Institut National de la Santé et de la Recherche Médicale, UMR 1087, l'Institut du ThoraxNantes, France; Centre National de la Recherche Scientifique, UMR 6291Nantes, France; Université de NantesNantes, France
| | - Francoise Le Bouffant
- Institut National de la Santé et de la Recherche Médicale, UMR 1087, l'Institut du ThoraxNantes, France; Centre National de la Recherche Scientifique, UMR 6291Nantes, France; Université de NantesNantes, France
| | - Gilles Toumaniantz
- Institut National de la Santé et de la Recherche Médicale, UMR 1087, l'Institut du ThoraxNantes, France; Centre National de la Recherche Scientifique, UMR 6291Nantes, France; Université de NantesNantes, France
| | - Julien Barc
- Institut National de la Santé et de la Recherche Médicale, UMR 1087, l'Institut du ThoraxNantes, France; Centre National de la Recherche Scientifique, UMR 6291Nantes, France; Université de NantesNantes, France
| | - Olfat A Malak
- Institut National de la Santé et de la Recherche Médicale, UMR 1087, l'Institut du ThoraxNantes, France; Centre National de la Recherche Scientifique, UMR 6291Nantes, France; Université de NantesNantes, France
| | - Véronique Fressart
- Assistance Publique - Hôpitaux de Paris (AP-HP), Hôpital de la Pitié Salpêtrière, Service de Biochimie Métabolique, Unité de Cardiogénétique et Myogénétique Paris, France
| | - Yann Péréon
- Centre Hospitalier Universitaire de Nantes, Centre de Référence Maladies Neuromusculaires Nantes-AngersNantes, France; Atlantic Gene Therapies - Biotherapy Institute for Rare DiseasesNantes, France
| | - Isabelle Baró
- Institut National de la Santé et de la Recherche Médicale, UMR 1087, l'Institut du ThoraxNantes, France; Centre National de la Recherche Scientifique, UMR 6291Nantes, France; Université de NantesNantes, France
| | - Flavien Charpentier
- Institut National de la Santé et de la Recherche Médicale, UMR 1087, l'Institut du ThoraxNantes, France; Centre National de la Recherche Scientifique, UMR 6291Nantes, France; Université de NantesNantes, France; Centre Hospitalier Universitaire de Nantes, l'Institut du ThoraxNantes, France
| |
Collapse
|
47
|
Abstract
Familial disorders of skeletal muscle excitability were initially described early in the last century and are now known to be caused by mutations of voltage-gated ion channels. The clinical manifestations are often striking, with an inability to relax after voluntary contraction (myotonia) or transient attacks of severe weakness (periodic paralysis). An essential feature of these disorders is fluctuation of symptoms that are strongly impacted by environmental triggers such as exercise, temperature, or serum K(+) levels. These phenomena have intrigued physiologists for decades, and in the past 25 years the molecular lesions underlying these disorders have been identified and mechanistic studies are providing insights for therapeutic strategies of disease modification. These familial disorders of muscle fiber excitability are "channelopathies" caused by mutations of a chloride channel (ClC-1), sodium channel (NaV1.4), calcium channel (CaV1.1), and several potassium channels (Kir2.1, Kir2.6, and Kir3.4). This review provides a synthesis of the mechanistic connections between functional defects of mutant ion channels, their impact on muscle excitability, how these changes cause clinical phenotypes, and approaches toward therapeutics.
Collapse
Affiliation(s)
- Stephen C Cannon
- Department of Physiology, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| |
Collapse
|
48
|
Lee YH, Lee HS, Lee HE, Hahn S, Nam TS, Shin HY, Choi YC, Kim SM. Whole-Body Muscle MRI in Patients with Hyperkalemic Periodic Paralysis Carrying the SCN4A Mutation T704M: Evidence for Chronic Progressive Myopathy with Selective Muscle Involvement. J Clin Neurol 2015; 11:331-8. [PMID: 26256659 PMCID: PMC4596100 DOI: 10.3988/jcn.2015.11.4.331] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Revised: 02/26/2015] [Accepted: 02/27/2015] [Indexed: 01/08/2023] Open
Abstract
Background and Purpose Hyperkalemic periodic paralysis (hyperKPP) is a muscle sodium-ion channelopathy characterized by recurrent paralytic attacks. A proportion of affected individuals develop fixed or chronic progressive weakness that results in significant disability. However, little is known about the pathology of hyperKPP-induced fixed weakness, including the pattern of muscle involvement. The aim of this study was to characterize the patterns of muscle involvement in hyperKPP by whole-body magnetic resonance imaging (MRI). Methods We performed whole-body muscle MRI in seven hyperKPP patients carrying the T704M mutation in the SCN4A skeletal sodium-channel gene. Muscle fat infiltration, suggestive of chronic progressive myopathy, was analyzed qualitatively using a grading system and was quantified by the two-point Dixon technique. Results Whole-body muscle MRI analysis revealed muscle atrophy and fatty infiltration in hyperKPP patients, especially in older individuals. Muscle involvement followed a selective pattern, primarily affecting the posterior compartment of the lower leg and anterior thigh muscles. The muscle fat fraction increased with patient age in the anterior thigh (r=0.669, p=0.009), in the deep posterior compartment of the lower leg (r=0.617, p=0.019), and in the superficial posterior compartment of the lower leg (r=0.777, p=0.001). Conclusions Our whole-body muscle MRI findings provide evidence for chronic progressive myopathy in hyperKPP patients. The reported data suggest that a selective pattern of muscle involvement-affecting the posterior compartment of the lower leg and the anterior thigh-is characteristic of chronic progressive myopathy in hyperKPP.
Collapse
Affiliation(s)
- Young Han Lee
- Department of Radiology, Research Institute of Radiological Science, Medical Convergence Research Institute, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Hyung Soo Lee
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea
| | - Hyo Eun Lee
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea
| | - Seok Hahn
- Department of Radiology, Research Institute of Radiological Science, Medical Convergence Research Institute, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Tai Seung Nam
- Department of Neurology, Chonnam National University Medical School, Gwangju, Korea
| | - Ha Young Shin
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea.
| | - Young Chul Choi
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea
| | - Seung Min Kim
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
49
|
Ptáček L, Jackson S. Louis Ptáček receives the 2015 ASCI/Stanley J. Korsmeyer Award. Interview by Sarah Jackson. J Clin Invest 2015; 125:1369-70. [PMID: 25831440 DOI: 10.1172/jci81185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
50
|
Protein mutated in paroxysmal dyskinesia interacts with the active zone protein RIM and suppresses synaptic vesicle exocytosis. Proc Natl Acad Sci U S A 2015; 112:2935-41. [PMID: 25730884 DOI: 10.1073/pnas.1501364112] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Paroxysmal nonkinesigenic dyskinesia (PNKD) is an autosomal dominant episodic movement disorder precipitated by coffee, alcohol, and stress. We previously identified the causative gene but the function of the encoded protein remains unknown. We also generated a PNKD mouse model that revealed dysregulated dopamine signaling in vivo. Here, we show that PNKD interacts with synaptic active zone proteins Rab3-interacting molecule (RIM)1 and RIM2, localizes to synapses, and modulates neurotransmitter release. Overexpressed PNKD protein suppresses release, and mutant PNKD protein is less effective than wild-type at inhibiting exocytosis. In PNKD KO mice, RIM1/2 protein levels are reduced and synaptic strength is impaired. Thus, PNKD is a novel synaptic protein with a regulatory role in neurotransmitter release.
Collapse
|