1
|
Canfora I, Altamura C, Desaphy JF, Boccanegra B, Vailati S, Caccia C, Melloni E, Padoani G, De Luca A, Pierno S. Preclinical study of the antimyotonic efficacy of safinamide in the myotonic mouse model. Neurotherapeutics 2024; 21:e00455. [PMID: 39322473 PMCID: PMC11586006 DOI: 10.1016/j.neurot.2024.e00455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 09/13/2024] [Accepted: 09/13/2024] [Indexed: 09/27/2024] Open
Abstract
Mexiletine is the first choice drug in the treatment of non-dystrophic myotonias. However, 30% of patients experience little benefit from mexiletine due to poor tolerability, contraindications and limited efficacy likely based on pharmacogenetic profile. Safinamide inhibits neuronal voltage-gated sodium and calcium channels and shows anticonvulsant activity, in addition to a reversible monoamine oxidase-B inhibition. We evaluated the preclinical effects of safinamide in an animal model of Myotonia Congenita, the ADR (arrested development of righting response) mouse. In vitro studies were performed using the two intracellular microelectrodes technique in current clamp mode. We analyzed sarcolemma excitability in skeletal muscle fibers isolated from male and female ADR (adr/adr) and from Wild-Type (wt/wt) mice, before and after the application of safinamide and the reference compound mexiletine. In ADR mice, the maximum number of action potentials (N-spikes) elicited by a fixed current is higher with respect to that of WT mice. Myotonic muscles show an involuntary firing of action potential called after-discharges. A more potent activity of safinamide compared to mexiletine has been demonstrated in reducing N-spikes and the after-discharges in myotonic muscle fibers. The time of righting reflex (TRR) before and after administration of safinamide and mexiletine was evaluated in vivo in ADR mice. Safinamide was able to reduce the TRR in ADR mice to a greater extent than mexiletine. In conclusion, safinamide counteracted the abnormal muscle hyperexcitability in myotonic mice both in vitro and in vivo suggesting it as an effective drug to be indicated in Myotonia Congenita.
Collapse
Affiliation(s)
- Ileana Canfora
- Department of Pharmacy & Drug Sciences, University of Bari Aldo Moro, Bari, Italy
| | - Concetta Altamura
- Department of Precision and Regenerative Medicine and Ionian Area, School of Medicine, University of Bari Aldo Moro, Bari, Italy
| | - Jean-Francois Desaphy
- Department of Precision and Regenerative Medicine and Ionian Area, School of Medicine, University of Bari Aldo Moro, Bari, Italy
| | - Brigida Boccanegra
- Department of Pharmacy & Drug Sciences, University of Bari Aldo Moro, Bari, Italy
| | - Silvia Vailati
- Global Medical Office and R&D, Zambon S.p.A., Bresso, MI, Italy
| | - Carla Caccia
- Global Medical Office and R&D, Zambon S.p.A., Bresso, MI, Italy
| | - Elsa Melloni
- Global Medical Office and R&D, Zambon S.p.A., Bresso, MI, Italy
| | - Gloria Padoani
- Global Medical Office and R&D, Zambon S.p.A., Bresso, MI, Italy
| | - Annamaria De Luca
- Department of Pharmacy & Drug Sciences, University of Bari Aldo Moro, Bari, Italy
| | - Sabata Pierno
- Department of Pharmacy & Drug Sciences, University of Bari Aldo Moro, Bari, Italy.
| |
Collapse
|
2
|
Tikhonova TB, Sharkov AA, Zhorov BS, Vassilevski AA. Diverse biophysical mechanisms in voltage-gated sodium channel Na v1.4 variants associated with myotonia. FASEB J 2024; 38:e23883. [PMID: 39150825 DOI: 10.1096/fj.202400867r] [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: 04/16/2024] [Revised: 07/14/2024] [Accepted: 07/31/2024] [Indexed: 08/18/2024]
Abstract
Mutations in SCN4A gene encoding Nav1.4 channel α-subunit, are known to cause neuromuscular disorders such as myotonia or paralysis. Here, we study the effect of two amino acid replacements, K1302Q and G1306E, in the DIII-IV loop of the channel, corresponding to mutations found in patients with myotonia. We combine clinical, electrophysiological, and molecular modeling data to provide a holistic picture of the molecular mechanisms operating in mutant channels and eventually leading to pathology. We analyze the existing clinical data for patients with the K1302Q substitution, which was reported for adults with or without myotonia phenotypes, and report two new unrelated patients with the G1306E substitution, who presented with severe neonatal episodic laryngospasm and childhood-onset myotonia. We provide a functional analysis of the mutant channels by expressing Nav1.4 α-subunit in Xenopus oocytes in combination with β1 subunit and recording sodium currents using two-electrode voltage clamp. The K1302Q variant exhibits abnormal voltage dependence of steady-state fast inactivation, being the likely cause of pathology. K1302Q does not lead to decelerated fast inactivation, unlike several other myotonic mutations such as G1306E. For both mutants, we observe increased window currents corresponding to a larger population of channels available for activation. To elaborate the structural rationale for our experimental data, we explore the contacts involving K/Q1302 and E1306 in the AlphaFold2 model of wild-type Nav1.4 and Monte Carlo-minimized models of mutant channels. Our data provide the missing evidence to support the classification of K1302Q variant as likely pathogenic and may be used by clinicians.
Collapse
Affiliation(s)
- Tatiana B Tikhonova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Artem A Sharkov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Veltischev Research and Clinical Institute for Pediatrics and Pediatric Surgery of the Pirogov Russian National Research Medical University, Moscow, Russia
- Genomed Ltd., Moscow, Russia
| | - Boris S Zhorov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Sechenov Institute of Evolutionary Physiology & Biochemistry, Russian Academy of Sciences, St. Petersburg, Russia
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Canada
| | - Alexander A Vassilevski
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Moscow Institute of Physics and Technology, State University, Dolgoprudny, Russia
| |
Collapse
|
3
|
Dinoi G, Conte E, Palumbo O, Benvenuto M, Coppola MA, Palumbo P, Lastella P, Boccanegra B, Di Muro E, Castori M, Carella M, Sciruicchio V, de Tommaso M, Liantonio A, De Luca A, La Neve A, Imbrici P. The Biallelic Inheritance of Two Novel SCN1A Variants Results in Developmental and Epileptic Encephalopathy Responsive to Levetiracetam. Biomedicines 2024; 12:1698. [PMID: 39200163 PMCID: PMC11351414 DOI: 10.3390/biomedicines12081698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 07/23/2024] [Accepted: 07/25/2024] [Indexed: 09/02/2024] Open
Abstract
Loss-, gain-of-function and mixed variants in SCN1A (Nav1.1 voltage-gated sodium channel) have been associated with a spectrum of neurologic disorders with different severity and drug-responsiveness. Most SCN1A variants are heterozygous changes occurring de novo or dominantly inherited; recessive inheritance has been reported in a few cases. Here, we report a family in which the biallelic inheritance of two novel SCN1A variants, N935Y and H1393Q, occurs in two siblings presenting with drug-responsive developmental and epileptic encephalopathy and born to heterozygous asymptomatic parents. To assess the genotype-phenotype correlation and support the treatment choice, HEK 293 cells were transfected with different combinations of the SCN1A WT and mutant cDNAs, and the resulting sodium currents were recorded through whole-cell patch-clamp. Functional studies showed that the N935Y and H1393Q channels and their combinations with the WT (WT + N935Y and WT + H1393Q) had current densities and biophysical properties comparable with those of their respective control conditions. This explains the asymptomatic condition of the probands' parents. The co-expression of the N935Y + H1393Q channels, mimicking the recessive inheritance of the two variants in siblings, showed ~20% reduced current amplitude compared with WT and with parental channels. This mild loss of Nav1.1 function may contribute in part to the disease pathogenesis, although other mechanisms may be involved.
Collapse
Affiliation(s)
- Giorgia Dinoi
- Department of Pharmacy—Drug Sciences, University of Bari “Aldo Moro”, 70125 Bari, Italy; (G.D.); (E.C.); (M.A.C.); (B.B.); (A.L.); (A.D.L.)
| | - Elena Conte
- Department of Pharmacy—Drug Sciences, University of Bari “Aldo Moro”, 70125 Bari, Italy; (G.D.); (E.C.); (M.A.C.); (B.B.); (A.L.); (A.D.L.)
| | - Orazio Palumbo
- Division of Medical Genetics, Fondazione IRCCS-Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy; (O.P.); (M.B.); (P.P.); (E.D.M.); (M.C.); (M.C.)
| | - Mario Benvenuto
- Division of Medical Genetics, Fondazione IRCCS-Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy; (O.P.); (M.B.); (P.P.); (E.D.M.); (M.C.); (M.C.)
| | - Maria Antonietta Coppola
- Department of Pharmacy—Drug Sciences, University of Bari “Aldo Moro”, 70125 Bari, Italy; (G.D.); (E.C.); (M.A.C.); (B.B.); (A.L.); (A.D.L.)
| | - Pietro Palumbo
- Division of Medical Genetics, Fondazione IRCCS-Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy; (O.P.); (M.B.); (P.P.); (E.D.M.); (M.C.); (M.C.)
| | - Patrizia Lastella
- Centro Sovraziendale Malattie Rare, UOC Medicina Interna Universitaria “C. Frugoni”, AOU Policlinico Consorziale di Bari, 70124 Bari, Italy;
| | - Brigida Boccanegra
- Department of Pharmacy—Drug Sciences, University of Bari “Aldo Moro”, 70125 Bari, Italy; (G.D.); (E.C.); (M.A.C.); (B.B.); (A.L.); (A.D.L.)
| | - Ester Di Muro
- Division of Medical Genetics, Fondazione IRCCS-Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy; (O.P.); (M.B.); (P.P.); (E.D.M.); (M.C.); (M.C.)
| | - Marco Castori
- Division of Medical Genetics, Fondazione IRCCS-Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy; (O.P.); (M.B.); (P.P.); (E.D.M.); (M.C.); (M.C.)
| | - Massimo Carella
- Division of Medical Genetics, Fondazione IRCCS-Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy; (O.P.); (M.B.); (P.P.); (E.D.M.); (M.C.); (M.C.)
| | - Vittorio Sciruicchio
- Children Epilepsy and EEG Center, Ospedale San Paolo di Bari, 70123 Bari, Italy;
| | - Marina de Tommaso
- DiBraiN Department, University of Bari “Aldo Moro”, 70124 Bari, Italy;
| | - Antonella Liantonio
- Department of Pharmacy—Drug Sciences, University of Bari “Aldo Moro”, 70125 Bari, Italy; (G.D.); (E.C.); (M.A.C.); (B.B.); (A.L.); (A.D.L.)
| | - Annamaria De Luca
- Department of Pharmacy—Drug Sciences, University of Bari “Aldo Moro”, 70125 Bari, Italy; (G.D.); (E.C.); (M.A.C.); (B.B.); (A.L.); (A.D.L.)
| | - Angela La Neve
- DiBraiN Department, University of Bari “Aldo Moro”, 70124 Bari, Italy;
| | - Paola Imbrici
- Department of Pharmacy—Drug Sciences, University of Bari “Aldo Moro”, 70125 Bari, Italy; (G.D.); (E.C.); (M.A.C.); (B.B.); (A.L.); (A.D.L.)
| |
Collapse
|
4
|
Zou X, Zhang Z, Lu H, Zhao W, Pan L, Chen Y. Functional effects of drugs and toxins interacting with Na V1.4. Front Pharmacol 2024; 15:1378315. [PMID: 38725668 PMCID: PMC11079311 DOI: 10.3389/fphar.2024.1378315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 04/08/2024] [Indexed: 05/12/2024] Open
Abstract
NaV1.4 is a voltage-gated sodium channel subtype that is predominantly expressed in skeletal muscle cells. It is essential for producing action potentials and stimulating muscle contraction, and mutations in NaV1.4 can cause various muscle disorders. The discovery of the cryo-EM structure of NaV1.4 in complex with β1 has opened new possibilities for designing drugs and toxins that target NaV1.4. In this review, we summarize the current understanding of channelopathies, the binding sites and functions of chemicals including medicine and toxins that interact with NaV1.4. These substances could be considered novel candidate compounds or tools to develop more potent and selective drugs targeting NaV1.4. Therefore, studying NaV1.4 pharmacology is both theoretically and practically meaningful.
Collapse
Affiliation(s)
- Xinyi Zou
- Zhejiang Provincial Key Laboratory of Resources Protection and Innovation of Traditional Chinese Medicine, College of Food and Health, Zhejiang Agriculture and Forestry University, Hangzhou, China
| | - Zixuan Zhang
- Zhejiang Provincial Key Laboratory of Resources Protection and Innovation of Traditional Chinese Medicine, College of Food and Health, Zhejiang Agriculture and Forestry University, Hangzhou, China
| | - Hui Lu
- Zhejiang Provincial Key Laboratory of Resources Protection and Innovation of Traditional Chinese Medicine, College of Food and Health, Zhejiang Agriculture and Forestry University, Hangzhou, China
| | - Wei Zhao
- Zhejiang Provincial Key Laboratory of Resources Protection and Innovation of Traditional Chinese Medicine, College of Food and Health, Zhejiang Agriculture and Forestry University, Hangzhou, China
| | - Lanying Pan
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Yuan Chen
- Zhejiang Provincial Key Laboratory of Resources Protection and Innovation of Traditional Chinese Medicine, College of Food and Health, Zhejiang Agriculture and Forestry University, Hangzhou, China
| |
Collapse
|
5
|
Lai HJ, Lee MJ, Yu HW, Chen KW, Tsai KL, Lin PC, Huang CW. Biophysical mechanisms underlying tefluthrin-induced modulation of gating changes and resurgent current generation in the human Na v1.4 channel. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2024; 200:105833. [PMID: 38582596 DOI: 10.1016/j.pestbp.2024.105833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 02/14/2024] [Accepted: 02/17/2024] [Indexed: 04/08/2024]
Abstract
Human skeletal muscle contraction is triggered by activation of Nav1.4 channels. Nav1.4 channels can generate resurgent currents by channel reopening at hyperpolarized potentials through a gating transition dependent on the intracellular Navβ4 peptide in the physiological conditions. Tefluthrin (TEF) is a pyrethroid insecticide that can disrupt electrical signaling in nerves and skeletal muscle, resulting in seizures, muscle spasms, fasciculations, and mental confusion. TEF can also induce tail currents through other voltage-gated sodium channels in the absence of Navβ4 peptide, suggesting that muscle spasms may be caused by resurgent currents. Further, intracellular Navβ4 peptide and extracellular TEF may show competitive or synergistic effects; however, their binding sites are still unknown. To address these issues, electrophysiological recordings were performed on CHO-K1 cells expressing Nav1.4 channels with intracellular Navβ4 peptide, extracellular TEF, or both. TEF and Navβ4 peptide induced a hyperpolarizing shift of activation and inactivation curves in the Nav1.4 channel. TEF also substantially prolonged the inactivation time constants, while simultaneous application of Navβ4 peptide partially reversed this effect. Resurgent currents were enhanced by TEF and Navβ4 peptide at negative potentials, but TEF more potently enhances resurgent currents and dampens decay of resurgent currents. With longer depolarization, peak resurgent currents decay was fastest with the TEF alone. Molecular docking suggested that TEF and Navβ4 peptide binding site(s) are not in the narrowest part of the channel pore, but rather in the bundle-crossing regions and in the domain linkers, respectively. TEF can induce resurgent currents independently and synergistically with Navβ4 peptide, which may explain the muscle spasms observed in TEF intoxication.
Collapse
Affiliation(s)
- Hsing-Jung Lai
- Department of Neurology, National Taiwan University Hospital, Taipei 10617, Taiwan; Department of Physiology, National Taiwan University, Taipei 10617, Taiwan
| | - Ming-Jen Lee
- Department of Neurology, National Taiwan University Hospital, Taipei 10617, Taiwan; Department of Medical genetics, National Taiwan University Hospital, Taipei 10617, Taiwan
| | - Hsin-Wei Yu
- Department of Physiology, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Kuan-Wen Chen
- Genetics Generation Advancement Corporation, Taipei 11494, Taiwan
| | - Ke-Li Tsai
- Department of Physiology, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Pi-Chen Lin
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
| | - Chiung-Wei Huang
- Department of Physiology, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Department of Post-Baccalaureate Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
| |
Collapse
|
6
|
Campanale C, Laghetti P, Saltarella I, Altamura C, Canioni E, Iosa E, Maggi L, Brugnoni R, Tacconi P, Desaphy JF. A c.1775C > T Point Mutation of Sodium Channel Alfa Subunit Gene (SCN4A) in a Three-Generation Sardinian Family with Sodium Channel Myotonia. J Neuromuscul Dis 2024; 11:725-734. [PMID: 38427496 DOI: 10.3233/jnd-230134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2024]
Abstract
Background 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. Methods Next-generation sequencing including the CLCN1 and SCN4A genes was performed in patients with clinical neuromuscular disorders. Electromyography, Short Exercise Test, in vivo and in vitro electrophysiology, site-directed mutagenesis and heterologous expression were collected. Results A heterozygous point mutation (c.1775C > T, p.Thr592Ile) of muscle voltage-gated sodium channel α subunit gene (SCN4A) has been identified in five female patients over three generations, in a family with non-dystrophic myotonia. The muscle stiffness and myotonia involve mainly the face and hands, but also affect walking and running, appearing early after birth and presenting a clear cold sensitivity. Very hot temperatures, menstruation and pregnancy also exacerbate the symptoms; muscle pain and a warm-up phenomenon are variable features. Neither paralytic attacks nor post-exercise weakness has been reported. Muscle hypertrophy with cramp-like pain and increased stiffness developed during pregnancy. The symptoms were controlled with both mexiletine and acetazolamide. The Short Exercise Test after muscle cooling revealed two different patterns, with moderate absolute changes of compound muscle action potential amplitude. Conclusions The p.Thr592Ile mutation in the SCN4A gene identified in this Sardinian family was responsible of clinical phenotype of myotonia.
Collapse
Affiliation(s)
- Carmen Campanale
- Department of Precision and Regenerative Medicine, Section of Pharmacology, School of Medicine, University of Bari Aldo Moro, Bari, Italy
| | - Paola Laghetti
- Department of Precision and Regenerative Medicine, Section of Pharmacology, School of Medicine, University of Bari Aldo Moro, Bari, Italy
| | - Ilaria Saltarella
- Department of Precision and Regenerative Medicine, Section of Pharmacology, School of Medicine, University of Bari Aldo Moro, Bari, Italy
| | - Concetta Altamura
- Department of Precision and Regenerative Medicine, Section of Pharmacology, School of Medicine, University of Bari Aldo Moro, Bari, Italy
| | - Eleonora Canioni
- Neurology IV - Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Emanuele Iosa
- Neurology IV - Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Lorenzo Maggi
- Neurology IV - Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Raffaella Brugnoni
- Neurology IV - Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Paolo Tacconi
- Centro Regionale per la Sclerosi Multipla, Ospedale Binaghi, Cagliari, Italy
| | - Jean-François Desaphy
- Department of Precision and Regenerative Medicine, Section of Pharmacology, School of Medicine, University of Bari Aldo Moro, Bari, Italy
| |
Collapse
|
7
|
Vacchiano V, Brugnoni R, Campanale C, Imbrici P, Dinoi G, Canioni E, Laghetti P, Saltarella I, Altamura C, Maggi L, Liguori R, Donadio V, Desaphy JF. Coexistence of SCN4A and CLCN1 mutations in a family with atypical myotonic features: A clinical and functional study. Exp Neurol 2023; 362:114342. [PMID: 36720299 DOI: 10.1016/j.expneurol.2023.114342] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 01/12/2023] [Accepted: 01/26/2023] [Indexed: 01/30/2023]
Abstract
Non-dystrophic myotonias include several entities with possible clinical overlap, i.e. myotonia congenita caused by CLCN1 gene mutations, as well as paramyotonia congenita and sodium channel myotonia caused by SCN4A gene mutations. Herein, we describe the clinical features of five relatives affected by clinical and neurophysiological myotonia, with an aspecific and mixed phenotype. Next-generation sequencing identified the novel p.K1302R variant in SCN4A and the p.H838P variant in CLCN1. Segregation of the two mutations with the disease was confirmed by genotyping affected and non-affected family members. Patch-clamp experiments showed that sodium currents generated by p.K1302R and WT hNav1.4 were very similar. Mutant channel showed a small negative shift (5 mV) in the voltage-dependence of activation, which increased the likelihood of the channel to open at more negative voltages. The p.H838P mutation caused a reduction in chloride current density and a small voltage-dependence shift towards less negative potentials, in agreement with its position into the CBS2 domain of the C-terminus. Our results demonstrated that the mild functional alterations induced by p.K1302R and p.H838P in combination may be responsible for the mixed myotonic phenotypes. The K1302R mutant was sensitive to mexiletine and lamotrigine, suggesting that both drugs might be useful for the K1302R carriers.
Collapse
Affiliation(s)
- Veria Vacchiano
- IRCSS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy.
| | - Raffaella Brugnoni
- Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Italy
| | - Carmen Campanale
- Dept. of Precision and Regenerative Medicine, School of Medicine, University of Bari Aldo Moro, Bari, Italy
| | - Paola Imbrici
- Dept. of Pharmacy - Drug Sciences, University of Bari Aldo Moro, Bari, Italy
| | - Giorgia Dinoi
- Dept. of Pharmacy - Drug Sciences, University of Bari Aldo Moro, Bari, Italy
| | - Eleonora Canioni
- Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Italy
| | - Paola Laghetti
- Dept. of Precision and Regenerative Medicine, School of Medicine, University of Bari Aldo Moro, Bari, Italy
| | - Ilaria Saltarella
- Dept. of Precision and Regenerative Medicine, School of Medicine, University of Bari Aldo Moro, Bari, Italy
| | - Concetta Altamura
- Dept. of Precision and Regenerative Medicine, School of Medicine, University of Bari Aldo Moro, Bari, Italy
| | - Lorenzo Maggi
- Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Italy
| | - Rocco Liguori
- IRCSS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Vincenzo Donadio
- IRCSS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Jean-François Desaphy
- Dept. of Precision and Regenerative Medicine, School of Medicine, University of Bari Aldo Moro, Bari, Italy
| |
Collapse
|
8
|
Altamura C, Saltarella I, Campanale C, Laghetti P, Desaphy JF. Drug repurposing in skeletal muscle ion channelopathies. Curr Opin Pharmacol 2023; 68:102329. [PMID: 36512979 DOI: 10.1016/j.coph.2022.102329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 10/26/2022] [Accepted: 11/02/2022] [Indexed: 12/14/2022]
Abstract
Skeletal muscle ion channelopathies are rare genetic diseases mainly characterized by myotonia (muscle stiffness) or periodic paralysis (muscle weakness). Here, we reviewed the available therapeutic options in non-dystrophic myotonias (NDM) and periodic paralyses (PP), which consists essentially in drug repositioning to address stiffness or weakness attacks. Empirical use followed by successful randomized clinical trials eventually led to the orphan drug designation and marketing authorization granting of mexiletine for NDM and dichlorphenamide for PP. Yet, these treatments neither consider the genetic cause of the diseases nor address the individual variability in drug response. Thus, ongoing research aims at the identification of repurposed drugs alternative to mexiletine and dichlorphenamide to allow personalization of treatment. This review highlights how drug repurposing may represent an efficient strategy in rare diseases, allowing reduction of drug development time and costs in a context in which the return on investment may be particularly challenging.
Collapse
Affiliation(s)
- Concetta Altamura
- Section of Pharmacology, Department of Precision and Regenerative Medicine, School of Medicine, University of Bari Aldo Moro, Piazza Giulo Cesare, 70124, Bari, Italy
| | - Ilaria Saltarella
- Section of Pharmacology, Department of Precision and Regenerative Medicine, School of Medicine, University of Bari Aldo Moro, Piazza Giulo Cesare, 70124, Bari, Italy
| | - Carmen Campanale
- Section of Pharmacology, Department of Precision and Regenerative Medicine, School of Medicine, University of Bari Aldo Moro, Piazza Giulo Cesare, 70124, Bari, Italy
| | - Paola Laghetti
- Section of Pharmacology, Department of Precision and Regenerative Medicine, School of Medicine, University of Bari Aldo Moro, Piazza Giulo Cesare, 70124, Bari, Italy
| | - Jean-François Desaphy
- Section of Pharmacology, Department of Precision and Regenerative Medicine, School of Medicine, University of Bari Aldo Moro, Piazza Giulo Cesare, 70124, Bari, Italy.
| |
Collapse
|
9
|
De Bellis M, Boccanegra B, Cerchiara AG, Imbrici P, De Luca A. Blockers of Skeletal Muscle Na v1.4 Channels: From Therapy of Myotonic Syndrome to Molecular Determinants of Pharmacological Action and Back. Int J Mol Sci 2023; 24:ijms24010857. [PMID: 36614292 PMCID: PMC9821513 DOI: 10.3390/ijms24010857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 12/28/2022] [Accepted: 12/30/2022] [Indexed: 01/05/2023] Open
Abstract
The voltage-gated sodium channels represent an important target for drug discovery since a large number of physiological processes are regulated by these channels. In several excitability disorders, including epilepsy, cardiac arrhythmias, chronic pain, and non-dystrophic myotonia, blockers of voltage-gated sodium channels are clinically used. Myotonia is a skeletal muscle condition characterized by the over-excitability of the sarcolemma, resulting in delayed relaxation after contraction and muscle stiffness. The therapeutic management of this disorder relies on mexiletine and other sodium channel blockers, which are not selective for the Nav1.4 skeletal muscle sodium channel isoform. Hence, the importance of deepening the knowledge of molecular requirements for developing more potent and use-dependent drugs acting on Nav1.4. Here, we review the available treatment options for non-dystrophic myotonia and the structure-activity relationship studies performed in our laboratory with a focus on new compounds with potential antimyotonic activity.
Collapse
|
10
|
Ke Q, Zhao Y, Li Y, Ye J, Tang S, He F, Ji F, Dai X, Ni J, Li Y, Griggs RC, Cheng X. Clinical comparison and functional study of the L703P: a recurrent mutation in human SCN4A that causes sodium channel myotonia. Neuromuscul Disord 2022; 32:811-819. [PMID: 36050252 DOI: 10.1016/j.nmd.2022.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 08/10/2022] [Accepted: 08/16/2022] [Indexed: 12/01/2022]
Abstract
The non-dystrophic myotonias are inherited skeletal muscle disorders characterized by skeletal muscle stiffness after voluntary contraction, without muscle atrophy. Based on their clinical features, non-dystrophic myotonias are classified into myotonia congenita, paramyotonia congenita, and sodium channel myotonia. Using whole-exome next-generation sequencing, we identified a L703P mutation (c.2108T>C, p.L703P) in SCN4A in a Chinese family diagnosed with non-dystrophic myotonias. The clinical findings of patients in this family included muscle stiffness and hypertrophy. The biophysical properties of wildtype and mutant channels were investigated using whole-cell patch clamp. L703P causes both gain-of-function and loss-of-function changes in Nav1.4 properties, including decreased current density, impaired recovery, enhanced activation and slow inactivation. Our study demonstrates that L703P is a pathogenic variant for myotonia, and provides additional electrophysiological information for understanding the pathogenic mechanism of SCN4A-associated channelopathies.
Collapse
Affiliation(s)
- Qing Ke
- Department of Neurology, the First Affiliated Hospital, Zhejiang University School of Medicine. #79 Qingchun Road, Hangzhou, Zhejiang 310003, P.R. China.
| | - Youcheng Zhao
- Department of Anesthesiology, Zhejiang Provincial People's Hospital, P.R. China
| | - Yuezhou Li
- The Children's hospital, Zhejiang university school of medicine, P.R. China
| | - Jia Ye
- The Children's hospital, Zhejiang university school of medicine, P.R. China
| | - Siyang Tang
- The Children's hospital, Zhejiang university school of medicine, P.R. China
| | - Fangping He
- Department of Neurology, the First Affiliated Hospital, Zhejiang University School of Medicine. #79 Qingchun Road, Hangzhou, Zhejiang 310003, P.R. China
| | - Fang Ji
- Department of Neurology, the First Affiliated Hospital, Zhejiang University School of Medicine. #79 Qingchun Road, Hangzhou, Zhejiang 310003, P.R. China
| | - Xuejiao Dai
- Department of Neurology, the First Affiliated Hospital, Zhejiang University School of Medicine. #79 Qingchun Road, Hangzhou, Zhejiang 310003, P.R. China
| | - Jie Ni
- Department of Neurology, the First Affiliated Hospital, Zhejiang University School of Medicine. #79 Qingchun Road, Hangzhou, Zhejiang 310003, P.R. China
| | - Yi Li
- Department of Neurology, the First Affiliated Hospital, Zhejiang University School of Medicine. #79 Qingchun Road, Hangzhou, Zhejiang 310003, P.R. China
| | - Robert C Griggs
- Department of Neurology, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA.
| | - Xiaoyang Cheng
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut 06510, US; Center for Neuroscience & Regeneration Research, Yale University, New Haven, Connecticut 06510, US; Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, Connecticut 06516, US.
| |
Collapse
|
11
|
Kinetic Alterations in Resurgent Sodium Currents of Mutant Nav1.4 Channel in Two Patients Affected by Paramyotonia Congenita. BIOLOGY 2022; 11:biology11040613. [PMID: 35453812 PMCID: PMC9031228 DOI: 10.3390/biology11040613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 04/11/2022] [Accepted: 04/14/2022] [Indexed: 11/17/2022]
Abstract
Simple Summary Paramyotonia congenita (PMC) is a rare muscle disorder that causes myotonia and weakness of facial and limb muscles. The electromyography in PMC shows continual spontaneous, high-frequency spike potentials in skeletal muscles. Genetic mutations in the Nav1.4 channel that cause hyperexcitability of muscle fibers are responsible for PMC. However, the genotype–phenotype relationship is highly diversified, and the molecular pathology remains unclear. Here, we investigated the electrophysiology in the Nav1.4 channel with mutations, p.V781I and p.A1737T, which were found in two Taiwanese patients. We identified the distinct changes in gating mechanisms altered by mutations which may underlie the clinical phenotype. Abstract Paramyotonia congenita (PMC) is a rare skeletal muscle disorder characterized by muscle stiffness upon repetitive exercise and cold exposure. PMC was reported to be caused by dominant mutations in the SCN4A gene encoding the α subunit of the Nav1.4 channel. Recently, we identified two missense mutations of the SCN4A gene, p.V781I and p.A1737T, in two PMC families. To evaluate the changes in electrophysiological properties caused by the mutations, both mutant and wild-type (WT) SCN4A genes were expressed in CHO-K1 and HEK-293T cells. Then, whole-cell patch-clamp recording was employed to study the altered gating of mutant channels. The activation curve of transient current showed a hyperpolarizing shift in both mutant Nav1.4 channels as compared to the WT channel, whereas there was a depolarizing shift in the fast inactivation curve. These changes confer to an increase in window current in the mutant channels. Further investigations demonstrated that the mutated channel proteins generate significantly larger resurgent currents as compared to the WT channel and take longer to attain the peak of resurgent current than the WT channel. In conclusion, the current study demonstrates that p.V781I and p.A1737T mutations in the Nav1.4 channel increase both the sustained and the resurgent Na+ current, leading to membrane hyperexcitability with a lower firing threshold, which may influence the clinical phenotype.
Collapse
|
12
|
Altamura C, Conte D, Carratù MR, Desaphy JF. Target mutation-driven drug discovery. Curr Med Chem 2022; 29:5156-5158. [PMID: 35440295 DOI: 10.2174/1389450123666220418111200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 01/10/2022] [Accepted: 02/09/2022] [Indexed: 11/22/2022]
Affiliation(s)
- Concetta Altamura
- Department of Biomedical Sciences and Human Oncology, School of Medicine, University of Bari Aldo Moro, Bari, I-70124 Italy
| | | | - Maria Rosaria Carratù
- Department of Biomedical Sciences and Human Oncology, School of Medicine, University of Bari Aldo Moro, Bari, I-70124 Italy
| | - Jean-François Desaphy
- Department of Biomedical Sciences and Human Oncology, School of Medicine, University of Bari Aldo Moro, Bari, I-70124 Italy
| |
Collapse
|
13
|
Milani G, Cavalluzzi MM, Altamura C, Santoro A, Perrone M, Muraglia M, Colabufo NA, Corbo F, Casalino E, Franchini C, Pisano I, Desaphy J, Carrieri A, Carocci A, Lentini G. Bioisosteric Modification of To042: Synthesis and Evaluation of Promising Use-Dependent Inhibitors of Voltage-Gated Sodium Channels. ChemMedChem 2021; 16:3588-3599. [PMID: 34519427 PMCID: PMC9293070 DOI: 10.1002/cmdc.202100496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 09/01/2021] [Indexed: 11/07/2022]
Abstract
Three analogues of To042, a tocainide-related lead compound recently reported for the treatment of myotonia, were synthesized and evaluated in vitro as skeletal muscle sodium channel blockers possibly endowed with enhanced use-dependent behavior. Patch-clamp experiments on hNav1.4 expressed in HEK293 cells showed that N-[(naphthalen-1-yl)methyl]-4-[(2,6-dimethyl)phenoxy]butan-2-amine, the aryloxyalkyl bioisostere of To042, exerted a higher use-dependent block than To042 thus being able to preferentially block the channels in over-excited membranes while preserving healthy tissue function. It also showed the lowest active transport across BBB according to the results of P-glycoprotein (P-gp) interacting activity evaluation and the highest cytoprotective effect on HeLa cells. Quantum mechanical calculations and dockings gave insights on the most probable conformation of the aryloxyalkyl bioisostere of To042 in solution and the target residues involved in the binding, respectively. Both approaches indicated the conformations that might be adopted in both the unbound and bound state of the ligand. Overall, N-[(naphthalen-1-yl)methyl]-4-[(2,6-dimethyl)phenoxy]butan-2-amine exhibits an interesting toxico-pharmacological profile and deserves further investigation.
Collapse
Affiliation(s)
- Gualtiero Milani
- Department of Pharmacy – Pharmaceutical SciencesUniversity of Bari Aldo MoroVia E. Orabona 470125BariItaly
| | - Maria Maddalena Cavalluzzi
- Department of Pharmacy – Pharmaceutical SciencesUniversity of Bari Aldo MoroVia E. Orabona 470125BariItaly
| | - Concetta Altamura
- Department of Biomedical Sciences and Human OncologySchool of MedicineUniversity of Bari Aldo Moro PoliclinicoPiazza Giulio Cesare70124BariItaly
| | - Antonella Santoro
- Department of Bioscience, Biotechnology and BiopharmaceuticsUniversity of Bari Aldo MoroVia Orabona 470125BariItaly
| | - Mariagrazia Perrone
- Department of Pharmacy – Pharmaceutical SciencesUniversity of Bari Aldo MoroVia E. Orabona 470125BariItaly
| | - Marilena Muraglia
- Department of Pharmacy – Pharmaceutical SciencesUniversity of Bari Aldo MoroVia E. Orabona 470125BariItaly
| | - Nicola Antonio Colabufo
- Department of Pharmacy – Pharmaceutical SciencesUniversity of Bari Aldo MoroVia E. Orabona 470125BariItaly
| | - Filomena Corbo
- Department of Pharmacy – Pharmaceutical SciencesUniversity of Bari Aldo MoroVia E. Orabona 470125BariItaly
| | - Elisabetta Casalino
- Department of Veterinary MedicineUniversity of Bari Aldo MoroVia E. Orabona 470125BariItaly
| | - Carlo Franchini
- Department of Pharmacy – Pharmaceutical SciencesUniversity of Bari Aldo MoroVia E. Orabona 470125BariItaly
| | - Isabella Pisano
- Department of Bioscience, Biotechnology and BiopharmaceuticsUniversity of Bari Aldo MoroVia Orabona 470125BariItaly
| | - Jean‐François Desaphy
- Department of Biomedical Sciences and Human OncologySchool of MedicineUniversity of Bari Aldo Moro PoliclinicoPiazza Giulio Cesare70124BariItaly
| | - Antonio Carrieri
- Department of Pharmacy – Pharmaceutical SciencesUniversity of Bari Aldo MoroVia E. Orabona 470125BariItaly
| | - Alessia Carocci
- Department of Pharmacy – Pharmaceutical SciencesUniversity of Bari Aldo MoroVia E. Orabona 470125BariItaly
| | - Giovanni Lentini
- Department of Pharmacy – Pharmaceutical SciencesUniversity of Bari Aldo MoroVia E. Orabona 470125BariItaly
| |
Collapse
|
14
|
Functional cross-talk between phosphorylation and disease-causing mutations in the cardiac sodium channel Na v1.5. Proc Natl Acad Sci U S A 2021; 118:2025320118. [PMID: 34373326 PMCID: PMC8379932 DOI: 10.1073/pnas.2025320118] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
The cardiac sodium channel (Nav1.5) is crucial for generating a regular heartbeat. It is thus not surprising that Nav1.5 mutations have been linked to life-threatening arrhythmias. Interestingly, Nav1.5 activity can also be altered by posttranslational modifications, such as tyrosine phosphorylation. Our combination of protein engineering and molecular modeling has revealed that the detrimental effect of a long QT3 patient mutation is only exposed when a proximal tyrosine is phosphorylated. This suggests a dynamic cross-talk between the genetic mutation and a neighboring phosphorylation, a phenomenon that could be important in other classes of proteins. Additionally, we show that phosphorylation can affect the channel’s sensitivity toward clinically relevant drugs, a finding that may prove important when devising patient-specific treatment plans. The voltage-gated sodium channel Nav1.5 initiates the cardiac action potential. Alterations of its activation and inactivation properties due to mutations can cause severe, life-threatening arrhythmias. Yet despite intensive research efforts, many functional aspects of this cardiac channel remain poorly understood. For instance, Nav1.5 undergoes extensive posttranslational modification in vivo, but the functional significance of these modifications is largely unexplored, especially under pathological conditions. This is because most conventional approaches are unable to insert metabolically stable posttranslational modification mimics, thus preventing a precise elucidation of the contribution by these modifications to channel function. Here, we overcome this limitation by using protein semisynthesis of Nav1.5 in live cells and carry out complementary molecular dynamics simulations. We introduce metabolically stable phosphorylation mimics on both wild-type (WT) and two pathogenic long-QT mutant channel backgrounds and decipher functional and pharmacological effects with unique precision. We elucidate the mechanism by which phosphorylation of Y1495 impairs steady-state inactivation in WT Nav1.5. Surprisingly, we find that while the Q1476R patient mutation does not affect inactivation on its own, it enhances the impairment of steady-state inactivation caused by phosphorylation of Y1495 through enhanced unbinding of the inactivation particle. We also show that both phosphorylation and patient mutations can impact Nav1.5 sensitivity toward the clinically used antiarrhythmic drugs quinidine and ranolazine, but not flecainide. The data highlight that functional effects of Nav1.5 phosphorylation can be dramatically amplified by patient mutations. Our work is thus likely to have implications for the interpretation of mutational phenotypes and the design of future drug regimens.
Collapse
|
15
|
Taleb A, Zhou YP, Meng LT, Zhu MY, Zhang Q, Naveed M, Li LD, Wang P, Zhou QG, Meng F, Han F. New application of an old drug proparacaine in treating epilepsy via liposomal hydrogel formulation. Pharmacol Res 2021; 169:105636. [PMID: 33932606 DOI: 10.1016/j.phrs.2021.105636] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 04/16/2021] [Accepted: 04/20/2021] [Indexed: 01/06/2023]
Abstract
Proparacaine (PPC) is a previously discovered topical anesthetic for ophthalmic optometry and surgery by blocking the central Nav1.3. In this study, we found that proparacaine hydrochloride (PPC-HCl) exerted an acute robust antiepileptic effect in pilocarpine-induced epilepsy mice. More importantly, chronic treatment with PPC-HCl totally terminated spontaneous recurrent seizure occurrence without significant toxicity. Chronic treatment with PPC-HCl did not cause obvious cytotoxicity, neuropsychiatric adverse effects, hepatotoxicity, cardiotoxicity, and even genotoxicity that evaluated by whole genome-scale transcriptomic analyses. Only when in a high dose (50 mg/kg), the QRS interval measured by electrocardiography was slightly prolonged, which was similar to the impact of levetiracetam. Nevertheless, to overcome this potential issue, we adopt a liposome encapsulation strategy that could alleviate cardiotoxicity and prepared a type of hydrogel containing PPC-HCl for sustained release. Implantation of thermosensitive chitosan-based hydrogel containing liposomal PPC-HCl into the subcutaneous tissue exerted immediate and long-lasting remission from spontaneous recurrent seizure in epileptic mice without affecting QRS interval. Therefore, this new liposomal hydrogel formulation of proparacaine could be developed as a transdermal patch for treating epilepsy, avoiding the severe toxicity after chronic treatment with current antiepileptic drugs in clinic.
Collapse
Affiliation(s)
- Abdoh Taleb
- Key Lab of Cardiovascular and Cerebrovascular Medicine, Drug Target and Drug Discovery Center, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Ya-Ping Zhou
- Department of Clinical Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Ling-Tong Meng
- Key Lab of Cardiovascular and Cerebrovascular Medicine, Drug Target and Drug Discovery Center, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Ming-Yi Zhu
- Department of Clinical Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Qiao Zhang
- Department of Clinical Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Muhammad Naveed
- Key Lab of Cardiovascular and Cerebrovascular Medicine, Drug Target and Drug Discovery Center, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Lian-Di Li
- Department of Clinical Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Peng Wang
- State Key Laboratory of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Pharmaceutical Co., Ltd., Nanjing 210042, China
| | - Qi-Gang Zhou
- Key Lab of Cardiovascular and Cerebrovascular Medicine, Drug Target and Drug Discovery Center, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China; Department of Clinical Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China; Sir Run Run Hospital, Nanjing Medical University, Nanjing 211166, Jiangsu Province, China.
| | - Fan Meng
- Department of Clinical Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China; State Key Laboratory of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Pharmaceutical Co., Ltd., Nanjing 210042, China.
| | - Feng Han
- Key Lab of Cardiovascular and Cerebrovascular Medicine, Drug Target and Drug Discovery Center, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China; Department of Clinical Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China.
| |
Collapse
|
16
|
Balla C, Conte E, Selvatici R, Marsano RM, Gerbino A, Farnè M, Blunck R, Vitali F, Armaroli A, Brieda A, Liantonio A, De Luca A, Ferlini A, Rapezzi C, Bertini M, Gualandi F, Imbrici P. Functional Characterization of Two Novel Mutations in SCN5A Associated with Brugada Syndrome Identified in Italian Patients. Int J Mol Sci 2021; 22:ijms22126513. [PMID: 34204499 PMCID: PMC8234720 DOI: 10.3390/ijms22126513] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 06/11/2021] [Accepted: 06/14/2021] [Indexed: 12/19/2022] Open
Abstract
Background. Brugada syndrome (BrS) is an autosomal dominantly inherited cardiac disease characterized by “coved type” ST-segment elevation in the right precordial leads, high susceptibility to ventricular arrhythmia and a family history of sudden cardiac death. The SCN5A gene, encoding for the cardiac voltage-gated sodium channel Nav1.5, accounts for ~20–30% of BrS cases and is considered clinically relevant. Methods. Here, we describe the clinical findings of two Italian families affected by BrS and provide the functional characterization of two novel SCN5A mutations, the missense variant Pro1310Leu and the in-frame insertion Gly1687_Ile1688insGlyArg. Results. Despite being clinically different, both patients have a family history of sudden cardiac death and had history of arrhythmic events. The Pro1310Leu mutation significantly reduced peak sodium current density without affecting channel membrane localization. Changes in the gating properties of expressed Pro1310Leu channel likely account for the loss-of-function phenotype. On the other hand, Gly1687_Ile1688insGlyArg channel, identified in a female patient, yielded a nearly undetectable sodium current. Following mexiletine incubation, the Gly1687_Ile1688insGlyArg channel showed detectable, albeit very small, currents and biophysical properties similar to those of the Nav1.5 wild-type channel. Conclusions. Overall, our results suggest that the degree of loss-of-function shown by the two Nav1.5 mutant channels correlates with the aggressive clinical phenotype of the two probands. This genotype-phenotype correlation is fundamental to set out appropriate therapeutical intervention.
Collapse
Affiliation(s)
- Cristina Balla
- Cardiological Center, University of Ferrara, 44121 Ferrara, Italy; (C.B.); (F.V.); (A.B.); (C.R.); (M.B.)
| | - Elena Conte
- Department of Pharmacy-Drug Sciences, University of Bari “Aldo Moro”, 70125 Bari, Italy; (E.C.); (A.L.); (A.D.L.)
| | - Rita Selvatici
- Unit of Medical Genetics, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (R.S.); (M.F.); (A.A.); (A.F.)
| | | | - Andrea Gerbino
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari “Aldo Moro”, 70125 Bari, Italy;
| | - Marianna Farnè
- Unit of Medical Genetics, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (R.S.); (M.F.); (A.A.); (A.F.)
| | - Rikard Blunck
- Department of Physics, Université de Montréal, Montréal, QC H3C 3J7, Canada;
| | - Francesco Vitali
- Cardiological Center, University of Ferrara, 44121 Ferrara, Italy; (C.B.); (F.V.); (A.B.); (C.R.); (M.B.)
| | - Annarita Armaroli
- Unit of Medical Genetics, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (R.S.); (M.F.); (A.A.); (A.F.)
| | - Alessandro Brieda
- Cardiological Center, University of Ferrara, 44121 Ferrara, Italy; (C.B.); (F.V.); (A.B.); (C.R.); (M.B.)
| | - Antonella Liantonio
- Department of Pharmacy-Drug Sciences, University of Bari “Aldo Moro”, 70125 Bari, Italy; (E.C.); (A.L.); (A.D.L.)
| | - Annamaria De Luca
- Department of Pharmacy-Drug Sciences, University of Bari “Aldo Moro”, 70125 Bari, Italy; (E.C.); (A.L.); (A.D.L.)
| | - Alessandra Ferlini
- Unit of Medical Genetics, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (R.S.); (M.F.); (A.A.); (A.F.)
| | - Claudio Rapezzi
- Cardiological Center, University of Ferrara, 44121 Ferrara, Italy; (C.B.); (F.V.); (A.B.); (C.R.); (M.B.)
- Maria Cecilia Hospital, GVM Care & Research, 48033 Cotignola, Italy
| | - Matteo Bertini
- Cardiological Center, University of Ferrara, 44121 Ferrara, Italy; (C.B.); (F.V.); (A.B.); (C.R.); (M.B.)
| | - Francesca Gualandi
- Unit of Medical Genetics, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (R.S.); (M.F.); (A.A.); (A.F.)
- Correspondence: (F.G.); (P.I.)
| | - Paola Imbrici
- Department of Pharmacy-Drug Sciences, University of Bari “Aldo Moro”, 70125 Bari, Italy; (E.C.); (A.L.); (A.D.L.)
- Correspondence: (F.G.); (P.I.)
| |
Collapse
|
17
|
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: 30] [Impact Index Per Article: 7.5] [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
|
18
|
Changes in Resurgent Sodium Current Contribute to the Hyperexcitability of Muscles in Patients with Paramyotonia Congenita. Biomedicines 2021; 9:biomedicines9010051. [PMID: 33430134 PMCID: PMC7826575 DOI: 10.3390/biomedicines9010051] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 01/01/2021] [Accepted: 01/06/2021] [Indexed: 11/17/2022] Open
Abstract
Paramyotonia congenita (PMC) is a rare hereditary skeletal muscle disorder. The major symptom, muscle stiffness, is frequently induced by cold exposure and repetitive exercise. Mutations in human SCN4A gene, which encodes the α-subunit of Nav1.4 channel, are responsible for PMC. Mutation screening of SCN4A gene from two PMC families identified two missense mutations, p.T1313M and p.R1448H. To elucidate the electrophysiological abnormalities caused by the mutations, the p.T1313M, p.R1448H, and wild-type (WT) SCN4A genes were transient expressed on Chinese hamster ovary (CHO-K1) cells. The detailed study on the gating defects of the mutant channels using the whole-cell patch clamping technique was performed. The mutant Nav1.4 channels impaired the basic gating properties with increasing sustained and window currents during membrane depolarization and facilitated the genesis of resurgent currents during repolarization. The mutations caused a hyperpolarization shift in the fast inactivation and slightly enhanced the slow inactivation with an increase in half-maximal inactivation voltage. No differences were found in the decay kinetics of the tail current between mutant and WT channels. In addition to generating the larger resurgent sodium current, the time to peak in the mutant channels was longer than that in the WT channels. In conclusion, our results demonstrated that the mutations p.T1313M and p.R1448H in Nav1.4 channels can enhance fast inactivation, slow inactivation, and resurgent current, revealing that subtle changes in gating processes can influence the clinical phenotype.
Collapse
|
19
|
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: 5.3] [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
|
20
|
Non-dystrophic myotonias: clinical and mutation spectrum of 70 German patients. J Neurol 2020; 268:1708-1720. [PMID: 33263785 PMCID: PMC8068660 DOI: 10.1007/s00415-020-10328-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 11/09/2020] [Accepted: 11/19/2020] [Indexed: 12/17/2022]
Abstract
Introduction Non-dystrophic myotonias (NDM) are heterogeneous diseases caused by mutations in CLCN1 and SCN4A. The study aimed to describe the clinical and genetic spectrum of NDM in a large German cohort. Methods We retrospectively identified all patients with genetically confirmed NDM diagnosed in our center. The following data were analyzed: demographics, family history, muscular features, cardiac involvement, CK, EMG, genotype, other tested genes, treatment perceived efficacy. Results 70 patients (age 40.2 years ± 14.9; 52.8% males) were included in our study (48 NDM-CLCN1, 22 NDM-SCN4A). The most frequent presenting symptoms were myotonia (NDM-CLCN1 83.3%, NDM-SCN4A 72.2%) and myalgia (NDM-CLCN1 57.4%, NDM-SCN4A 52.6%). Besides a more prominent facial involvement in NDM-SCN4A and cold-sensitivity in NDM-CLCN1, no other significant differences were observed between groups. Cardiac arrhythmia or conduction defects were documented in sixNDM-CLCN1 patients (three of them requiring a pacemaker) and one patient with NDM-SCN4A. CK was normal in 40% of patients. Myotonic runs in EMG were detected in 89.1% of CLCN1 and 78.9% of SCN4A. 50% of NDM-CLCN1 patients had the classic c.2680C>T (p.Arg894*) mutation. 12 new genetic variants are reported. About 50% of patients were not taking any anti-myotonic drug at the last follow-up. The anti-myotonic drugs with the best patient’s perceived efficacy were mexiletine and lamotrigine. Conclusion This study highlights the relevant clinical overlap between NDM-CLCN1 and NDM-SCN4A patients and warrants the use of early and broad genetic investigation for the precise identification of the NDM subtype. Besides the clinical and genetic heterogeneity, the limited response to current anti-myotonic drugs constitutes a continuing challenge. Supplementary Information The online version contains supplementary material available at 10.1007/s00415-020-10328-1.
Collapse
|
21
|
Taminato T, Mori-Yoshimura M, Miki J, Sasaki R, Sato N, Oya Y, Nishino I, Takahashi Y. Paramyotonia Congenita with Persistent Distal and Facial Muscle Weakness: A Case Report with Literature Review. J Neuromuscul Dis 2020; 7:193-201. [PMID: 32083589 DOI: 10.3233/jnd-190440] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Paramyotonia congenita (PC; OMIM 168300) is a non-dystrophic myotonia caused by mutations in the SCN4A gene. Transient muscle stiffness, usually induced by exposure to cold and aggravated by exercise, is the predominant clinical symptom, and interictal persistent weakness is uncommon. CASE REPORT We report a family with a history of PC accompanied by persistent hand muscle weakness with masticatory muscle involvement. Persistent weakness was exacerbated with age, and MR analysis showed marked atrophy of temporal, masseter, and finger flexor muscles with fatty replacement. The PC causative mutation T1313M in the SCN4A gene was prevalent in the family. Administration of acetazolamide chloride improved clinical symptoms and the results of cold and short exercise tests. Phenotypic variation within the family was remarkable, as the two younger affected patients did not present with persistent weakness or muscle atrophy. CONCLUSIONS PC associated with the T1313M mutation is a possible cause of persistent distal hand weakness.
Collapse
Affiliation(s)
- Tomoya Taminato
- Department of Neurology, National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Madoka Mori-Yoshimura
- Department of Neurology, National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Jun Miki
- Department of Neurology, Saku Central Hospital, Nagano Prefectural Federation of Agricultural Cooperatives for Health and Welfare, Nagano, Japan
| | - Ryogen Sasaki
- Department of Neurology, Kuwana City Medical Center, Mie, Japan
| | - Noriko Sato
- Department of Radiology, National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Yasushi Oya
- Department of Neurology, National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Ichizo Nishino
- Department of Genome Medicine Development, Medical Genome Center, National Center of Neurology and Psychiatry, Tokyo, Japan.,Department of Neuromuscular Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Yuji Takahashi
- Department of Neurology, National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan
| |
Collapse
|
22
|
Abstract
PURPOSE OF REVIEW This article aims to review the current and upcoming treatment options of primary muscle channelopathies including the non-dystrophic myotonias and periodic paralyses. RECENT FINDINGS The efficacy of mexiletine in the treatment of myotonia is now supported by two randomised placebo-controlled trials, one of which utilised a novel aggregated n-of-1 design. This has resulted in licencing of the drug via orphan drug status. There is also good evidence that mexiletine is well tolerated and safe in this patient group without the need for intensive monitoring. A range of alternative antimyotonic treatment options include lamotrigine, carbamazepine and ranolazine exist with variable evidence base. In vitro studies have shown insight into reasons for treatment failure of some medications with certain genotypes opening the era of mutation-specific therapy such as use of flecainide. In the periodic paralyses, the ability of MRI to distinguish between reversible oedema and irreversible fatty replacement makes it an increasingly useful tool to guide and assess pharmacological treatment. Unfortunately, the striking efficacy of bumetanide in hypokalaemic periodic paralysis animal models was not replicated in a recent pilot study in humans. SUMMARY The treatment of skeletal muscle channelopathies combines dietary and lifestyle advice together with pharmacological interventions. The rarity of these conditions remains a barrier for clinical studies but the example of the aggregated n-of-1 trial of mexiletine shows that innovative trial design can overcome these hurdles. Further research is required to test efficacy of drugs shown to have promising characteristics in preclinical experiments such as safinamide, riluzule and magnesium for myotonia or bumetanide for hypokalaemic periodic paralysis.
Collapse
Affiliation(s)
- Nantaporn Jitpimolmard
- Queen Square Centre for Neuromuscular Diseases, UCL Queen Square Institute of Neurology, UCL, London, UK
- Rehabilitation Medicine Department, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Emma Matthews
- Queen Square Centre for Neuromuscular Diseases, UCL Queen Square Institute of Neurology, UCL, London, UK
- Atkinson-Morley Neuromuscular Centre, St George’s University Hospitals Foundation Trust, London, UK
| | - Doreen Fialho
- Queen Square Centre for Neuromuscular Diseases, UCL Queen Square Institute of Neurology, UCL, London, UK
| |
Collapse
|
23
|
Maggi L, Brugnoni R, Canioni E, Tonin P, Saletti V, Sola P, Piccinelli SC, Colleoni L, Ferrigno P, Pini A, Masson R, Manganelli F, Lietti D, Vercelli L, Ricci G, Bruno C, Tasca G, Pizzuti A, Padovani A, Fusco C, Pegoraro E, Ruggiero L, Ravaglia S, Siciliano G, Morandi L, Dubbioso R, Mongini T, Filosto M, Tramacere I, Mantegazza R, Bernasconi P. Clinical and Molecular Spectrum of Myotonia and Periodic Paralyses Associated With Mutations in SCN4A in a Large Cohort of Italian Patients. Front Neurol 2020; 11:646. [PMID: 32849172 PMCID: PMC7403394 DOI: 10.3389/fneur.2020.00646] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 05/29/2020] [Indexed: 11/25/2022] Open
Abstract
Background: Four main clinical phenotypes have been traditionally described in patients mutated in SCN4A, including sodium-channel myotonia (SCM), paramyotonia congenita (PMC), Hypokaliemic type II (HypoPP2), and Hyperkaliemic/Normokaliemic periodic paralysis (HyperPP/NormoPP); in addition, rare phenotypes associated with mutations in SCN4A are congenital myasthenic syndrome and congenital myopathy. However, only scarce data have been reported in literature on large patient cohorts including phenotypes characterized by myotonia and episodes of paralysis. Methods: We retrospectively investigated clinical and molecular features of 80 patients fulfilling the following criteria: (1) clinical and neurophysiological diagnosis of myotonia, or clinical diagnosis of PP, and (2) presence of a pathogenic SCN4A gene variant. Patients presenting at birth with episodic laryngospasm or congenital myopathy-like phenotype with later onset of myotonia were considered as neonatal SCN4A. Results: PMC was observed in 36 (45%) patients, SCM in 30 (37.5%), Hyper/NormoPP in 7 (8.7%), HypoPP2 in 3 (3.7%), and neonatal SCN4A in 4 (5%). The median age at onset was significantly earlier in PMC than in SCM (p < 0.01) and in Hyper/NormoPP than in HypoPP2 (p = 0.02). Cold-induced myotonia was more frequently observed in PMC (n = 34) than in SCM (n = 23) (p = 0.04). No significant difference was found in age at onset of episodes of paralysis among PMC and PP or in frequency of permanent weakness between PP (n = 4), SCM (n = 5), and PMC (n = 10). PP was more frequently associated with mutations in the S4 region of the NaV1.4 channel protein compared to SCM and PMC (p < 0.01); mutations causing PMC were concentrated in the C-terminal region of the protein, while SCM-associated mutations were detected in all the protein domains. Conclusions: Our data suggest that skeletal muscle channelopathies associated with mutations in SCN4A represent a continuum in the clinical spectrum.
Collapse
Affiliation(s)
- Lorenzo Maggi
- Neuroimmunology and Neuromuscular Diseases, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Raffaella Brugnoni
- Neuroimmunology and Neuromuscular Diseases, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Eleonora Canioni
- Neuroimmunology and Neuromuscular Diseases, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Paola Tonin
- Section of Clinical Neurology, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Veronica Saletti
- Developmental Neurology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Patrizia Sola
- Clinica Neurologica, Azienda Ospedaliero Universitaria di Modena, Modena, Italy
| | - Stefano Cotti Piccinelli
- Unit of Neurology, Center for Neuromuscular Diseases, ASST Spedali Civili and University of Brescia, Brescia, Italy
| | - Lara Colleoni
- Neuroimmunology and Neuromuscular Diseases, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Paola Ferrigno
- SC Neurologia e Stroke Unit, Azienda Ospedaliera Brotzu, Cagliari, Italy
| | - Antonella Pini
- Child Neurology and Psychiatry Unit, IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Riccardo Masson
- Developmental Neurology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Fiore Manganelli
- Department of Neurosciences, Reproductive Sciences and Odontostomatology, University of Naples "Federico II", Naples, Italy
| | | | - Liliana Vercelli
- Department of Neurosciences Rita Levi Montalcini, University of Turin, Turin, Italy
| | - Giulia Ricci
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Claudio Bruno
- Center of Translational and Experimental Myology, Istituto Giannina Gaslini, Genova, Italy
| | - Giorgio Tasca
- Unità Operativa Complessa di Neurologia, Dipartimento di Scienze Dell'Invecchiamento, Neurologiche, Ortopediche e della Testa-Collo, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Antonio Pizzuti
- Fondazione IRCCS Casa Sollievo della Sofferenza, Laboratory of Medical Genetics, San Giovanni Rotondo, Italy.,Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Alessandro Padovani
- Unit of Neurology, Center for Neuromuscular Diseases, ASST Spedali Civili and University of Brescia, Brescia, Italy
| | - Carlo Fusco
- Dipartimento Materno-Infantile, S.C. Neuropsichiatria Infantile, Presidio Ospedaliero Provinciale Santa Maria Nuova, IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Elena Pegoraro
- Department of Neurosciences, University of Padova, Padova, Italy
| | - Lucia Ruggiero
- Department of Neurosciences, Reproductive Sciences and Odontostomatology, University of Naples "Federico II", Naples, Italy
| | | | - Gabriele Siciliano
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Lucia Morandi
- Neuroimmunology and Neuromuscular Diseases, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Raffaele Dubbioso
- Department of Neurosciences, Reproductive Sciences and Odontostomatology, University of Naples "Federico II", Naples, Italy
| | - Tiziana Mongini
- Department of Neurosciences Rita Levi Montalcini, University of Turin, Turin, Italy
| | - Massimiliano Filosto
- Unit of Neurology, Center for Neuromuscular Diseases, ASST Spedali Civili and University of Brescia, Brescia, Italy
| | - Irene Tramacere
- Research and Clinical Development Department, Scientific Directorate, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Renato Mantegazza
- Neuroimmunology and Neuromuscular Diseases, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Pia Bernasconi
- Neuroimmunology and Neuromuscular Diseases, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| |
Collapse
|
24
|
Desaphy JF, Farinato A, Altamura C, De Bellis M, Imbrici P, Tarantino N, Caccia C, Melloni E, Padoani G, Vailati S, Keywood C, Carratù MR, De Luca A, Conte D, Pierno S. Safinamide's potential in treating nondystrophic myotonias: Inhibition of skeletal muscle voltage-gated sodium channels and skeletal muscle hyperexcitability in vitro and in vivo. Exp Neurol 2020; 328:113287. [PMID: 32205118 DOI: 10.1016/j.expneurol.2020.113287] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 03/14/2020] [Accepted: 03/19/2020] [Indexed: 12/26/2022]
Abstract
The antiarrhythmic sodium-channel blocker mexiletine is used to treat patients with myotonia. However, around 30% of patients do not benefit from mexiletine due to poor tolerability or suboptimal response. Safinamide is an add-on therapy to levodopa for Parkinson's disease. In addition to MAOB inhibition, safinamide inhibits neuronal sodium channels, conferring anticonvulsant activity in models of epilepsy. Here, we investigated the effects of safinamide on skeletal muscle hNav1.4 sodium channels and in models of myotonia, in-vitro and in-vivo. Using patch-clamp, we showed that safinamide reversibly inhibited sodium currents in HEK293T cells transfected with hNav1.4. At the holding potential (hp) of -120 mV, the half-maximum inhibitory concentrations (IC50) were 160 and 33 μM at stimulation frequencies of 0.1 and 10 Hz, respectively. The calculated affinity constants of safinamide were dependent on channel state: 420 μM for closed channels and 9 μM for fast-inactivated channels. The p.F1586C mutation in hNav1.4 greatly impaired safinamide inhibition, suggesting that the drug binds to the local anesthetic receptor site in the channel pore. In a condition mimicking myotonia, i.e. hp. of -90 mV and 50-Hz stimulation, safinamide inhibited INa with an IC50 of 6 μM, being two-fold more potent than mexiletine. Using the two-intracellular microelectrodes current-clamp method, action potential firing was recorded in vitro in rat skeletal muscle fibers in presence of the chloride channel blocker, 9-anthracene carboxylic acid (9-AC), to increase excitability. Safinamide counteracted muscle fiber hyperexcitability with an IC50 of 13 μM. In vivo, oral safinamide was tested in the rat model of myotonia. In this model, intraperitoneal injection of 9-AC greatly increased the time of righting reflex (TRR) due to development of muscle stiffness. Safinamide counteracted 9-AC induced TRR increase with an ED50 of 1.2 mg/kg, which is 7 times lower than that previously determined for mexiletine. In conclusion, safinamide is a potent voltage and frequency dependent blocker of skeletal muscle sodium channels. Accordingly, the drug was able to counteract abnormal muscle hyperexcitability induced by 9-AC, both in vitro and in vivo. Thus, this study suggests that safinamide may have potential in treating myotonia and warrants further preclinical and human studies to fully evaluate this possibility.
Collapse
Affiliation(s)
- Jean-François Desaphy
- Department of Biomedical Sciences and Human Oncology, School of Medicine, University of Bari Aldo Moro, Bari, Italy.
| | - Alessandro Farinato
- Department of Pharmacy & Drug Sciences, 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
| | - Michela De Bellis
- Department of Pharmacy & Drug Sciences, University of Bari Aldo Moro, Bari, Italy
| | - Paola Imbrici
- Department of Pharmacy & Drug Sciences, University of Bari Aldo Moro, Bari, Italy
| | - Nancy Tarantino
- Department of Pharmacy & Drug Sciences, University of Bari Aldo Moro, Bari, Italy
| | - Carla Caccia
- Open R&D Department, Zambon S.p.A., Bresso, MI, Italy
| | - Elsa Melloni
- Open R&D Department, Zambon S.p.A., Bresso, MI, Italy
| | | | | | | | - Maria Rosaria Carratù
- Department of Biomedical Sciences and Human Oncology, School of Medicine, University of Bari Aldo Moro, Bari, Italy
| | - Annamaria De Luca
- Department of Pharmacy & Drug Sciences, University of Bari Aldo Moro, Bari, Italy
| | - Diana Conte
- Department of Pharmacy & Drug Sciences, University of Bari Aldo Moro, Bari, Italy
| | - Sabata Pierno
- Department of Pharmacy & Drug Sciences, University of Bari Aldo Moro, Bari, Italy
| |
Collapse
|
25
|
Suetterlin KJ, Raja Rayan D, Matthews E, Hanna MG. Mexiletine (NaMuscla) for the treatment of myotonia in non-dystrophic myotonic disorders. Expert Opin Orphan Drugs 2020. [DOI: 10.1080/21678707.2020.1739519] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Affiliation(s)
- Karen J. Suetterlin
- MRC Centre for Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
| | - Dipa Raja Rayan
- MRC Centre for Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
| | - Emma Matthews
- MRC Centre for Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
| | - Michael G Hanna
- MRC Centre for Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
| |
Collapse
|
26
|
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: 2.2] [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
|