1
|
Jo S, Fujita A, Osorno T, Stewart RG, Vaelli PM, Bean BP. Differential state-dependent Nav1.8 inhibition by suzetrigine, LTGO-33, and A-887826. J Gen Physiol 2025; 157:e202413719. [PMID: 40136042 PMCID: PMC11938940 DOI: 10.1085/jgp.202413719] [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: 10/29/2024] [Revised: 02/18/2025] [Accepted: 03/09/2025] [Indexed: 03/27/2025] Open
Abstract
Nav1.8 sodium channels are expressed in pain-sensing neurons, and some Nav1.8 inhibitors significantly reduce pain in clinical trials. Several Nav1.8 inhibitors have an unusual state dependence whereby inhibition is relieved by depolarization. We compared the state-dependent action of several Nav1.8 channel inhibitors to test whether inhibition is relieved during action potential (AP) firing under physiological conditions to produce "reverse use dependence." A-887826 inhibition was substantially relieved by AP waveforms applied at 20 Hz at 37°C. In contrast, there was no relief during AP trains with suzetrigine (VX-548) or LTGO-33, even though inhibition could be effectively removed by long, strong depolarizations. These differences were explained by differences in the voltage dependence and kinetics with which the compounds dissociate from depolarized channels and rebind to resting state channels. Suzetrigine required the strongest depolarizations for relief (midpoint +33 mV) and relief was slow (tau >300 ms at +20 mV), so almost no relief occurred during an AP waveform. Relief from A-887826 required weaker depolarizations (midpoint +13 mV) and was much faster, so some relief occurred during each AP waveform and accumulated during 20-Hz trains. LTGO-33 required the weakest depolarizations for relief (midpoint -11 mV) and relief was even faster than for A-887826, but reinhibition between AP waveforms was far faster than for A-887826, so that relief did not accumulate during AP trains at 20 Hz. The results show that, unlike A-887826, there is no use-dependent relief of inhibition by suzetrigine or LTGO-33 with physiological voltage waveforms at physiological temperatures, but each for different reasons.
Collapse
Affiliation(s)
- Sooyeon Jo
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Akie Fujita
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Tomás Osorno
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | | | - Patric M. Vaelli
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Bruce P. Bean
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
2
|
Xie YF. Nav1.8 and Chronic Pain: From Laboratory Animals to Clinical Patients. Biomolecules 2025; 15:694. [PMID: 40427587 PMCID: PMC12108746 DOI: 10.3390/biom15050694] [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: 04/05/2025] [Revised: 05/07/2025] [Accepted: 05/08/2025] [Indexed: 05/29/2025] Open
Abstract
As a subtype of voltage-gated sodium channel and predominantly expressed in the sensory neurons located in the dorsal root ganglion (DRG), the Nav1.8 channel encoded by the SCN10A gene is found to have different variants in patients suffering chronic pain or insensitivity to pain due to the gain-of-function or loss-of-function of Nav1.8 channels. In animal models of chronic pain, Nav1.8 is also verified to be involved, suggesting that Nav1.8 may be a potential target for treatment of chronic pain. Another voltage-gated sodium channel, Nav1.7, is also proposed to be a target for chronic pain, supported by clinical findings in patients and laboratory animal models; however, there is no Nav1.7-specific drug that has passed clinical trials, although they demonstrated satisfactory effects in laboratory animals. This discrepancy between clinical and preclinical studies may be related to the differences between humans and laboratory animals or due to the degeneracy in different sodium channels governing the DRG neuronal excitability, which is thought of as the underlying machinery of chronic pain and mostly studied. This review summarizes recent findings of Nav1.8 in chronic pain from clinics and laboratories and discusses the difference, which may be helpful for future investigation of Nav1.8 in chronic pain, considering the dilemma of the Nav1.7 channel in chronic pain.
Collapse
Affiliation(s)
- Yu-Feng Xie
- Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| |
Collapse
|
3
|
Alsaloum M, Dib-Hajj SD, Page DA, Ruben PC, Krainer AR, Waxman SG. Voltage-gated sodium channels in excitable cells as drug targets. Nat Rev Drug Discov 2025; 24:358-378. [PMID: 39901031 DOI: 10.1038/s41573-024-01108-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/21/2024] [Indexed: 02/05/2025]
Abstract
Excitable cells - including neurons, muscle cells and cardiac myocytes - are unique in expressing high densities of voltage-gated sodium (NaV) channels. This molecular adaptation enables these cells to produce action potentials, and is essential to their function. With the advent of the molecular revolution, the concept of 'the' sodium channel has been supplanted by understanding that excitable cells in mammals can express any of nine different forms of sodium channels (NaV1.1-NaV1.9). Selective expression in particular types of cells, together with a key role in controlling action potential firing, makes some of these NaV subtypes especially attractive molecular targets for drug development. Although these different channel subtypes display a common overall structure, differences in their amino acid sequences have provided a basis for the development of subtype-specific drugs. This approach has resulted in exciting progress in the development of drugs for epilepsy, cardiac disorders and pain. In this Review, we discuss recent progress in the development of drugs that selectively target each of the sodium channel subtypes.
Collapse
Affiliation(s)
- Matthew Alsaloum
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | | | - Dana A Page
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Peter C Ruben
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, British Columbia, Canada
| | | | - Stephen G Waxman
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
4
|
Abbott GW, Manville RW. Na Vigating nociceptive neuron signaling pathways to arrive at pain relief. J Pharmacol Exp Ther 2025; 392:100037. [PMID: 40023591 DOI: 10.1016/j.jpet.2024.100037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Accepted: 08/16/2024] [Indexed: 03/04/2025] Open
Affiliation(s)
- Geoffrey W Abbott
- Bioelectricity Laboratory, Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, California.
| | - Rían W Manville
- Bioelectricity Laboratory, Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, California
| |
Collapse
|
5
|
Müller P, Draguhn A, Egorov AV. Persistent sodium currents in neurons: potential mechanisms and pharmacological blockers. Pflugers Arch 2024; 476:1445-1473. [PMID: 38967655 PMCID: PMC11381486 DOI: 10.1007/s00424-024-02980-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 06/07/2024] [Accepted: 06/11/2024] [Indexed: 07/06/2024]
Abstract
Persistent sodium current (INaP) is an important activity-dependent regulator of neuronal excitability. It is involved in a variety of physiological and pathological processes, including pacemaking, prolongation of sensory potentials, neuronal injury, chronic pain and diseases such as epilepsy and amyotrophic lateral sclerosis. Despite its importance, neither the molecular basis nor the regulation of INaP are sufficiently understood. Of particular significance is a solid knowledge and widely accepted consensus about pharmacological tools for analysing the function of INaP and for developing new therapeutic strategies. However, the literature on INaP is heterogeneous, with varying definitions and methodologies used across studies. To address these issues, we provide a systematic review of the current state of knowledge on INaP, with focus on mechanisms and effects of this current in the central nervous system. We provide an overview of the specificity and efficacy of the most widely used INaP blockers: amiodarone, cannabidiol, carbamazepine, cenobamate, eslicarbazepine, ethosuximide, gabapentin, GS967, lacosamide, lamotrigine, lidocaine, NBI-921352, oxcarbazepine, phenytoine, PRAX-562, propofol, ranolazine, riluzole, rufinamide, topiramate, valproaic acid and zonisamide. We conclude that there is strong variance in the pharmacological effects of these drugs, and in the available information. At present, GS967 and riluzole can be regarded bona fide INaP blockers, while phenytoin and lacosamide are blockers that only act on the slowly inactivating component of sodium currents.
Collapse
Affiliation(s)
- Peter Müller
- Department Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tuebingen , Hoppe-Seyler-Straße 3, 72076, Tübingen, Germany.
| | - Andreas Draguhn
- Institute for Physiology and Pathophysiology, Medical Faculty, Heidelberg University, Im Neuenheimer Feld 326, 69120, Heidelberg, Germany
| | - Alexei V Egorov
- Institute for Physiology and Pathophysiology, Medical Faculty, Heidelberg University, Im Neuenheimer Feld 326, 69120, Heidelberg, Germany
| |
Collapse
|
6
|
Liedtke W. [Additional experience with medicinal treatment of trigeminal nerve pain]. Schmerz 2024:10.1007/s00482-024-00820-2. [PMID: 39141098 DOI: 10.1007/s00482-024-00820-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/14/2024] [Indexed: 08/15/2024]
Affiliation(s)
- Wolfgang Liedtke
- US Facial Pain Association, 7778 McGinnis Ferry Road, 30024, Suwanee, GA, USA.
- Department of Neurology, Anesthesiology and Neurobiology, Duke University, Durham, NC, USA.
- Department of Molecular Pathobiology - Dental Pain Research, New York University, College of Dentistry, New York, NY, USA.
- New York University, Pain Research Center, New York, NY, USA.
| |
Collapse
|
7
|
Sánchez JD, Gómez-Carpintero J, González JF, Menéndez JC. Twenty-first century antiepileptic drugs. An overview of their targets and synthetic approaches. Eur J Med Chem 2024; 272:116476. [PMID: 38759456 DOI: 10.1016/j.ejmech.2024.116476] [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: 02/21/2024] [Revised: 05/01/2024] [Accepted: 05/02/2024] [Indexed: 05/19/2024]
Abstract
The therapeutic use of the traditional drugs against epilepsy has been hindered by their toxicity and low selectivity. These limitations have stimulated the design and development of new generations of antiepileptic drugs. This review explores the molecular targets and synthesis of the antiepileptic drugs that have entered the market in the 21st century, with a focus on manufacturer synthesis.
Collapse
Affiliation(s)
- J Domingo Sánchez
- Unidad de Química Orgánica y Farmacéutica, Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense, 28040, Madrid, Spain
| | - Jorge Gómez-Carpintero
- Unidad de Química Orgánica y Farmacéutica, Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense, 28040, Madrid, Spain
| | - Juan F González
- Unidad de Química Orgánica y Farmacéutica, Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense, 28040, Madrid, Spain
| | - J Carlos Menéndez
- Unidad de Química Orgánica y Farmacéutica, Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense, 28040, Madrid, Spain.
| |
Collapse
|
8
|
Fogel EL, Easler JJ, Yuan Y, Yadav D, Conwell DL, Vege SS, Han SY, Park W, Patrick V, White FA. Safety, Tolerability, and Dose-Limiting Toxicity of Lacosamide in Patients With Painful Chronic Pancreatitis: Protocol for a Phase 1 Clinical Trial to Determine Safety and Identify Side Effects. JMIR Res Protoc 2024; 13:e50513. [PMID: 38451604 PMCID: PMC10958339 DOI: 10.2196/50513] [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: 07/03/2023] [Revised: 10/24/2023] [Accepted: 10/25/2023] [Indexed: 03/08/2024] Open
Abstract
BACKGROUND Chronic abdominal pain is the hallmark symptom of chronic pancreatitis (CP), with 50% to 80% of patients seeking medical attention for pain control. Although several management options are available, outcomes are often disappointing, and opioids remain a mainstay of therapy. Opioid-induced hyperalgesia is a phenomenon resulting in dose escalation, which may occur partly because of the effects of opioids on voltage-gated sodium channels associated with pain. Preclinical observations demonstrate that the combination of an opioid and the antiseizure drug lacosamide diminishes opioid-induced hyperalgesia and improves pain control. OBJECTIVE In this phase 1 trial, we aim to determine the safety, tolerability, and dose-limiting toxicity of adding lacosamide to opioids for the treatment of painful CP and assess the feasibility of performance of a pilot study of adding lacosamide to opioid therapy in patients with CP. As an exploratory aim, we will assess the efficacy of adding lacosamide to opioid therapy in patients with painful CP. METHODS Using the Bayesian optimal interval design, we will conduct a dose-escalation trial of adding lacosamide to opioid therapy in patients with painful CP enrolled in cohorts of size 3. The initial dose will be 50 mg taken orally twice a day, followed by incremental increases to a maximum dose of 400 mg/day, with lacosamide administered for 7 days at each dose level. Adverse events will be documented according to Common Terminology Criteria for Adverse Events (version 5.0). RESULTS As of December 2023, we have currently enrolled 6 participants. The minimum number of participants to be enrolled is 12 with a maximum of 24. We expect to publish the results by March 2025. CONCLUSIONS This trial will test the feasibility of the study design and provide reassurance regarding the tolerability and safety of opioids in treating painful CP. It is anticipated that lacosamide will prove to be safe and well tolerated, supporting a subsequent phase 2 trial assessing the efficacy of lacosamide+opioid therapy in patients with painful CP, and that lacosamide combined with opiates will lower the opioid dose necessary for pain relief and improve the safety profile of opioid use in treating painful CP. TRIAL REGISTRATION Clinicaltrials.gov NCT05603702; https://clinicaltrials.gov/study/NCT05603702. INTERNATIONAL REGISTERED REPORT IDENTIFIER (IRRID) PRR1-10.2196/50513.
Collapse
Affiliation(s)
- Evan L Fogel
- Department of Medicine, School of Medicine, Indiana University, Indianapolis, IN, United States
| | - Jeffrey J Easler
- Department of Medicine, School of Medicine, Indiana University, Indianapolis, IN, United States
| | - Ying Yuan
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Dhiraj Yadav
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Darwin L Conwell
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | | | - Samuel Y Han
- Department of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Walter Park
- Department of Medicine, Stanford University Medical Center, Stanford, CA, United States
| | - Vanessa Patrick
- Department of Medicine, School of Medicine, Indiana University, Indianapolis, IN, United States
| | - Fletcher A White
- Department of Anesthesia, School of Medicine, Indiana University, Indianapolis, IN, United States
| |
Collapse
|
9
|
da Silva MDV, Piva M, Martelossi-Cebinelli G, Stinglin Rosa Ribas M, Hoffmann Salles Bianchini B, K Heintz O, Casagrande R, Verri WA. Stem cells and pain. World J Stem Cells 2023; 15:1035-1062. [PMID: 38179216 PMCID: PMC10762525 DOI: 10.4252/wjsc.v15.i12.1035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 11/06/2023] [Accepted: 11/30/2023] [Indexed: 12/26/2023] Open
Abstract
Pain can be defined as an unpleasant sensory and emotional experience caused by either actual or potential tissue damage or even resemble that unpleasant experience. For years, science has sought to find treatment alternatives, with minimal side effects, to relieve pain. However, the currently available pharmacological options on the market show significant adverse events. Therefore, the search for a safer and highly efficient analgesic treatment has become a priority. Stem cells (SCs) are non-specialized cells with a high capacity for replication, self-renewal, and a wide range of differentiation possibilities. In this review, we provide evidence that the immune and neuromodulatory properties of SCs can be a valuable tool in the search for ideal treatment strategies for different types of pain. With the advantage of multiple administration routes and dosages, therapies based on SCs for pain relief have demonstrated meaningful results with few downsides. Nonetheless, there are still more questions than answers when it comes to the mechanisms and pathways of pain targeted by SCs. Thus, this is an evolving field that merits further investigation towards the development of SC-based analgesic therapies, and this review will approach all of these aspects.
Collapse
Affiliation(s)
- Matheus Deroco Veloso da Silva
- Department of Pathology, Laboratory of Pain, Inflammation, Neuropathy and Cancer, State University of Londrina, Londrina 86057-970, Paraná, Brazil
| | - Maiara Piva
- Department of Pathology, Laboratory of Pain, Inflammation, Neuropathy and Cancer, State University of Londrina, Londrina 86057-970, Paraná, Brazil
| | - Geovana Martelossi-Cebinelli
- Department of Pathology, Laboratory of Pain, Inflammation, Neuropathy and Cancer, State University of Londrina, Londrina 86057-970, Paraná, Brazil
| | - Mariana Stinglin Rosa Ribas
- Department of Pathology, Laboratory of Pain, Inflammation, Neuropathy and Cancer, State University of Londrina, Londrina 86057-970, Paraná, Brazil
| | - Beatriz Hoffmann Salles Bianchini
- Department of Pathology, Laboratory of Pain, Inflammation, Neuropathy and Cancer, State University of Londrina, Londrina 86057-970, Paraná, Brazil
| | - Olivia K Heintz
- Morningside Graduate School of Biomedical Sciences, University of Massachusetts Chan Medical School, Worcester, MA 01655, United States
| | - Rubia Casagrande
- Department of Pharmaceutical Sciences, Center of Health Science, State University of Londrina, Londrina 86038-440, Paraná, Brazil
| | - Waldiceu A Verri
- Department of Pathology, Laboratory of Pain, Inflammation, Neuropathy and Cancer, Center of Biological Sciences, State University of Londrina, Londrina 86057-970, Paraná, Brazil.
| |
Collapse
|
10
|
Lv S, He E, Luo J, Liu Y, Liang W, Xu S, Zhang K, Yang Y, Wang M, Song Y, Wu Y, Cai X. Using Human-Induced Pluripotent Stem Cell Derived Neurons on Microelectrode Arrays to Model Neurological Disease: A Review. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2301828. [PMID: 37863819 PMCID: PMC10667858 DOI: 10.1002/advs.202301828] [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] [Received: 03/22/2023] [Revised: 09/04/2023] [Indexed: 10/22/2023]
Abstract
In situ physiological signals of in vitro neural disease models are essential for studying pathogenesis and drug screening. Currently, an increasing number of in vitro neural disease models are established using human-induced pluripotent stem cell (hiPSC) derived neurons (hiPSC-DNs) to overcome interspecific gene expression differences. Microelectrode arrays (MEAs) can be readily interfaced with two-dimensional (2D), and more recently, three-dimensional (3D) neural stem cell-derived in vitro models of the human brain to monitor their physiological activity in real time. Therefore, MEAs are emerging and useful tools to model neurological disorders and disease in vitro using human iPSCs. This is enabling a real-time window into neuronal signaling at the network scale from patient derived. This paper provides a comprehensive review of MEA's role in analyzing neural disease models established by hiPSC-DNs. It covers the significance of MEA fabrication, surface structure and modification schemes for hiPSC-DNs culturing and signal detection. Additionally, this review discusses advances in the development and use of MEA technology to study in vitro neural disease models, including epilepsy, autism spectrum developmental disorder (ASD), and others established using hiPSC-DNs. The paper also highlights the application of MEAs combined with hiPSC-DNs in detecting in vitro neurotoxic substances. Finally, the future development and outlook of multifunctional and integrated devices for in vitro medical diagnostics and treatment are discussed.
Collapse
Affiliation(s)
- Shiya Lv
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Enhui He
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
- The State Key Lab of Brain‐Machine IntelligenceZhejiang UniversityHangzhou321100China
| | - Jinping Luo
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Yaoyao Liu
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Wei Liang
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Shihong Xu
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Kui Zhang
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Yan Yang
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Mixia Wang
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Yilin Song
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Yirong Wu
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Xinxia Cai
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| |
Collapse
|
11
|
He Z, Li J. The therapeutic effects of lacosamide on epilepsy-associated comorbidities. Front Neurol 2023; 14:1063703. [PMID: 37006477 PMCID: PMC10062524 DOI: 10.3389/fneur.2023.1063703] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 03/02/2023] [Indexed: 03/18/2023] Open
Abstract
Epilepsy is a chronic neurological disorder associated with severe social and psychological effects, and most epilepsy patients often report at least one comorbidity. Accumulating evidence have suggested that lacosamide, a new generation of anti-seizure medications, may exhibit efficacy in the management of both epilepsy and its related comorbidities. Therefore, this narrative review aimed to elucidate the recent advancements regarding the therapeutic role of lacosamide in epilepsy-associated comorbidities. The possible pathophysiological mechanisms between epilepsy and epilepsy-associated comorbidities have been also partially described. Whether lacosamide improves cognitive and behavioral functions in patients with epilepsy has not been conclusively established. Some studies support that lacosamide may alleviate anxiety and depression in epilepsy patients. In addition, lacosamide has been found to be safe and effective in the treatment of epilepsy in people with intellectual disabilities, epilepsy of cerebrovascular etiology, and epilepsy associated with brain tumors. Moreover, lacosamide treatment has demonstrated fewer side effects on other systems. Hence, future larger and higher quality clinical studies are needed to further explore both the safety and efficacy of lacosamide in the treatment of epilepsy-associated comorbidities.
Collapse
|
12
|
Castro PA, Pinto-Borguero I, Yévenes GE, Moraga-Cid G, Fuentealba J. Antiseizure medication in early nervous system development. Ion channels and synaptic proteins as principal targets. Front Pharmacol 2022; 13:948412. [PMID: 36313347 PMCID: PMC9614143 DOI: 10.3389/fphar.2022.948412] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 09/05/2022] [Indexed: 12/04/2022] Open
Abstract
The main strategy for the treatment of epilepsy is the use of pharmacological agents known as antiseizure medication (ASM). These drugs control the seizure onset and improves the life expectancy and quality of life of patients. Several ASMs are contraindicated during pregnancy, due to a potential teratogen risk. For this reason, the pharmacological treatments of the pregnant Women with Epilepsy (WWE) need comprehensive analyses to reduce fetal risk during the first trimester of pregnancy. The mechanisms by which ASM are teratogens are still under study and scientists in the field, propose different hypotheses. One of them, which will be addressed in this review, corresponds to the potential alteration of ASM on ion channels and proteins involved in relevant signaling and cellular responses (i.e., migration, differentiation) during embryonic development. The actual information related to the action of ASM and its possible targets it is poorly understood. In this review, we will focus on describing the eventual presence of some ion channels and synaptic proteins of the neurotransmitter signaling pathways present during early neural development, which could potentially interacting as targets of ASM. This information leads to elucidate whether these drugs would have the ability to affect critical signaling during periods of neural development that in turn could explain the fetal malformations observed by the use of ASM during pregnancy.
Collapse
Affiliation(s)
- Patricio A. Castro
- Laboratory of Physiology and Pharmacology for Neural Development, LAND, Departamento de Fisiología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
- *Correspondence: Patricio A. Castro,
| | - Ingrid Pinto-Borguero
- Laboratory of Physiology and Pharmacology for Neural Development, LAND, Departamento de Fisiología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Gonzalo E. Yévenes
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Gustavo Moraga-Cid
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Jorge Fuentealba
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| |
Collapse
|
13
|
Kitano Y, Shinozuka T. Inhibition of Na V1.7: the possibility of ideal analgesics. RSC Med Chem 2022; 13:895-920. [PMID: 36092147 PMCID: PMC9384491 DOI: 10.1039/d2md00081d] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 07/25/2022] [Indexed: 08/03/2023] Open
Abstract
The selective inhibition of NaV1.7 is a promising strategy for developing novel analgesic agents with fewer adverse effects. Although the potent selective inhibition of NaV1.7 has been recently achieved, multiple NaV1.7 inhibitors failed in clinical development. In this review, the relationship between preclinical in vivo efficacy and NaV1.7 coverage among three types of voltage-gated sodium channel (VGSC) inhibitors, namely conventional VGSC inhibitors, sulphonamides and acyl sulphonamides, is discussed. By demonstrating the PK/PD discrepancy of preclinical studies versus in vivo models and clinical results, the potential reasons behind the disconnect between preclinical results and clinical outcomes are discussed together with strategies for developing ideal analgesic agents.
Collapse
Affiliation(s)
- Yutaka Kitano
- R&D Division, Daiichi Sankyo Co., Ltd. 1-2-58 Hiromachi Shinagawa-ku Tokyo 140-8710 Japan
| | - Tsuyoshi Shinozuka
- R&D Division, Daiichi Sankyo Co., Ltd. 1-2-58 Hiromachi Shinagawa-ku Tokyo 140-8710 Japan
| |
Collapse
|
14
|
Jia L, Eroglu TE, Wilders R, Verkerk AO, Tan HL. Carbamazepine Increases the Risk of Sudden Cardiac Arrest by a Reduction of the Cardiac Sodium Current. Front Cell Dev Biol 2022; 10:891996. [PMID: 35721495 PMCID: PMC9204209 DOI: 10.3389/fcell.2022.891996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 05/09/2022] [Indexed: 11/21/2022] Open
Abstract
Aim: To assess the risk of sudden cardiac arrest (SCA) associated with the use of carbamazepine (CBZ) and establish the possible underlying cellular electrophysiological mechanisms. Methods: The SCA risk association with CBZ was studied in general population cohorts using a case–control design (n = 5,473 SCA cases, 21,866 non-SCA controls). Effects of 1–100 µM CBZ on action potentials (APs) and individual membrane currents were determined in isolated rabbit and human cardiomyocytes using the patch clamp technique. Results: CBZ use was associated with increased risk of SCA compared with no use (adjusted odds ratio 1.90 [95% confidence interval: 1.12–3.24]). CBZ reduced the AP upstroke velocity of rabbit and human cardiomyocytes, without prominent changes in other AP parameters. The reduction occurred at ≥30 µM and was frequency-dependent with a more pronounced reduction at high stimulus frequencies. The cardiac sodium current (INa) was reduced at ≥30 μM; this was accompanied by a hyperpolarizing shift in the voltage-dependency of inactivation. The recovery from inactivation was slower, which is consistent with the more pronounced AP upstroke velocity reduction at high stimulus frequencies. The main cardiac K+ and Ca2+ currents were unaffected, except reduction of L-type Ca2+ current by 100 µM CBZ. Conclusion: CBZ use is associated with an increased risk of SCA in the general population. At concentrations of 30 µM and above, CBZ reduces AP upstroke velocity and INa in cardiomyocytes. Since the concentration of 30 µM is well within the therapeutic range (20–40 µM), we conclude that CBZ increases the risk of SCA by a reduction of the cardiac INa.
Collapse
Affiliation(s)
- Lixia Jia
- Department of Clinical and Experimental Cardiology, Heart Center, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Talip E. Eroglu
- Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
- Department of Cardiology, Herlev and Gentofte Hospital, University of Copenhagen, Gentofte, Denmark
| | - Ronald Wilders
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Arie O. Verkerk
- Department of Clinical and Experimental Cardiology, Heart Center, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Hanno L. Tan
- Department of Clinical and Experimental Cardiology, Heart Center, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- Netherlands Heart Institute, Utrecht, Netherlands
- *Correspondence: Hanno L. Tan,
| |
Collapse
|
15
|
Djamgoz MBA. Combinatorial Therapy of Cancer: Possible Advantages of Involving Modulators of Ionic Mechanisms. Cancers (Basel) 2022; 14:2703. [PMID: 35681682 PMCID: PMC9179511 DOI: 10.3390/cancers14112703] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 05/22/2022] [Accepted: 05/27/2022] [Indexed: 02/04/2023] Open
Abstract
Cancer is a global health problem that 1 in 2-3 people can expect to experience during their lifetime. Several different modalities exist for cancer management, but all of these suffer from significant shortcomings in both diagnosis and therapy. Apart from developing completely new therapies, a viable way forward is to improve the efficacy of the existing modalities. One way is to combine these with each other or with other complementary approaches. An emerging latter approach is derived from ionic mechanisms, mainly ion channels and exchangers. We evaluate the evidence for this systematically for the main treatment methods: surgery, chemotherapy, radiotherapy and targeted therapies (including monoclonal antibodies, steroid hormones, tyrosine kinase inhibitors and immunotherapy). In surgery, the possible systemic use of local anesthetics to suppress subsequent relapse is still being discussed. For all the other methods, there is significant positive evidence for several cancers and a range of modulators of ionic mechanisms. This applies also to some of the undesirable side effects of the treatments. In chemotherapy, for example, there is evidence for co-treatment with modulators of the potassium channel (Kv11.1), pH regulation (sodium-hydrogen exchanger) and Na+-K+-ATPase (digoxin). Voltage-gated sodium channels, shown previously to promote metastasis, appear to be particularly useful for co-targeting with inhibitors of tyrosine kinases, especially epidermal growth factor. It is concluded that combining current orthodox treatment modalities with modulators of ionic mechanisms can produce beneficial effects including (i) making the treatment more effective, e.g., by lowering doses; (ii) avoiding the onset of resistance to therapy; (iii) reducing undesirable side effects. However, in many cases, prospective clinical trials are needed to put the findings firmly into clinical context.
Collapse
Affiliation(s)
- Mustafa B. A. Djamgoz
- Department of Life Sciences, Imperial College London, South Kensington Campus, London SW7 2AZ, UK; ; Tel.: +44-796-181-6959
- Biotechnology Research Centre, Cyprus International University, Haspolat, Mersin 10, Turkey
| |
Collapse
|
16
|
Qiu SB, Xiao JH, Chen PR, Ai KL, Pan KL, Chen JK, Chen YW, Pan PS. Robust Synthesis of Tetra‐Boronate Esters Analogues and the Corresponding Boronic Acids Derivatives. European J Org Chem 2022. [DOI: 10.1002/ejoc.202200379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Shuo-Bei Qiu
- National Defense Medical Center Institute of Life Sciences TAIWAN
| | | | - Pin-Rui Chen
- Tamkang University Department of Chemistry TAIWAN
| | - Kuan-Lin Ai
- Tamkang University Department of Chemistry TAIWAN
| | - Kuan-Lin Pan
- Tamkang University Department of Chemistry TAIWAN
| | - Jen-Kun Chen
- National Health Research Institutes Institute of Biomedical Engineering and Nanomedicine TAIWAN
| | - Yi-Wei Chen
- Taipei Veterans General Hospital Oncology New Taipei City TAIWAN
| | - Po-Shen Pan
- Tamkang University Chemistry No.151, Yingzhuan Rd., Tamsui Dist., 25137 New Taipei City TAIWAN
| |
Collapse
|
17
|
Lin YC, Lai YC, Lin TH, Yang YC, Kuo CC. Selective stabilization of the intermediate inactivated Na+ channel by the new-generation anticonvulsant rufinamide. Biochem Pharmacol 2022; 197:114928. [DOI: 10.1016/j.bcp.2022.114928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 01/13/2022] [Accepted: 01/13/2022] [Indexed: 11/27/2022]
|
18
|
Cho HY, Chuang TH, Wu SN. Evidence for Inhibitory Perturbations on the Amplitude, Gating, and Hysteresis of A-Type Potassium Current, Produced by Lacosamide, a Functionalized Amino Acid with Anticonvulsant Properties. Int J Mol Sci 2022; 23:1171. [PMID: 35163091 PMCID: PMC8835568 DOI: 10.3390/ijms23031171] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 01/11/2022] [Accepted: 01/19/2022] [Indexed: 02/05/2023] Open
Abstract
Lacosamide (Vimpat®, LCS) is widely known as a functionalized amino acid with promising anti-convulsant properties; however, adverse events during its use have gradually appeared. Despite its inhibitory effect on voltage-gated Na+ current (INa), the modifications on varying types of ionic currents caused by this drug remain largely unexplored. In pituitary tumor (GH3) cells, we found that the presence of LCS concentration-dependently decreased the amplitude of A-type K+ current (IK(A)) elicited in response to membrane depolarization. The IK(A) amplitude in these cells was sensitive to attenuation by the application of 4-aminopyridine, 4-aminopyridine-3-methanol, or capsaicin but not by that of tetraethylammonium chloride. The effective IC50 value required for its reduction in peak or sustained IK(A) was calculated to be 102 or 42 µM, respectively, while the value of the dissociation constant (KD) estimated from the slow component in IK(A) inactivation at varying LCS concentrations was 52 µM. By use of two-step voltage protocol, the presence of this drug resulted in a rightward shift in the steady-state inactivation curve of IK(A) as well as in a slowing in the recovery time course of the current block; however, no change in the gating charge of the inactivation curve was detected in its presence. Moreover, the LCS addition led to an attenuation in the degree of voltage-dependent hysteresis for IK(A) elicitation by long-duration triangular ramp voltage commands. Likewise, the IK(A) identified in mouse mHippoE-14 neurons was also sensitive to block by LCS, coincident with an elevation in the current inactivation rate. Collectively, apart from its canonical action on INa inhibition, LCS was effective at altering the amplitude, gating, and hysteresis of IK(A) in excitable cells. The modulatory actions on IK(A), caused by LCS, could interfere with the functional activities of electrically excitable cells (e.g., pituitary tumor cells or hippocampal neurons).
Collapse
Affiliation(s)
- Hsin-Yen Cho
- Department of Physiology, College of Medicine, National Cheng Kung University, Tainan City 70101, Taiwan; (H.-Y.C.); (T.-H.C.)
| | - Tzu-Hsien Chuang
- Department of Physiology, College of Medicine, National Cheng Kung University, Tainan City 70101, Taiwan; (H.-Y.C.); (T.-H.C.)
| | - Sheng-Nan Wu
- Department of Physiology, College of Medicine, National Cheng Kung University, Tainan City 70101, Taiwan; (H.-Y.C.); (T.-H.C.)
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan City 70101, Taiwan
| |
Collapse
|
19
|
Labau JIR, Alsaloum M, Estacion M, Tanaka B, Dib-Hajj FB, Lauria G, Smeets HJM, Faber CG, Dib-Hajj S, Waxman SG. Lacosamide Inhibition of Na V1.7 Channels Depends on its Interaction With the Voltage Sensor Domain and the Channel Pore. Front Pharmacol 2022; 12:791740. [PMID: 34992539 PMCID: PMC8724789 DOI: 10.3389/fphar.2021.791740] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 12/02/2021] [Indexed: 12/14/2022] Open
Abstract
Lacosamide, developed as an anti-epileptic drug, has been used for the treatment of pain. Unlike typical anticonvulsants and local anesthetics which enhance fast-inactivation and bind within the pore of sodium channels, lacosamide enhances slow-inactivation of these channels, suggesting different binding mechanisms and mode of action. It has been reported that lacosamide's effect on NaV1.5 is sensitive to a mutation in the local anesthetic binding site, and that it binds with slow kinetics to the fast-inactivated state of NaV1.7. We recently showed that the NaV1.7-W1538R mutation in the voltage-sensing domain 4 completely abolishes NaV1.7 inhibition by clinically-achievable concentration of lacosamide. Our molecular docking analysis suggests a role for W1538 and pore residues as high affinity binding sites for lacosamide. Aryl sulfonamide sodium channel blockers are also sensitive to substitutions of the W1538 residue but not of pore residues. To elucidate the mechanism by which lacosamide exerts its effects, we used voltage-clamp recordings and show that lacosamide requires an intact local anesthetic binding site to inhibit NaV1.7 channels. Additionally, the W1538R mutation does not abrogate local anesthetic lidocaine-induced blockade. We also show that the naturally occurring arginine in NaV1.3 (NaV1.3-R1560), which corresponds to NaV1.7-W1538R, is not sufficient to explain the resistance of NaV1.3 to clinically-relevant concentrations of lacosamide. However, the NaV1.7-W1538R mutation conferred sensitivity to the NaV1.3-selective aryl-sulfonamide blocker ICA-121431. Together, the W1538 residue and an intact local anesthetic site are required for lacosamide's block of NaV1.7 at a clinically-achievable concentration. Moreover, the contribution of W1538 to lacosamide inhibitory effects appears to be isoform-specific.
Collapse
Affiliation(s)
- Julie I R Labau
- Department of Neurology, Yale University School of Medicine, New Haven, CT, United States.,Center for Neuroscience and Regeneration Research, Yale University, West Haven, CT, United States.,Rehabilitation Research Center, Veteran Affairs Connecticut Healthcare System, West Haven, CT, United States.,Department of Toxicogenomics, Clinical Genomics, Maastricht University Medical Centre+, Maastricht, Netherlands.,School of Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| | - Matthew Alsaloum
- Department of Neurology, Yale University School of Medicine, New Haven, CT, United States.,Center for Neuroscience and Regeneration Research, Yale University, West Haven, CT, United States.,Rehabilitation Research Center, Veteran Affairs Connecticut Healthcare System, West Haven, CT, United States.,Yale Medical Scientist Training Program, Yale School of Medicine, New Haven, CT, United States.,Interdepartmental Neuroscience Program, Yale School of Medicine, New Haven, CT, United States
| | - Mark Estacion
- Department of Neurology, Yale University School of Medicine, New Haven, CT, United States.,Center for Neuroscience and Regeneration Research, Yale University, West Haven, CT, United States.,Rehabilitation Research Center, Veteran Affairs Connecticut Healthcare System, West Haven, CT, United States
| | - Brian Tanaka
- Department of Neurology, Yale University School of Medicine, New Haven, CT, United States.,Center for Neuroscience and Regeneration Research, Yale University, West Haven, CT, United States.,Rehabilitation Research Center, Veteran Affairs Connecticut Healthcare System, West Haven, CT, United States
| | - Fadia B Dib-Hajj
- Department of Neurology, Yale University School of Medicine, New Haven, CT, United States.,Center for Neuroscience and Regeneration Research, Yale University, West Haven, CT, United States.,Rehabilitation Research Center, Veteran Affairs Connecticut Healthcare System, West Haven, CT, United States
| | - Giuseppe Lauria
- Neuroalgology Unit, IRCCS Foundation, "Carlo Besta" Neurological Institute, Milan, Italy.,Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Hubert J M Smeets
- Department of Toxicogenomics, Clinical Genomics, Maastricht University Medical Centre+, Maastricht, Netherlands.,School of Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| | - Catharina G Faber
- Department of Neurology, School of Mental Health and Neuroscience, Maastricht University Medical Center, Maastricht, Netherlands
| | - Sulayman Dib-Hajj
- Department of Neurology, Yale University School of Medicine, New Haven, CT, United States.,Center for Neuroscience and Regeneration Research, Yale University, West Haven, CT, United States.,Rehabilitation Research Center, Veteran Affairs Connecticut Healthcare System, West Haven, CT, United States
| | - Stephen G Waxman
- Department of Neurology, Yale University School of Medicine, New Haven, CT, United States.,Center for Neuroscience and Regeneration Research, Yale University, West Haven, CT, United States.,Rehabilitation Research Center, Veteran Affairs Connecticut Healthcare System, West Haven, CT, United States
| |
Collapse
|
20
|
Maadarani O, Bitar Z, Ashkanani A, Elzoueiry M, Elhabibi M, Gohar M, Almuwaizri M, Shoeb S, Alsaddah J. Cardiac Sodium Channel Blockade Due to Antiepileptic Drug Combination. Eur J Case Rep Intern Med 2021; 8:002839. [PMID: 34790627 DOI: 10.12890/2021_002839] [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: 08/20/2021] [Accepted: 08/26/2021] [Indexed: 11/05/2022] Open
Abstract
Drugs that inhibit voltage-dependent sodium channels are commonly used to treat epilepsy. Old and novel antiepileptic drugs are used either as monotherapy or in combination to control epilepsy. For a long time, carbamazepine has been used as the first choice for the treatment of simple and complex partial seizures. In the USA, lacosamide was approved in October 2008 as an adjunctive treatment for partial-onset seizures. We describe the effect of two sodium channel blockers on the heart of a patient with epilepsy. LEARNING POINTS Approximately 30% of patients with epilepsy require combination therapy with antiepileptic drugs for seizure reduction.Lacosamide and carbamazepine are both sodium channel blockers but exert their effects through different mechanisms.Electrocardiogram monitoring is necessary when lacosamide and carbamazepine are used together as this combination may predispose to seizures, conduction abnormalities and dysrhythmia.
Collapse
Affiliation(s)
- Ossama Maadarani
- Internal Medicine Department, Ahmadi Hospital, Kuwait Oil Company, Al Ahmadi, Kuwait
| | - Zouheir Bitar
- Internal Medicine Department, Ahmadi Hospital, Kuwait Oil Company, Al Ahmadi, Kuwait
| | - Abdelaziz Ashkanani
- Internal Medicine Department, Ahmadi Hospital, Kuwait Oil Company, Al Ahmadi, Kuwait
| | - Mahmoud Elzoueiry
- Internal Medicine Department, Ahmadi Hospital, Kuwait Oil Company, Al Ahmadi, Kuwait
| | - Mohamad Elhabibi
- Internal Medicine Department, Ahmadi Hospital, Kuwait Oil Company, Al Ahmadi, Kuwait
| | - Mohamad Gohar
- Internal Medicine Department, Ahmadi Hospital, Kuwait Oil Company, Al Ahmadi, Kuwait
| | - Mohamad Almuwaizri
- Nursing Department, Ahmadi Hospital, Kuwait Oil Company, Al Ahmadi, Kuwait
| | - Sania Shoeb
- Internal Medicine Department, Al Salam Hospital, Kuwait
| | - Jadan Alsaddah
- Department of Cardiology, Chest Diseases Hospital, Kuwait
| |
Collapse
|
21
|
Lou S, Cui S. Drug treatment of epilepsy: From serendipitous discovery to evolutionary mechanisms. Curr Med Chem 2021; 29:3366-3391. [PMID: 34514980 DOI: 10.2174/0929867328666210910124727] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 07/17/2021] [Accepted: 07/21/2021] [Indexed: 11/22/2022]
Abstract
Epilepsy is a chronic brain disorder caused by abnormal firing of neurons. Up to now, using antiepileptic drugs is the main method of epilepsy treatment. The development of antiepileptic drugs lasted for centuries. In general, most agents entering clinical practice act on the balance mechanisms of brain "excitability-inhibition". More specifically, they target voltage-gated ion channels, GABAergic transmission and glutamatergic transmission. In recent years, some novel drugs representing new mechanisms of action have been discovered. Although there are about 30 available drugs in the market, it is still in urgent need of discovering more effective and safer drugs. The development of new antiepileptic drugs is into a new era: from serendipitous discovery to evolutionary mechanism-based design. This article presents an overview of drug treatment of epilepsy, including a series of traditional and novel drugs.
Collapse
Affiliation(s)
- Shengying Lou
- Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou. China
| | - Sunliang Cui
- Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou. China
| |
Collapse
|
22
|
Roy PK, Rajesh Y, Mandal M. Therapeutic targeting of membrane-associated proteins in central nervous system tumors. Exp Cell Res 2021; 406:112760. [PMID: 34339674 DOI: 10.1016/j.yexcr.2021.112760] [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: 04/30/2021] [Revised: 06/28/2021] [Accepted: 07/28/2021] [Indexed: 12/09/2022]
Abstract
The activity of the most complex system, the central nervous system (CNS) is profoundly regulated by a huge number of membrane-associated proteins (MAP). A minor change stimulates immense chemical changes and the elicited response is organized by MAP, which acts as a receptor of that chemical or channel enabling the flow of ions. Slight changes in the activity or expression of these MAPs lead to severe consequences such as cognitive disorders, memory loss, or cancer. CNS tumors are heterogeneous in nature and hard-to-treat due to random mutations in MAPs; like as overexpression of EGFRvIII/TGFβR/VEGFR, change in adhesion molecules α5β3 integrin/SEMA3A, imbalance in ion channel proteins, etc. Extensive research is under process for developing new therapeutic approaches using these proteins such as targeted cytotoxic radiotherapy, drug-delivery, and prodrug activation, blocking of receptors like GluA1, developing viral vector against cell surface receptor. The combinatorial approach of these strategies along with the conventional one might be more potential. Henceforth, our review focuses on in-depth analysis regarding MAPs aiming for a better understanding for developing an efficient therapeutic approach for targeting CNS tumors.
Collapse
Affiliation(s)
- Pritam Kumar Roy
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, India
| | - Yetirajam Rajesh
- Department of Human and Molecular Genetics, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Mahitosh Mandal
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, India.
| |
Collapse
|
23
|
Kraus RL, Zhao F, Pall PS, Zhou D, Vardigan JD, Danziger A, Li Y, Daley C, Ballard JE, Clements MK, Klein RM, Holahan MA, Greshock TJ, Kim RM, Layton ME, Burgey CS, Serra J, Henze DA, Houghton AK. Na v1.7 target modulation and efficacy can be measured in nonhuman primate assays. Sci Transl Med 2021; 13:13/594/eaay1050. [PMID: 34011626 DOI: 10.1126/scitranslmed.aay1050] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 06/03/2020] [Accepted: 02/20/2021] [Indexed: 12/13/2022]
Abstract
Humans with loss-of-function mutations in the Nav1.7 channel gene (SCN9A) show profound insensitivity to pain, whereas those with gain-of-function mutations can have inherited pain syndromes. Therefore, inhibition of the Nav1.7 channel with a small molecule has been considered a promising approach for the treatment of various human pain conditions. To date, clinical studies conducted using selective Nav1.7 inhibitors have not provided analgesic efficacy sufficient to warrant further investment. Clinical studies to date used multiples of in vitro IC50 values derived from electrophysiological studies to calculate anticipated human doses. To increase the chance of clinical success, we developed rhesus macaque models of action potential propagation, nociception, and olfaction, to measure Nav1.7 target modulation in vivo. The potent and selective Nav1.7 inhibitors SSCI-1 and SSCI-2 dose-dependently blocked C-fiber nociceptor conduction in microneurography studies and inhibited withdrawal responses to noxious heat in rhesus monkeys. Pharmacological Nav1.7 inhibition also reduced odor-induced activation of the olfactory bulb (OB), measured by functional magnetic resonance imaging (fMRI) studies consistent with the anosmia reported in Nav1.7 loss-of-function patients. These data demonstrate that it is possible to measure Nav1.7 target modulation in rhesus macaques and determine the plasma concentration required to produce a predetermined level of inhibition. The calculated plasma concentration for preclinical efficacy could be used to guide human efficacious exposure estimates. Given the translatable nature of the assays used, it is anticipated that they can be also used in phase 1 clinical studies to measure target modulation and aid in the interpretation of phase 1 clinical data.
Collapse
Affiliation(s)
- Richard L Kraus
- Merck & Co. Inc., WP-14, 770 Sumneytown Pike, P.O. Box 4, West Point, PA 19486, USA.
| | - Fuqiang Zhao
- Merck & Co. Inc., WP-14, 770 Sumneytown Pike, P.O. Box 4, West Point, PA 19486, USA
| | - Parul S Pall
- Merck & Co. Inc., WP-14, 770 Sumneytown Pike, P.O. Box 4, West Point, PA 19486, USA
| | - Dan Zhou
- Merck & Co. Inc., WP-14, 770 Sumneytown Pike, P.O. Box 4, West Point, PA 19486, USA
| | - Joshua D Vardigan
- Merck & Co. Inc., WP-14, 770 Sumneytown Pike, P.O. Box 4, West Point, PA 19486, USA
| | - Andrew Danziger
- Merck & Co. Inc., WP-14, 770 Sumneytown Pike, P.O. Box 4, West Point, PA 19486, USA
| | - Yuxing Li
- Merck & Co. Inc., WP-14, 770 Sumneytown Pike, P.O. Box 4, West Point, PA 19486, USA
| | - Christopher Daley
- Merck & Co. Inc., WP-14, 770 Sumneytown Pike, P.O. Box 4, West Point, PA 19486, USA
| | - Jeanine E Ballard
- Merck & Co. Inc., WP-14, 770 Sumneytown Pike, P.O. Box 4, West Point, PA 19486, USA
| | - Michelle K Clements
- Merck & Co. Inc., WP-14, 770 Sumneytown Pike, P.O. Box 4, West Point, PA 19486, USA
| | - Rebecca M Klein
- Merck & Co. Inc., WP-14, 770 Sumneytown Pike, P.O. Box 4, West Point, PA 19486, USA
| | - Marie A Holahan
- Merck & Co. Inc., WP-14, 770 Sumneytown Pike, P.O. Box 4, West Point, PA 19486, USA
| | - Thomas J Greshock
- Merck & Co. Inc., WP-14, 770 Sumneytown Pike, P.O. Box 4, West Point, PA 19486, USA
| | - Ronald M Kim
- Merck & Co. Inc., WP-14, 770 Sumneytown Pike, P.O. Box 4, West Point, PA 19486, USA
| | - Mark E Layton
- Merck & Co. Inc., WP-14, 770 Sumneytown Pike, P.O. Box 4, West Point, PA 19486, USA
| | - Christopher S Burgey
- Merck & Co. Inc., WP-14, 770 Sumneytown Pike, P.O. Box 4, West Point, PA 19486, USA
| | - Jordi Serra
- Department of Clinical Neurophysiology, Ruskin Wing, King's College Hospital, Denmark Hill, London SE5 9RS, UK
| | - Darrell A Henze
- Merck & Co. Inc., WP-14, 770 Sumneytown Pike, P.O. Box 4, West Point, PA 19486, USA
| | - Andrea K Houghton
- Merck & Co. Inc., WP-14, 770 Sumneytown Pike, P.O. Box 4, West Point, PA 19486, USA
| |
Collapse
|
24
|
Luna C, Mizerska K, Quirce S, Belmonte C, Gallar J, Acosta MDC, Meseguer V. Sodium Channel Blockers Modulate Abnormal Activity of Regenerating Nociceptive Corneal Nerves After Surgical Lesion. Invest Ophthalmol Vis Sci 2021; 62:2. [PMID: 33393968 PMCID: PMC7797933 DOI: 10.1167/iovs.62.1.2] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Purpose To test the effect of different sodium channel blockers on the electrical activity of corneal nociceptors in intact and surgically injured corneas. Methods In anesthetized guinea pigs, a 4-mm diameter corneal flap was performed in one eye at a midstromal depth using a custom-made microkeratome. At different times after surgery (3 hours to 15 days), the electrical activity of corneal nociceptor fibers was recorded from ciliary nerve filaments in the superfused eye in vitro. Mechanical threshold was measured using calibrated von Frey hairs; chemical stimulation was performed applying 30-second CO2 gas pulses. The characteristics of the spontaneous and stimulus-evoked activity of corneal nociceptors recorded from intact and lesioned corneas, before and after treatment with the sodium channel blockers lidocaine, carbamazepine, and amitriptyline, were compared. Results No spontaneous or stimulus-evoked impulse activity was detected inside the flap at any of the studied time points. However, both were recorded from mechanonociceptor and polymodal nociceptors fibers in the surrounding corneal tissue, being significantly higher (sensitization) 24 to 48 hours after surgery. In these fibers, none of the tested drugs affected mechanical threshold, but they significantly reduced the CO2 response of polymodal nociceptors of intact and injured corneas. Likewise, they diminished significantly the transient increase in spontaneous and stimulus-evoked activity of sensitized polymodal nociceptors. Conclusions Na+ channel blockers decrease the excitability of intact and sensitized corneal nociceptor fibers, thus acting as potential tools to attenuate their abnormal activity, which underlies the spontaneous pain, hyperalgesia, and allodynia often accompanying surgical corneal lesions, as occurs after photorefractive surgery.
Collapse
Affiliation(s)
- Carolina Luna
- Instituto de Neurociencias, Universidad Miguel Hernández - CSIC, San Juan de Alicante, Spain
| | - Kamila Mizerska
- Instituto de Neurociencias, Universidad Miguel Hernández - CSIC, San Juan de Alicante, Spain
| | - Susana Quirce
- Instituto de Neurociencias, Universidad Miguel Hernández - CSIC, San Juan de Alicante, Spain
| | - Carlos Belmonte
- Instituto de Neurociencias, Universidad Miguel Hernández - CSIC, San Juan de Alicante, Spain
| | - Juana Gallar
- Instituto de Neurociencias, Universidad Miguel Hernández - CSIC, San Juan de Alicante, Spain.,Instituto de Investigación Sanitaria y Biomédica de Alicante, San Juan de Alicante, Spain
| | - María Del Carmen Acosta
- Instituto de Neurociencias, Universidad Miguel Hernández - CSIC, San Juan de Alicante, Spain
| | - Víctor Meseguer
- Instituto de Neurociencias, Universidad Miguel Hernández - CSIC, San Juan de Alicante, Spain
| |
Collapse
|
25
|
Griffin AM, Kahlig KM, Hatch RJ, Hughes ZA, Chapman ML, Antonio B, Marron BE, Wittmann M, Martinez-Botella G. Discovery of the First Orally Available, Selective K Na1.1 Inhibitor: In Vitro and In Vivo Activity of an Oxadiazole Series. ACS Med Chem Lett 2021; 12:593-602. [PMID: 33859800 PMCID: PMC8040054 DOI: 10.1021/acsmedchemlett.0c00675] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 03/01/2021] [Indexed: 12/15/2022] Open
Abstract
The gene KCNT1 encodes the sodium-activated potassium channel KNa1.1 (Slack, Slo2.2). Variants in the KCNT1 gene induce a gain-of-function (GoF) phenotype in ionic currents and cause a spectrum of intractable neurological disorders in infants and children, including epilepsy of infancy with migrating focal seizures (EIMFS) and autosomal dominant nocturnal frontal lobe epilepsy (ADNFLE). Effective treatment options for KCNT1-related disease are absent, and novel therapies are urgently required. We describe the development of a novel class of oxadiazole KNa1.1 inhibitors, leading to the discovery of compound 31 that reduced seizures and interictal spikes in a mouse model of KCNT1 GoF.
Collapse
Affiliation(s)
- Andrew M. Griffin
- Praxis
Precision Medicines, Research Innovation, Cambridge, Massachusetts 02142, United States
| | - Kristopher M. Kahlig
- Praxis
Precision Medicines, Research Innovation, Cambridge, Massachusetts 02142, United States
| | - Robert John Hatch
- Praxis
Precision Medicines, Research Innovation, Cambridge, Massachusetts 02142, United States
- The
Florey Institute of Neuroscience and Mental Health, Melbourne, VIC 3052, Australia
| | - Zoë A. Hughes
- Praxis
Precision Medicines, Research Innovation, Cambridge, Massachusetts 02142, United States
| | | | | | - Brian E. Marron
- Praxis
Precision Medicines, Research Innovation, Cambridge, Massachusetts 02142, United States
| | - Marion Wittmann
- Praxis
Precision Medicines, Research Innovation, Cambridge, Massachusetts 02142, United States
| | | |
Collapse
|
26
|
Xu Y, Yu Y, Wang Q, Li W, Zhang S, Liao X, Liu Y, Su Y, Zhao M, Zhang J. Active components of Bupleurum chinense and Angelica biserrata showed analgesic effects in formalin induced pain by acting on Nav1.7. JOURNAL OF ETHNOPHARMACOLOGY 2021; 269:113736. [PMID: 33359917 DOI: 10.1016/j.jep.2020.113736] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 12/09/2020] [Accepted: 12/19/2020] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGY RELEVANCE Pain is an unpleasant sensory and emotional experience, often accompanied by the occurrence of a variety of diseases. More than 800 kinds of traditional Chinese medicines (TCM) has now been reported for pain relief and several monomers have been developed into novel analgesic drugs. Bupleurum chinense and Angelica biserrata were representatives of the TCM that are currently available for the treatment of pain. AIM OF THE STUDY The study aims to detect the potential analgesic activity of each monomer of Bupleurum chinense and Angelica biserrata and to explore whether Nav1.7 is one of the targets for its analgesic activity. MATERIALS AND METHODS In this study, five monomers from Bupleurum chinense (Saikosaponin A, Saikosaponin B1, Saikosaponin B2, Saikosaponin C, Saikosaponin D) and five monomers from the Angelica biserrata (Osthole, Xanthotoxin, Imperatorin, Isoimperatorin, Psoralen) were examined by whole-cell patch-clamp on Nav1.7, which was closely associated with pain. Classical mouse pain models were also used to further verify the analgesic activity in vivo. RESULTS The results showed that monomers of Saikosaponins and Angelica biserrata all inhibited the peak currents of Nav1.7, indicating that Nav1.7 might be involved in the analgesic mechanism of Saikosaponins and Angelica biserrata. Among them, Saikosaponin A and Imperatorin showed the strongest inhibitory effect on Nav1.7. Furthermore, both Saikosaponin A and Imperatorin showed inhibitory effects on thermal pain and formalin-induced pain in phase II in vivo. CONCLUSION The results provide valuable information for future studies on the potential of TCM in alleviating pain.
Collapse
Affiliation(s)
- Yijia Xu
- School of Life Sciences and Biopharmaceutical Science, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, China
| | - Yue Yu
- College of Traditional Chinese Medicine, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, China
| | - Qi Wang
- School of Life Sciences and Biopharmaceutical Science, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, China
| | - Wenwen Li
- School of Medical Devices, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, China
| | - Suli Zhang
- School of Life Sciences and Biopharmaceutical Science, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, China
| | - Xiaoyuan Liao
- School of Medical Devices, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, China
| | - Yanfeng Liu
- School of Life Sciences and Biopharmaceutical Science, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, China
| | - Yang Su
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110004, China
| | - Mingyi Zhao
- School of Life Sciences and Biopharmaceutical Science, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, China.
| | - Jinghai Zhang
- School of Medical Devices, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, China.
| |
Collapse
|
27
|
Bladen C, Mirlohi S, Santiago M, Longworth M, Kassiou M, Banister S, Connor M. Modulation of human T-type calcium channels by synthetic cannabinoid receptor agonists in vitro. Neuropharmacology 2021; 187:108478. [PMID: 33600843 DOI: 10.1016/j.neuropharm.2021.108478] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 01/07/2021] [Accepted: 01/26/2021] [Indexed: 01/01/2023]
Abstract
BACKGROUND AND PURPOSE Consumption of Synthetic Cannabinoid Receptor agonists (SCRAs) is associated with severe adverse reactions including seizures, arrhythmias and death, but the molecular mechanisms surrounding SCRA toxicity are not yet established. These disease-like symptoms are also synonymous with altered T-type calcium channel activity which controls rhythmicity in the heart and brain. This study examined whether SCRAs alter T-type activity and whether this represents a possible mechanism of toxicity. EXPERIMENTAL APPROACH Fluorescence-based and electrophysiology assays were used to screen 16 structurally related synthetic cannabinoids for their ability to inhibit human T-type calcium channels expressed in HEK293 cells. The most potent compounds were then further examined using patch clamp electrophysiology. KEY RESULTS MDMB-CHMICA and AMB-CHMINACA potently blocked Cav3.2 with IC50 values of 1.5 and 0.74 μM respectively. Current inhibition increased from 47 to 80% and 45-87% respectively when the channel was in slow-inactivated state. Both SCRAs had little effect on steady state inactivation, however MDMB-CHMICA significantly shifted the half activation potential by -7mV. Neither drug produced frequency dependent block, in contrast to the phytocannabinoid Δ9-THC. CONCLUSIONS AND IMPLICATIONS SCRAs are potent agonists of CB1 receptors and can be extremely toxic, but observed toxicity also resembles symptoms associated with altered Cav3.2 activity. Many SCRAs tested were potent modulators of Cav3.2, raising the possibility that SC toxicity may be due in part to Cav3.2 modulation. This potent T-type channel modulation suggests the possibility of SCRAs as a new drug class with potential to treat diseases associated with altered T-type channel activity. This article is part of the special issue on 'Cannabinoids'.
Collapse
Affiliation(s)
- Chris Bladen
- Biomedical Sciences, Macquarie University, Sydney, NSW, Australia.
| | - Somayeh Mirlohi
- Biomedical Sciences, Macquarie University, Sydney, NSW, Australia
| | - Marina Santiago
- Biomedical Sciences, Macquarie University, Sydney, NSW, Australia
| | | | - Michael Kassiou
- School of Chemistry, University of Sydney, Sydney, NSW, 2006, Australia
| | - Sam Banister
- School of Chemistry, University of Sydney, Sydney, NSW, 2006, Australia; The Lambert Initiative for Cannabinoid Therapeutics, University of Sydney, Sydney, NSW, 2006, Australia
| | - Mark Connor
- Biomedical Sciences, Macquarie University, Sydney, NSW, Australia
| |
Collapse
|
28
|
Hinckley CA, Kuryshev Y, Sers A, Barre A, Buisson B, Naik H, Hajos M. Characterization of Vixotrigine, a Broad-Spectrum Voltage-Gated Sodium Channel Blocker. Mol Pharmacol 2021; 99:49-59. [PMID: 33298520 DOI: 10.1124/molpharm.120.000079] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 10/15/2020] [Indexed: 02/02/2023] Open
Abstract
Voltage-gated sodium channels (Navs) are promising targets for analgesic and antiepileptic therapies. Although specificity between Nav subtypes may be desirable to target specific neural types, such as nociceptors in pain, many broadly acting Nav inhibitors are clinically beneficial in neuropathic pain and epilepsy. Here, we present the first systematic characterization of vixotrigine, a Nav blocker. Using recombinant systems, we find that vixotrigine potency is enhanced in a voltage- and use-dependent manner, consistent with a state-dependent block of Navs. Furthermore, we find that vixotrigine potently inhibits sodium currents produced by both peripheral and central nervous system Nav subtypes, with use-dependent IC50 values between 1.76 and 5.12 μM. Compared with carbamazepine, vixotrigine shows higher potency and more profound state-dependent inhibition but a similar broad spectrum of action distinct from Nav1.7- and Nav1.8-specific blockers. We find that vixotrigine rapidly inhibits Navs and prolongs recovery from the fast-inactivated state. In native rodent dorsal root ganglion sodium channels, we find that vixotrigine shifts steady-state inactivation curves. Based on these results, we conclude that vixotrigine is a broad-spectrum, state-dependent Nav blocker. SIGNIFICANCE STATEMENT: Vixotrigine blocks both peripheral and central voltage-gated sodium channel subtypes. Neurophysiological approaches in recombinant systems and sensory neurons suggest this block is state-dependent.
Collapse
Affiliation(s)
- Christopher A Hinckley
- Biogen, Cambridge, Massachusetts (C.A.H., H.N.); Charles River Laboratories Cleveland, Inc., Cleveland, Ohio (Y.K.); Neuroservice, Aix-en-Provence, France (A.S., A.B., B.B.); and Department of Comparative Medicine, Yale University, New Haven, Connecticut (M.H.)
| | - Yuri Kuryshev
- Biogen, Cambridge, Massachusetts (C.A.H., H.N.); Charles River Laboratories Cleveland, Inc., Cleveland, Ohio (Y.K.); Neuroservice, Aix-en-Provence, France (A.S., A.B., B.B.); and Department of Comparative Medicine, Yale University, New Haven, Connecticut (M.H.)
| | - Alissende Sers
- Biogen, Cambridge, Massachusetts (C.A.H., H.N.); Charles River Laboratories Cleveland, Inc., Cleveland, Ohio (Y.K.); Neuroservice, Aix-en-Provence, France (A.S., A.B., B.B.); and Department of Comparative Medicine, Yale University, New Haven, Connecticut (M.H.)
| | - Alexander Barre
- Biogen, Cambridge, Massachusetts (C.A.H., H.N.); Charles River Laboratories Cleveland, Inc., Cleveland, Ohio (Y.K.); Neuroservice, Aix-en-Provence, France (A.S., A.B., B.B.); and Department of Comparative Medicine, Yale University, New Haven, Connecticut (M.H.)
| | - Bruno Buisson
- Biogen, Cambridge, Massachusetts (C.A.H., H.N.); Charles River Laboratories Cleveland, Inc., Cleveland, Ohio (Y.K.); Neuroservice, Aix-en-Provence, France (A.S., A.B., B.B.); and Department of Comparative Medicine, Yale University, New Haven, Connecticut (M.H.)
| | - Himanshu Naik
- Biogen, Cambridge, Massachusetts (C.A.H., H.N.); Charles River Laboratories Cleveland, Inc., Cleveland, Ohio (Y.K.); Neuroservice, Aix-en-Provence, France (A.S., A.B., B.B.); and Department of Comparative Medicine, Yale University, New Haven, Connecticut (M.H.)
| | - Mihaly Hajos
- Biogen, Cambridge, Massachusetts (C.A.H., H.N.); Charles River Laboratories Cleveland, Inc., Cleveland, Ohio (Y.K.); Neuroservice, Aix-en-Provence, France (A.S., A.B., B.B.); and Department of Comparative Medicine, Yale University, New Haven, Connecticut (M.H.)
| |
Collapse
|
29
|
Peng YS, Wu HT, Lai YC, Chen JL, Yang YC, Kuo CC. Inhibition of neuronal Na+ currents by lacosamide: Differential binding affinity and kinetics to different inactivated states. Neuropharmacology 2020; 179:108266. [DOI: 10.1016/j.neuropharm.2020.108266] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 07/06/2020] [Accepted: 07/28/2020] [Indexed: 10/23/2022]
|
30
|
Alelwani W, Elmorsy E, Kattan SW, Babteen NA, Alnajeebi AM, Al-Ghafari A, Carter WG. Carbamazepine induces a bioenergetics disruption to microvascular endothelial cells from the blood-brain barrier. Toxicol Lett 2020; 333:184-191. [PMID: 32805338 DOI: 10.1016/j.toxlet.2020.08.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 07/15/2020] [Accepted: 08/11/2020] [Indexed: 10/23/2022]
Abstract
Carbamazepine (CBZ) is a widely employed anti-seizure medication that crosses the blood-brain barrier (BBB) to exert its anti-convulsant action. The effects of CBZ on components of the BBB have yet to be completely delineated. Hence the current study evaluated the effects of CBZ upon mitochondrial functionality of BBB-derived microvascular endothelial cells isolated from Albino rats. The influence of CBZ on cell viability and barrier functions were evaluated by 3-(4,5 dimethylthiazol-2-yl)-2,5-diphenyl-tetrazolium bromide (MTT), lactate dehydrogenase, and electrophysiological assays over a drug concentration range of 0.1-1000 μM. Bioenergetics effects were measured via ATP production, mitochondrial complexes I and III activities, lactate production, and oxygen consumption rates (OCRs), and mitochondrial membrane potential, fluidity and lipid content. CBZ was cytotoxic to microvascular endothelial cells in a concentration and duration dependent manner. CBZ significantly diminished the endothelial cell's barrier functions, and impacted upon cellular bioenergetics: reducing mitochondrial complex activities with a parallel decrease in OCRs and increased anaerobic lactate production. CBZ significantly decreased mitochondrial membrane potential and induced an increase of membrane fluidity and decrease in levels of mitochondrial saturated and unsaturated fatty acids. In summary, CBZ disrupted functional activity of BBB endothelial cells via damage and modification of mitochondria functionality at therapeutically relevant concentrations.
Collapse
Affiliation(s)
- Walla Alelwani
- Department of Biochemistry, College of Science, University of Jeddah, Jeddah 80203, Saudi Arabia
| | - Ekramy Elmorsy
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Mansoura University, Mansoura, Egypt; School of Medicine, University of Nottingham, Royal Derby Hospital Centre, Derby, UK
| | - Shahad W Kattan
- Medical Laboratory Department, College of Applied Medical Sciences, Taibah University, Yanbu, Saudi Arabia
| | - Nouf Abubakr Babteen
- Department of Biochemistry, College of Science, University of Jeddah, Jeddah 80203, Saudi Arabia
| | - Afnan M Alnajeebi
- Department of Biochemistry, College of Science, University of Jeddah, Jeddah 80203, Saudi Arabia
| | - Ayat Al-Ghafari
- Biochemistry Department, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Wayne G Carter
- School of Medicine, University of Nottingham, Royal Derby Hospital Centre, Derby, UK.
| |
Collapse
|
31
|
Menezes LFS, Sabiá Júnior EF, Tibery DV, Carneiro LDA, Schwartz EF. Epilepsy-Related Voltage-Gated Sodium Channelopathies: A Review. Front Pharmacol 2020; 11:1276. [PMID: 33013363 PMCID: PMC7461817 DOI: 10.3389/fphar.2020.01276] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 07/31/2020] [Indexed: 12/29/2022] Open
Abstract
Epilepsy is a disease characterized by abnormal brain activity and a predisposition to generate epileptic seizures, leading to neurobiological, cognitive, psychological, social, and economic impacts for the patient. There are several known causes for epilepsy; one of them is the malfunction of ion channels, resulting from mutations. Voltage-gated sodium channels (NaV) play an essential role in the generation and propagation of action potential, and malfunction caused by mutations can induce irregular neuronal activity. That said, several genetic variations in NaV channels have been described and associated with epilepsy. These mutations can affect channel kinetics, modifying channel activation, inactivation, recovery from inactivation, and/or the current window. Among the NaV subtypes related to epilepsy, NaV1.1 is doubtless the most relevant, with more than 1500 mutations described. Truncation and missense mutations are the most observed alterations. In addition, several studies have already related mutated NaV channels with the electrophysiological functioning of the channel, aiming to correlate with the epilepsy phenotype. The present review provides an overview of studies on epilepsy-associated mutated human NaV1.1, NaV1.2, NaV1.3, NaV1.6, and NaV1.7.
Collapse
Affiliation(s)
- Luis Felipe Santos Menezes
- Laboratório de Neurofarmacologia, Departamento de Ciências Fisiológicas, Universidade de Brasília, Brasília, Brazil
| | - Elias Ferreira Sabiá Júnior
- Laboratório de Neurofarmacologia, Departamento de Ciências Fisiológicas, Universidade de Brasília, Brasília, Brazil
| | - Diogo Vieira Tibery
- Laboratório de Neurofarmacologia, Departamento de Ciências Fisiológicas, Universidade de Brasília, Brasília, Brazil
| | - Lilian Dos Anjos Carneiro
- Faculdade de Medicina, Centro Universitário Euro Americano, Brasília, Brazil.,Faculdade de Medicina, Centro Universitário do Planalto Central, Brasília, Brazil
| | - Elisabeth Ferroni Schwartz
- Laboratório de Neurofarmacologia, Departamento de Ciências Fisiológicas, Universidade de Brasília, Brasília, Brazil
| |
Collapse
|
32
|
Labau JIR, Estacion M, Tanaka BS, de Greef BTA, Hoeijmakers JGJ, Geerts M, Gerrits MM, Smeets HJM, Faber CG, Merkies ISJ, Lauria G, Dib-Hajj SD, Waxman SG. Differential effect of lacosamide on Nav1.7 variants from responsive and non-responsive patients with small fibre neuropathy. Brain 2020; 143:771-782. [PMID: 32011655 PMCID: PMC7089662 DOI: 10.1093/brain/awaa016] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 11/13/2019] [Accepted: 12/06/2019] [Indexed: 12/20/2022] Open
Abstract
Small fibre neuropathy is a common pain disorder, which in many cases fails to respond to treatment with existing medications. Gain-of-function mutations of voltage-gated sodium channel Nav1.7 underlie dorsal root ganglion neuronal hyperexcitability and pain in a subset of patients with small fibre neuropathy. Recent clinical studies have demonstrated that lacosamide, which blocks sodium channels in a use-dependent manner, attenuates pain in some patients with Nav1.7 mutations; however, only a subgroup of these patients responded to the drug. Here, we used voltage-clamp recordings to evaluate the effects of lacosamide on five Nav1.7 variants from patients who were responsive or non-responsive to treatment. We show that, at the clinically achievable concentration of 30 μM, lacosamide acts as a potent sodium channel inhibitor of Nav1.7 variants carried by responsive patients, via a hyperpolarizing shift of voltage-dependence of both fast and slow inactivation and enhancement of use-dependent inhibition. By contrast, the effects of lacosamide on slow inactivation and use-dependence in Nav1.7 variants from non-responsive patients were less robust. Importantly, we found that lacosamide selectively enhances fast inactivation only in variants from responders. Taken together, these findings begin to unravel biophysical underpinnings that contribute to responsiveness to lacosamide in patients with small fibre neuropathy carrying select Nav1.7 variants.
Collapse
Affiliation(s)
- Julie I R Labau
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510, USA.,Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06510, USA.,Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA.,Department of Neurology, School of Mental Health and Neuroscience, Maastricht University Medical Center, Maastricht, The Netherlands.,Department of Genetics and Cell Biology, Clinical Genomics Unit, Maastricht University, Maastricht, The Netherlands
| | - Mark Estacion
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510, USA.,Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06510, USA.,Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Brian S Tanaka
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510, USA.,Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06510, USA.,Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Bianca T A de Greef
- Department of Neurology, School of Mental Health and Neuroscience, Maastricht University Medical Center, Maastricht, The Netherlands.,Department of Clinical Epidemiology and Medical Technology Assessment (KEMTA), Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Janneke G J Hoeijmakers
- Department of Neurology, School of Mental Health and Neuroscience, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Margot Geerts
- Department of Neurology, School of Mental Health and Neuroscience, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Monique M Gerrits
- Department of Clinical Genetics, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Hubert J M Smeets
- Department of Genetics and Cell Biology, Clinical Genomics Unit, Maastricht University, Maastricht, The Netherlands
| | - Catharina G Faber
- Department of Neurology, School of Mental Health and Neuroscience, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Ingemar S J Merkies
- Department of Neurology, School of Mental Health and Neuroscience, Maastricht University Medical Center, Maastricht, The Netherlands.,Department of Neurology, St. Elisabeth Hospital, Willemstad, Curaçao
| | - Giuseppe Lauria
- Neuroalgology Unit, IRCCS Foundation, "Carlo Besta" Neurological Institute, Milan, Italy.,Department of Biomedical and Clinical Sciences "Luigi Sacco", University of Milan, Italy
| | - Sulayman D Dib-Hajj
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510, USA.,Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06510, USA.,Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Stephen G Waxman
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510, USA.,Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06510, USA.,Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
| |
Collapse
|
33
|
Recent advances in treatment of epilepsy-related sodium channelopathies. Eur J Paediatr Neurol 2020; 24:123-128. [PMID: 31889633 DOI: 10.1016/j.ejpn.2019.12.009] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 12/06/2019] [Indexed: 11/22/2022]
Abstract
Voltage-gated sodium channels (VGSCs) play a crucial role in generation of action potentials. Pathogenic variants in the five human brain expressed VGSC genes, SCN1A, SCN2A, SCN3A, SCN8A and SCN1B have been associated with a spectrum of epilepsy phenotypes and neurodevelopmental disorders. In the last decade, next generation sequencing techniques have revolutionized the way we diagnose these channelopathies, which is paving the way towards precision medicine. Knowing the functional effect (Loss-of-function versus Gain-of-function) of a variant is not only important for understanding the underlying pathophysiology, but it is particularly crucial to orient therapeutic decisions. Here we provide a review of the literature dealing with treatment options in epilepsy-related sodium channelopathies, including the current and emerging medications.
Collapse
|
34
|
Moutal A, White KA, Chefdeville A, Laufmann RN, Vitiello PF, Feinstein D, Weimer JM, Khanna R. Dysregulation of CRMP2 Post-Translational Modifications Drive Its Pathological Functions. Mol Neurobiol 2019; 56:6736-6755. [PMID: 30915713 PMCID: PMC6728212 DOI: 10.1007/s12035-019-1568-4] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 03/15/2019] [Indexed: 12/13/2022]
Abstract
Collapsin response mediator proteins (CRMPs) are a family of ubiquitously expressed, homologous phosphoproteins best known for coordinating cytoskeletal formation and regulating cellular division, migration, polarity, and synaptic connection. CRMP2, the most studied of the five family members, is best known for its affinity for tubulin heterodimers and function in regulating the microtubule network. These functions are tightly regulated by post-translational modifications including phosphorylation, SUMOylation, oxidation, and O-GlcNAcylation. While CRMP2's physiological functions rely mostly on its non-phosphorylated state, dysregulation of CRMP2 phosphorylation and SUMOylation has been reported to be involved in the pathophysiology of multiple diseases including cancer, chronic pain, spinal cord injury, neurofibromatosis type 1, and others. Here, we provide a consolidated update on what is known about CRMP2 signaling and function, first focusing on axonal growth and neuronal polarity, then illustrating the link between dysregulated CRMP2 post-translational modifications and diseases. We additionally discuss the roles of CRMP2 in non-neuronal cells, both in the CNS and regions of the periphery. Finally, we offer thoughts on the therapeutic implications of modulating CRMP2 function in a variety of diseases.
Collapse
Affiliation(s)
- Aubin Moutal
- Department of Pharmacology, College of Medicine, University of Arizona, 1501 North Campbell Drive, P.O. Box 245050, Tucson, AZ, 85724, USA
| | - Katherine A White
- Pediatrics and Rare Diseases Group, Sanford Research, 2301 E 60th St N, Sioux Falls, SD, 57104, USA
| | - Aude Chefdeville
- Department of Pharmacology, College of Medicine, University of Arizona, 1501 North Campbell Drive, P.O. Box 245050, Tucson, AZ, 85724, USA
| | - Rachel N Laufmann
- Pediatrics and Rare Diseases Group, Sanford Research, 2301 E 60th St N, Sioux Falls, SD, 57104, USA
| | - Peter F Vitiello
- Department of Pharmacology, College of Medicine, University of Arizona, 1501 North Campbell Drive, P.O. Box 245050, Tucson, AZ, 85724, USA
- Department of Pediatrics, Sanford School of Medicine, University of South Dakota, Sioux Falls, SD, USA
| | - Douglas Feinstein
- Department of Veterans Affairs, Jesse Brown VA Medical Center, University of Illinois at Chicago, Chicago, IL, USA
| | - Jill M Weimer
- Department of Pharmacology, College of Medicine, University of Arizona, 1501 North Campbell Drive, P.O. Box 245050, Tucson, AZ, 85724, USA.
- Department of Pediatrics, Sanford School of Medicine, University of South Dakota, Sioux Falls, SD, USA.
| | - Rajesh Khanna
- Department of Pharmacology, College of Medicine, University of Arizona, 1501 North Campbell Drive, P.O. Box 245050, Tucson, AZ, 85724, USA.
- Pediatrics and Rare Diseases Group, Sanford Research, 2301 E 60th St N, Sioux Falls, SD, 57104, USA.
- Department of Anesthesiology, University of Arizona, Tucson, AZ, USA.
- The Center for Innovation in Brain Sciences, The University of Arizona Health Sciences, Tucson, AZ, USA.
| |
Collapse
|
35
|
Flood E, Boiteux C, Lev B, Vorobyov I, Allen TW. Atomistic Simulations of Membrane Ion Channel Conduction, Gating, and Modulation. Chem Rev 2019; 119:7737-7832. [DOI: 10.1021/acs.chemrev.8b00630] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Emelie Flood
- School of Science, RMIT University, Melbourne, Victoria 3000, Australia
| | - Céline Boiteux
- School of Science, RMIT University, Melbourne, Victoria 3000, Australia
| | - Bogdan Lev
- School of Science, RMIT University, Melbourne, Victoria 3000, Australia
| | - Igor Vorobyov
- Department of Physiology & Membrane Biology/Department of Pharmacology, University of California, Davis, 95616, United States
| | - Toby W. Allen
- School of Science, RMIT University, Melbourne, Victoria 3000, Australia
| |
Collapse
|
36
|
Caroleo MC, Brizzi A, De Rosa M, Pandey A, Gallelli L, Badolato M, Carullo G, Cione E. Targeting Neuropathic Pain: Pathobiology, Current Treatment and Peptidomimetics as a New Therapeutic Opportunity. Curr Med Chem 2019; 27:1469-1500. [PMID: 31142248 DOI: 10.2174/0929867326666190530121133] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 01/25/2019] [Accepted: 02/15/2019] [Indexed: 12/18/2022]
Abstract
There is a huge need for pharmaceutical agents for the treatment of chronic Neuropathic Pain (NP), a complex condition where patients can suffer from either hyperalgesia or allodynia originating from central or peripheral nerve injuries. To date, the therapeutic guidelines include the use of tricyclic antidepressants, serotonin-noradrenaline reuptake inhibitors and anticonvulsants, beside the use of natural compounds and non-pharmacological options. Unfortunately, these drugs suffer from limited efficacy and serious dose-dependent adverse effects. In the last decades, the heptapeptide SP1-7, the major bioactive metabolite produced by Substance P (SP) cleavage, has been extensively investigated as a potential target for the development of novel peptidomimetic molecules to treat NP. Although the physiological effects of this SP fragment have been studied in detail, the mechanism behind its action is not fully clarified and the target for SP1-7 has not been identified yet. Nevertheless, specific binding sites for the heptapeptide have been found in brain and spinal cord of both mouse and rats. Several Structure-Affinity Relationship (SAR) studies on SP1-7 and some of its synthetic analogues have been carried out aiming to developing more metabolically stable and effective small molecule SP1-7-related amides that could be used as research tools for a better understanding of the SP1-7 system and, in a longer perspective, as potential therapeutic agents for future treatment of NP.
Collapse
Affiliation(s)
- Maria Cristina Caroleo
- Department of Pharmacy and Health and Nutrition Sciences, University of Calabria, Edificio Polifunzionale, 87026 Rende (CS), Italy
| | - Antonella Brizzi
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Polo Scientifico San Miniato, Via A. Moro 2, 53100 Siena, Italy
| | - Maria De Rosa
- Drug Discovery Unit, Ri.MED Foundation, Palermo 90133, Italy
| | - Ankur Pandey
- Department of Chemistry and Center of Advanced Studies in Chemistry, Punjab University, Chandigarh, India
| | - Luca Gallelli
- Department of Health Science, School of Medicine, University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| | - Mariateresa Badolato
- Department of Pharmacy and Health and Nutrition Sciences, University of Calabria, Edificio Polifunzionale, 87026 Rende (CS), Italy
| | - Gabriele Carullo
- Department of Pharmacy and Health and Nutrition Sciences, University of Calabria, Edificio Polifunzionale, 87026 Rende (CS), Italy
| | - Erika Cione
- Department of Pharmacy and Health and Nutrition Sciences, University of Calabria, Edificio Polifunzionale, 87026 Rende (CS), Italy
| |
Collapse
|
37
|
Navarrete-Modesto V, Orozco-Suárez S, Feria-Romero IA, Rocha L. The molecular hallmarks of epigenetic effects mediated by antiepileptic drugs. Epilepsy Res 2019; 149:53-65. [DOI: 10.1016/j.eplepsyres.2018.11.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 10/16/2018] [Accepted: 11/14/2018] [Indexed: 02/06/2023]
|
38
|
Patel R, Kucharczyk M, Montagut‐Bordas C, Lockwood S, Dickenson AH. Neuropathy following spinal nerve injury shares features with the irritable nociceptor phenotype: A back-translational study of oxcarbazepine. Eur J Pain 2019; 23:183-197. [PMID: 30091265 PMCID: PMC6396087 DOI: 10.1002/ejp.1300] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 07/24/2018] [Accepted: 07/31/2018] [Indexed: 12/30/2022]
Abstract
BACKGROUND The term 'irritable nociceptor' was coined to describe neuropathic patients characterized by evoked hypersensitivity and preservation of primary afferent fibres. Oxcarbazepine is largely ineffectual in an overall patient population, but has clear efficacy in a subgroup with the irritable nociceptor profile. We examine whether neuropathy in rats induced by spinal nerve injury shares overlapping pharmacological sensitivity with the irritable nociceptor phenotype using drugs that target sodium channels. METHODS In vivo electrophysiology was performed in anaesthetized spinal nerve ligated (SNL) and sham-operated rats to record from wide dynamic range (WDR) neurones in the ventral posterolateral thalamus (VPL) and dorsal horn. RESULTS In neuropathic rats, spontaneous activity in the VPL was substantially attenuated by spinal lidocaine, an effect that was absent in sham rats. The former measure was in part dependent on ongoing peripheral activity as intraplantar lidocaine also reduced aberrant spontaneous thalamic firing. Systemic oxcarbazepine had no effect on wind-up of dorsal horn neurones in sham and SNL rats. However, in SNL rats, oxcarbazepine markedly inhibited punctate mechanical-, dynamic brush- and cold-evoked neuronal responses in the VPL and dorsal horn, with minimal effects on heat-evoked responses. In addition, oxcarbazepine inhibited spontaneous activity in the VPL. Intraplantar injection of the active metabolite licarbazepine replicated the effects of systemic oxcarbazepine, supporting a peripheral locus of action. CONCLUSIONS We provide evidence that ongoing activity in primary afferent fibres drives spontaneous thalamic firing after spinal nerve injury and that oxcarbazepine through a peripheral mechanism exhibits modality-selective inhibitory effects on sensory neuronal processing. SIGNIFICANCE The inhibitory effects of lidocaine and oxcarbazepine in this rat model of neuropathy resemble the clinical observations in the irritable nociceptor patient subgroup and support a mechanism-based rationale for bench-to-bedside translation when screening novel drugs.
Collapse
Affiliation(s)
- Ryan Patel
- Department of Neuroscience, Physiology and PharmacologyUniversity College LondonLondonUK
| | - Mateusz Kucharczyk
- Department of Neuroscience, Physiology and PharmacologyUniversity College LondonLondonUK
| | | | - Stevie Lockwood
- Department of Neuroscience, Physiology and PharmacologyUniversity College LondonLondonUK
| | - Anthony H. Dickenson
- Department of Neuroscience, Physiology and PharmacologyUniversity College LondonLondonUK
| |
Collapse
|
39
|
Fenestrations control resting-state block of a voltage-gated sodium channel. Proc Natl Acad Sci U S A 2018; 115:13111-13116. [PMID: 30518562 DOI: 10.1073/pnas.1814928115] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Potency of drug action is usually determined by binding to a specific receptor site on target proteins. In contrast to this conventional paradigm, we show here that potency of local anesthetics (LAs) and antiarrhythmic drugs (AADs) that block sodium channels is controlled by fenestrations that allow drug access to the receptor site directly from the membrane phase. Voltage-gated sodium channels initiate action potentials in nerve and cardiac muscle, where their hyperactivity causes pain and cardiac arrhythmia, respectively. LAs and AADs selectively block sodium channels in rapidly firing nerve and muscle cells to relieve these conditions. The structure of the ancestral bacterial sodium channel NaVAb, which is also blocked by LAs and AADs, revealed fenestrations connecting the lipid phase of the membrane to the central cavity of the pore. We cocrystallized lidocaine and flecainide with NavAb, which revealed strong drug-dependent electron density in the central cavity of the pore. Mutation of the contact residue T206 greatly reduced drug potency, confirming this site as the receptor for LAs and AADs. Strikingly, mutations of the fenestration cap residue F203 changed fenestration size and had graded effects on resting-state block by flecainide, lidocaine, and benzocaine, the potencies of which were altered from 51- to 2.6-fold in order of their molecular size. These results show that conserved fenestrations in the pores of sodium channels are crucial pharmacologically and determine the level of resting-state block by widely used drugs. Fine-tuning drug access through fenestrations provides an unexpected avenue for structure-based design of ion-channel-blocking drugs.
Collapse
|
40
|
Cuomo I, Piacentino D, Kotzalidis GD, Lionetto L, De Filippis S. Lacosamide in bipolar disorder: A 30-day comparison to a retrospective control group treated with other antiepileptics. Psychiatry Clin Neurosci 2018; 72:864-875. [PMID: 30251375 DOI: 10.1111/pcn.12784] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 09/03/2018] [Accepted: 09/19/2018] [Indexed: 12/21/2022]
Abstract
AIM Bipolar disorder (BD) is often treated with anticonvulsants. Lacosamide has not been tested in BD. We assessed its effects in a hospital setting in patients with BD without epilepsy. METHODS We treated 102 consecutive hospitalized patients with acute BD with lacosamide 50-300 mg/day. We compared this sample with a retrospective sample treated with other antiepileptics (OAE). We rated patients after 3, 7, 15, and 30 days of treatment with the Brief Psychiatric Rating Scale, Young Mania Rating Scale, Hamilton Depression Rating Scale, Hamilton Anxiety Rating Scale, Clinical Global Impressions - Severity, and Global Assessment of Functioning. RESULTS Patients receiving lacosamide were significantly younger and had fewer mixed episodes at intake, and less substance use disorder comorbidity than those receiving OAE. Both groups showed positive effects on all measures. The two groups did not differ on any clinical measure at baseline, but from the 3rd day on, lacosamide patients fared better than OAE patients on the Young Mania Rating Scale and Clinical Global Impressions - Severity and worse on the Hamilton Anxiety Rating Scale. From the 15th day, OAE patients scored better on the Brief Psychiatric Rating Scale. Global Assessment of Functioning scores were significantly more improved in the lacosamide patients. Age, substance use disorder comorbidity, episode type, and educational level significantly affected results. No interactions were found amongst these parameters. CONCLUSION Lacosamide was effective in reducing psychopathology, mania, depression, and anxiety and in improving global functioning in patients with BD-I/II disorder in the short term, with few side-effects. Lacosamide improved mania, clinical severity, and global functioning better than OAE at doses lower than those used in epilepsy.
Collapse
Affiliation(s)
- Ilaria Cuomo
- Department of Neuropsychiatry, Clinica Von Siebenthal Neuropsychiatric Hospital, Rome, Italy.,NESMOS Department, School of Medicine and Psychology, Sapienza University, Rome, Italy
| | - Daria Piacentino
- NESMOS Department, School of Medicine and Psychology, Sapienza University, Rome, Italy
| | - Georgios D Kotzalidis
- NESMOS Department, School of Medicine and Psychology, Sapienza University, Rome, Italy
| | - Luana Lionetto
- Department of Advanced Molecular Analysis, School of Medicine and Psychology, Sapienza University, Rome, Italy
| | - Sergio De Filippis
- NESMOS Department, School of Medicine and Psychology, Sapienza University, Rome, Italy
| |
Collapse
|
41
|
Namer B, Schmidt D, Eberhardt E, Maroni M, Dorfmeister E, Kleggetveit IP, Kaluza L, Meents J, Gerlach A, Lin Z, Winterpacht A, Dragicevic E, Kohl Z, Schüttler J, Kurth I, Warncke T, Jorum E, Winner B, Lampert A. Pain relief in a neuropathy patient by lacosamide: Proof of principle of clinical translation from patient-specific iPS cell-derived nociceptors. EBioMedicine 2018; 39:401-408. [PMID: 30503201 PMCID: PMC6354557 DOI: 10.1016/j.ebiom.2018.11.042] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 11/06/2018] [Accepted: 11/19/2018] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Small fiber neuropathy (SFN) is a severe and disabling chronic pain syndrome with no causal and limited symptomatic treatment options. Mechanistically based individual treatment is not available. We report an in-vitro predicted individualized treatment success in one therapy-refractory Caucasian patient suffering from SFN for over ten years. METHODS Intrinsic excitability of human induced pluripotent stem cell (iPSC) derived nociceptors from this patient and respective controls were recorded on multi-electrode (MEA) arrays, in the presence and absence of lacosamide. The patient's pain ratings were assessed by a visual analogue scale (10: worst pain, 0: no pain) and treatment effect was objectified by microneurography recordings of the patient's single nerve C-fibers. FINDINGS We identified patient-specific changes in iPSC-derived nociceptor excitability in MEA recordings, which were reverted by the FDA-approved compound lacosamide in vitro. Using this drug for individualized treatment of this patient, the patient's pain ratings decreased from 7.5 to 1.5. Consistent with the pain relief reported by the patient, microneurography recordings of the patient's single nerve fibers mirrored a reduced spontaneous nociceptor (C-fiber) activity in the patient during lacosamide treatment. Microneurography recordings yielded an objective measurement of altered peripheral nociceptor activity following treatment. INTERPRETATION Thus, we are here presenting one example of successful patient specific precision medicine using iPSC technology and individualized therapeutic treatment based on patient-derived sensory neurons.
Collapse
Affiliation(s)
- Barbara Namer
- Institute of Physiology and Pathophysiology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; Department of Experimental Pain Research, Medical Faculty Mannheim of Heidelberg University, Germany; Interdisciplinary Center for Clinical Research within the faculty of Medicine at the RWTH Aachen University, 52074 Aachen, Germany
| | - Diana Schmidt
- Department of Stem Cell Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Esther Eberhardt
- Department of Anesthesiology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Michele Maroni
- Department of Stem Cell Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; Department of Anesthesiology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Eva Dorfmeister
- Department of Stem Cell Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | | | - Luisa Kaluza
- Institute of Physiology, Medical Faculty, RWTH Aachen University, 52074 Aachen, Germany
| | - Jannis Meents
- Institute of Physiology, Medical Faculty, RWTH Aachen University, 52074 Aachen, Germany
| | | | | | - Andreas Winterpacht
- Institute of Human Genetics, Medical Faculty, RWTH Aachen University, 52074 Aachen, Germany
| | | | - Zacharias Kohl
- Department of Molecular Neurology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Jürgen Schüttler
- Department of Anesthesiology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Ingo Kurth
- Institute of Human Genetics, Medical Faculty, RWTH Aachen University, 52074 Aachen, Germany
| | - Torhild Warncke
- Department of Anesthesiology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Ellen Jorum
- Department of Neurology, Oslo University Hospital-Rikshospitalet, Oslo, Norway; Section of Clinical Neurophysiology, Department of Neurology, Oslo University Hospital-Rikshospitalet, Oslo, Norway
| | - Beate Winner
- Department of Stem Cell Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; Center of Rare Diseases Erlangen (ZSEER), Germany.
| | - Angelika Lampert
- Institute of Physiology, Medical Faculty, RWTH Aachen University, 52074 Aachen, Germany.
| |
Collapse
|
42
|
Ghovanloo MR, Shuart NG, Mezeyova J, Dean RA, Ruben PC, Goodchild SJ. Inhibitory effects of cannabidiol on voltage-dependent sodium currents. J Biol Chem 2018; 293:16546-16558. [PMID: 30219789 PMCID: PMC6204917 DOI: 10.1074/jbc.ra118.004929] [Citation(s) in RCA: 145] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 09/12/2018] [Indexed: 12/25/2022] Open
Abstract
Cannabis sativa contains many related compounds known as phytocannabinoids. The main psychoactive and nonpsychoactive compounds are Δ9-tetrahydrocannabinol (THC) and cannabidiol (CBD), respectively. Much of the evidence for clinical efficacy of CBD-mediated antiepileptic effects has been from case reports or smaller surveys. The mechanisms for CBD's anticonvulsant effects are unclear and likely involve noncannabinoid receptor pathways. CBD is reported to modulate several ion channels, including sodium channels (Nav). Evaluating the therapeutic mechanisms and safety of CBD demands a richer understanding of its interactions with central nervous system targets. Here, we used voltage-clamp electrophysiology of HEK-293 cells and iPSC neurons to characterize the effects of CBD on Nav channels. Our results show that CBD inhibits hNav1.1-1.7 currents, with an IC50 of 1.9-3.8 μm, suggesting that this inhibition could occur at therapeutically relevant concentrations. A steep Hill slope of ∼3 suggested multiple interactions of CBD with Nav channels. CBD exhibited resting-state blockade, became more potent at depolarized potentials, and also slowed recovery from inactivation, supporting the idea that CBD binding preferentially stabilizes inactivated Nav channel states. We also found that CBD inhibits other voltage-dependent currents from diverse channels, including bacterial homomeric Nav channel (NaChBac) and voltage-gated potassium channel subunit Kv2.1. Lastly, the CBD block of Nav was temperature-dependent, with potency increasing at lower temperatures. We conclude that CBD's mode of action likely involves 1) compound partitioning in lipid membranes, which alters membrane fluidity affecting gating, and 2) undetermined direct interactions with sodium and potassium channels, whose combined effects are loss of channel excitability.
Collapse
Affiliation(s)
- Mohammad-Reza Ghovanloo
- From the Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada and
- the Department of Cellular and Molecular Biology, Xenon Pharmaceuticals, Burnaby, British Columbia V5G 4W8, Canada
| | - Noah Gregory Shuart
- the Department of Cellular and Molecular Biology, Xenon Pharmaceuticals, Burnaby, British Columbia V5G 4W8, Canada
| | - Janette Mezeyova
- the Department of Cellular and Molecular Biology, Xenon Pharmaceuticals, Burnaby, British Columbia V5G 4W8, Canada
| | - Richard A Dean
- the Department of Cellular and Molecular Biology, Xenon Pharmaceuticals, Burnaby, British Columbia V5G 4W8, Canada
| | - Peter C Ruben
- From the Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada and
| | - Samuel J Goodchild
- the Department of Cellular and Molecular Biology, Xenon Pharmaceuticals, Burnaby, British Columbia V5G 4W8, Canada
| |
Collapse
|
43
|
Abstract
Neurofibromatosis type 1 (NF1), a genetic disorder linked to inactivating mutations or a homozygous deletion of the Nf1 gene, is characterized by tumorigenesis, cognitive dysfunction, seizures, migraine, and pain. Omic studies on human NF1 tissues identified an increase in the expression of collapsin response mediator protein 2 (CRMP2), a cytosolic protein reported to regulate the trafficking and activity of presynaptic N-type voltage-gated calcium (Cav2.2) channels. Because neurofibromin, the protein product of the Nf1 gene, binds to and inhibits CRMP2, the neurofibromin-CRMP2 signaling cascade will likely affect Ca channel activity and regulate nociceptive neurotransmission and in vivo responses to noxious stimulation. Here, we investigated the function of neurofibromin-CRMP2 interaction on Cav2.2. Mapping of >275 peptides between neurofibromin and CRMP2 identified a 15-amino acid CRMP2-derived peptide that, when fused to the tat transduction domain of HIV-1, inhibited Ca influx in dorsal root ganglion neurons. This peptide mimics the negative regulation of CRMP2 activity by neurofibromin. Neurons treated with tat-CRMP2/neurofibromin regulating peptide 1 (t-CNRP1) exhibited a decreased Cav2.2 membrane localization, and uncoupling of neurofibromin-CRMP2 and CRMP2-Cav2.2 interactions. Proteomic analysis of a nanodisc-solubilized membrane protein library identified syntaxin 1A as a novel CRMP2-binding protein whose interaction with CRMP2 was strengthened in neurofibromin-depleted cells and reduced by t-CNRP1. Stimulus-evoked release of calcitonin gene-related peptide from lumbar spinal cord slices was inhibited by t-CNRP1. Intrathecal administration of t-CNRP1 was antinociceptive in experimental models of inflammatory, postsurgical, and neuropathic pain. Our results demonstrate the utility of t-CNRP1 to inhibit CRMP2 protein-protein interactions for the potential treatment of pain.
Collapse
|
44
|
Zheng YM, Wang WF, Li YF, Yu Y, Gao ZB. Enhancing inactivation rather than reducing activation of Nav1.7 channels by a clinically effective analgesic CNV1014802. Acta Pharmacol Sin 2018; 39:587-596. [PMID: 29094728 PMCID: PMC5888685 DOI: 10.1038/aps.2017.151] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 09/03/2017] [Indexed: 12/16/2022]
Abstract
The Nav1.7 channel represents a promising target for pain relief. In the recent decades, a number of Nav1.7 channel inhibitors have been developed. According to the effects on channel kinetics, these inhibitors could be divided into two major classes: reducing activation or enhancing inactivation. To date, however, only several inhibitors have moved forward into phase 2 clinical trials and most of them display a less than ideal analgesic efficacy, thus intensifying the controversy regarding if an ideal candidate should preferentially affect the activation or inactivation state. In the present study, we investigated the action mechanisms of a recently clinically confirmed inhibitor CNV1014802 using both electrophysiology and site-directed mutagenesis. We found that CNV1014802 inhibited Nav1.7 channels through stabilizing a nonconductive inactivated state. When the cells expressing Nav1.7 channels were hold at 70 mV or 120 mV, the half maximal inhibitory concentration (IC50) values (with 95% confidence limits) were 1.77 (1.20-2.33) and 71.66 (46.85-96.48) μmol/L, respectively. This drug caused dramatic hyperpolarizing shift of channel inactivation but did not affect activation. Moreover, CNV1014802 accelerated the onset of inactivation and delayed the recovery from inactivation. Notably, application of CNV1014802 (30 μmol/L) could rescue the Nav1.7 mutations expressed in CHO cells that cause paroxysmal extreme pain disorder (PEPD), thereby restoring the impaired inactivation to those of the wild-type channel. Our study demonstrates that CNV1014802 enhances the inactivation but does not reduce the activation of Nav1.7 channels, suggesting that identifying inhibitors that preferentially affect inactivation is a promising approach for developing drugs targeting Nav1.7.
Collapse
Affiliation(s)
- Yue-ming Zheng
- CAS Key Laboratory of Receptor Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Wan-fu Wang
- CAS Key Laboratory of Receptor Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Yan-fen Li
- CAS Key Laboratory of Receptor Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Shanghai Key Laboratory of Bio-Energy Crops, School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Yong Yu
- Department of Neurosurgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Zhao-bing Gao
- CAS Key Laboratory of Receptor Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| |
Collapse
|
45
|
Zaman T, Helbig I, Božović IB, DeBrosse SD, Bergqvist AC, Wallis K, Medne L, Maver A, Peterlin B, Helbig KL, Zhang X, Goldberg EM. Mutations in SCN3A cause early infantile epileptic encephalopathy. Ann Neurol 2018; 83:703-717. [PMID: 29466837 DOI: 10.1002/ana.25188] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 02/01/2018] [Accepted: 02/18/2018] [Indexed: 12/19/2022]
Abstract
OBJECTIVE Voltage-gated sodium (Na+ ) channels underlie action potential generation and propagation and hence are central to the regulation of excitability in the nervous system. Mutations in the genes SCN1A, SCN2A, and SCN8A, encoding the Na+ channel pore-forming (α) subunits Nav1.1, 1.2, and 1.6, respectively, and SCN1B, encoding the accessory subunit β1 , are established causes of genetic epilepsies. SCN3A, encoding Nav1.3, is known to be highly expressed in brain, but has not previously been linked to early infantile epileptic encephalopathy. Here, we describe a cohort of 4 patients with epileptic encephalopathy and heterozygous de novo missense variants in SCN3A (p.Ile875Thr in 2 cases, p.Pro1333Leu, and p.Val1769Ala). METHODS All patients presented with treatment-resistant epilepsy in the first year of life, severe to profound intellectual disability, and in 2 cases (both with the variant p.Ile875Thr), diffuse polymicrogyria. RESULTS Electrophysiological recordings of mutant channels revealed prominent gain of channel function, with a markedly increased amplitude of the slowly inactivating current component, and for 2 of 3 mutants (p.Ile875Thr and p.Pro1333Leu), a leftward shift in the voltage dependence of activation to more hyperpolarized potentials. Gain of function was not observed for Nav1.3 variants known or presumed to be inherited (p.Arg1642Cys and p.Lys1799Gln). The antiseizure medications phenytoin and lacosamide selectively blocked slowly inactivating over transient current in wild-type and mutant Nav1.3 channels. INTERPRETATION These findings establish SCN3A as a new gene for infantile epileptic encephalopathy and suggest a potential pharmacologic intervention. These findings also reinforce the role of Nav1.3 as an important regulator of neuronal excitability in the developing brain, while providing additional insight into mechanisms of slow inactivation of Nav1.3. Ann Neurol 2018;83:703-717.
Collapse
Affiliation(s)
- Tariq Zaman
- Division of Neurology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Ingo Helbig
- Division of Neurology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA.,Department of Neuropediatrics, University Medical Center Schleswig-Holstein, Christian Albrecht University, Kiel, Germany.,Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Ivana Babić Božović
- Department of Biology and Medical Genetics, School of Medicine, University of Rijeka, Rijeka, Croatia
| | - Suzanne D DeBrosse
- Departments of Genetics and Genome Sciences, Pediatrics, and Neurology, and Center for Human Genetics, Case Western Reserve University School of Medicine, Cleveland, OH
| | - A Christina Bergqvist
- Division of Neurology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Kimberly Wallis
- Departments of Genetics and Genome Sciences, Pediatrics, and Neurology, and Center for Human Genetics, Case Western Reserve University School of Medicine, Cleveland, OH
| | - Livija Medne
- Division of Human Genetics, Department of Pediatrics, Individualized Medical Genetics Center, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Aleš Maver
- Clinical Institute of Medical Genetics, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Borut Peterlin
- Clinical Institute of Medical Genetics, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Katherine L Helbig
- Division of Neurology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA.,Division of Clinical Genomics, Ambry Genetics, Aliso Viejo, CA
| | - Xiaohong Zhang
- Division of Neurology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Ethan M Goldberg
- Division of Neurology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA.,Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA.,Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
46
|
Jang IJ, Davies AJ, Akimoto N, Back SK, Lee PR, Na HS, Furue H, Jung SJ, Kim YH, Oh SB. Acute inflammation reveals GABA A receptor-mediated nociception in mouse dorsal root ganglion neurons via PGE 2 receptor 4 signaling. Physiol Rep 2018; 5:5/8/e13178. [PMID: 28438981 PMCID: PMC5408276 DOI: 10.14814/phy2.13178] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 01/30/2017] [Indexed: 12/29/2022] Open
Abstract
Gamma‐aminobutyric acid (GABA) depolarizes dorsal root ganglia (DRG) primary afferent neurons through activation of Cl− permeable GABAA receptors but the physiologic role of GABAA receptors in the peripheral terminals of DRG neurons remains unclear. In this study, we investigated the role of peripheral GABAA receptors in nociception using a mouse model of acute inflammation. In vivo, peripheral administration of the selective GABAA receptor agonist muscimol evoked spontaneous licking behavior, as well as spinal wide dynamic range (WDR) neuron firing, after pre‐conditioning with formalin but had no effect in saline‐treated mice. GABAA receptor‐mediated pain behavior after acute formalin treatment was abolished by the GABAA receptor blocker picrotoxin and cyclooxygenase inhibitor indomethacin. In addition, treatment with prostaglandin E2 (PGE2) was sufficient to reveal muscimol‐induced licking behavior. In vitro, GABA induced sub‐threshold depolarization in DRG neurons through GABAA receptor activation. Both formalin and PGE2 potentiated GABA‐induced Ca2+ transients and membrane depolarization in capsaicin‐sensitive nociceptive DRG neurons; these effects were blocked by the prostaglandin E2 receptor 4 (EP4) antagonist AH23848 (10 μmol/L). Furthermore, potentiation of GABA responses by PGE2 was prevented by the selective Nav1.8 antagonist A887826 (100 nmol/L). Although the function of the Na+‐K+‐2Cl‐ co‐transporter NKCC1 was required to maintain the Cl‐ ion gradient in isolated DRG neurons, NKCC1 was not required for GABAA receptor‐mediated nociceptive behavior after acute inflammation. Taken together, these results demonstrate that GABAA receptors may contribute to the excitation of peripheral sensory neurons in inflammation through a combined effect involving PGE2‐EP4 signaling and Na+ channel sensitization.
Collapse
Affiliation(s)
- In Jeong Jang
- Pain Laboratory, Dental Research Institute and Department of Neurobiology and Physiology School of Dentistry Seoul National University, Seoul, Korea
| | - Alexander J Davies
- Pain Laboratory, Dental Research Institute and Department of Neurobiology and Physiology School of Dentistry Seoul National University, Seoul, Korea.,Department of Brain and Cognitive Sciences, College of Natural Sciences Seoul National University, Seoul, Korea
| | - Nozomi Akimoto
- Department of Information Physiology, National Institute for Physiological Sciences, Okazaki, Japan
| | - Seung Keun Back
- Department of Physiology, Korea University College of Medicine, Seoul, Korea.,Department of Pharmacology and Biotechnology, College of Medical Engineering Konyang University, Daejeon, Korea
| | - Pa Reum Lee
- Pain Laboratory, Dental Research Institute and Department of Neurobiology and Physiology School of Dentistry Seoul National University, Seoul, Korea.,Department of Brain and Cognitive Sciences, College of Natural Sciences Seoul National University, Seoul, Korea
| | - Heung Sik Na
- Department of Physiology, Korea University College of Medicine, Seoul, Korea
| | - Hidemasa Furue
- Department of Information Physiology, National Institute for Physiological Sciences, Okazaki, Japan
| | - Sung Jun Jung
- Department of Physiology, Hanyang University, Seoul, Korea
| | - Yong Ho Kim
- Pain Laboratory, Dental Research Institute and Department of Neurobiology and Physiology School of Dentistry Seoul National University, Seoul, Korea
| | - Seog Bae Oh
- Pain Laboratory, Dental Research Institute and Department of Neurobiology and Physiology School of Dentistry Seoul National University, Seoul, Korea .,Department of Brain and Cognitive Sciences, College of Natural Sciences Seoul National University, Seoul, Korea
| |
Collapse
|
47
|
Xu Y, Meng X, Hou X, Sun J, Kong X, Sun Y, Liu Z, Ma Y, Niu Y, Song Y, Cui Y, Zhao M, Zhang J. A mutant of the Buthus martensii Karsch antitumor-analgesic peptide exhibits reduced inhibition to hNa v1.4 and hNa v1.5 channels while retaining analgesic activity. J Biol Chem 2017; 292:18270-18280. [PMID: 28924048 DOI: 10.1074/jbc.m117.792697] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 09/06/2017] [Indexed: 12/13/2022] Open
Abstract
Scorpion toxins can kill other animals by inducing paralysis and arrhythmia, which limits the potential applications of these agents in the clinical management of diseases. Antitumor-analgesic peptide (AGAP), purified from Buthus martensii Karsch, has been proved to possess analgesic and antitumor activities. Trp38, a conserved aromatic residue of AGAP, might play an important role in mediating AGAP activities according to the sequence and homology-modeling analyses. Therefore, an AGAP mutant, W38G, was generated, and effects of both AGAP and the mutant W38G were examined by whole-cell patch clamp techniques on the sodium channels hNav1.4 and hNav1.5, which were closely associated with the biotoxicity of skeletal and cardiac muscles, respectively. The data showed that both W38G and AGAP inhibited the peak currents of hNav1.4 and hNav1.5; however, W38G induced a much weaker inhibition of both channels than AGAP. Accordingly, W38G exhibited much less toxic effect on both skeletal and cardiac muscles than AGAP in vivo The analgesic activity of W38G and AGAP were verified in vivo as well, and W38G retained analgesic activity similar to AGAP. Inhibition to both Nav1.7 and Nav1.8 was involved in the analgesic mechanism of AGAP and W38G. These findings indicated that Trp38 was a key amino acid involved in the biotoxicity of AGAP, and the AGAP mutant W38G might be a safer alternative for clinical application because it retains the analgesic efficacy with less toxicity to skeletal and cardiac muscles.
Collapse
Affiliation(s)
- Yijia Xu
- From the School of Life Sciences and Biopharmaceutical Science, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016
| | - Xiangxue Meng
- From the School of Life Sciences and Biopharmaceutical Science, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016
| | - Xue Hou
- From the School of Life Sciences and Biopharmaceutical Science, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016
| | - Jianfang Sun
- the College of Life and Health Sciences, Northeastern University, Shenyang, Liaoning 110004, and
| | - Xiaohua Kong
- From the School of Life Sciences and Biopharmaceutical Science, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016
| | - Yuqi Sun
- From the School of Life Sciences and Biopharmaceutical Science, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016
| | - Zeyu Liu
- From the School of Life Sciences and Biopharmaceutical Science, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016
| | - Yuanyuan Ma
- From the School of Life Sciences and Biopharmaceutical Science, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016
| | - Ye Niu
- From the School of Life Sciences and Biopharmaceutical Science, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016
| | - Yongbo Song
- From the School of Life Sciences and Biopharmaceutical Science, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016
| | - Yong Cui
- the School of Medical Devices, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Mingyi Zhao
- From the School of Life Sciences and Biopharmaceutical Science, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016,
| | - Jinghai Zhang
- the School of Medical Devices, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| |
Collapse
|
48
|
Nemes AD, O'Dwyer R, Najm IM, Ying Z, Gonzalez-Martinez J, Alexopoulos AV. Treatment with lacosamide impedes generalized seizures in a rodent model of cortical dysplasia. Epilepsia 2017; 58:1755-1761. [PMID: 28833036 DOI: 10.1111/epi.13856] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/30/2017] [Indexed: 11/26/2022]
Abstract
OBJECTIVE Epilepsy is a common neurologic disorder resulting in spontaneous, recurrent seizures. About 30-40% of patients are not responsive to pharmacologic therapies. This may be due to the differences between individual patients such as etiology, underlying pathophysiology, and seizure focus, and it highlights the importance of new drug discovery and testing in this field. Our goal was to determine the efficacy of lacosamide (LCM), a drug approved for the treatment of focal seizures, in a model of generalized epilepsy with cortical dysplasia (CD). We sought to compare LCM to levetiracetam (LEV), a drug that is currently used for the treatment of both partial and generalized epilepsy and to test its proficiency. METHODS Pregnant rats were irradiated to produce pups with malformed cortices in a model of CD, which will be referred to as the "first hit." Adult animals, developed normally (NL) and irradiated (XRT), were surgically implanted with electroencephalography (EEG) electrodes. Baseline EEG was recorded on all rats prior to pretreatments with either LCM, LEV, or placebo (PBO). After 30 min, all rats were injected with a subconvulsive dose of pentylenetetrazole (PTZ), a γ-aminobutyric acid receptor A (GABAA ) antagonist used to provoke generalized seizures as a "second hit." RESULTS LCM and LEV were both effective against seizures induced by PTZ. XRT rats had a higher seizure incidence with longer and more severe seizures than NL rats. Seizure duration was decreased with both LCM and LEV in all animals. In XRT rats, there was a significant reduction in acute seizure incidence and severity with both LCM and LEV after PTZ injection. SIGNIFICANCE Our results suggest that LCM could be used as a potential treatment option for generalized epilepsy with CD as the underlying pathology.
Collapse
Affiliation(s)
- Ashley D Nemes
- Epilepsy Center, Neurological Institute, Cleveland Clinic, Cleveland, Ohio, U.S.A
| | - Rebecca O'Dwyer
- Department of Neurological Sciences, Rush Medical College, Chicago, Illinois, U.S.A
| | - Imad M Najm
- Epilepsy Center, Neurological Institute, Cleveland Clinic, Cleveland, Ohio, U.S.A
| | - Zhong Ying
- Epilepsy Center, Neurological Institute, Cleveland Clinic, Cleveland, Ohio, U.S.A
| | | | | |
Collapse
|
49
|
Pryde DC, Swain NA, Stupple PA, West CW, Marron B, Markworth CJ, Printzenhoff D, Lin Z, Cox PJ, Suzuki R, McMurray S, Waldron GJ, Payne CE, Warmus JS, Chapman ML. The discovery of a potent Na v1.3 inhibitor with good oral pharmacokinetics. MEDCHEMCOMM 2017; 8:1255-1267. [PMID: 30108836 DOI: 10.1039/c7md00131b] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 04/26/2017] [Indexed: 11/21/2022]
Abstract
In this article, we describe the discovery of an aryl ether series of potent and selective Nav1.3 inhibitors. Based on structural analogy to a similar series of compounds we have previously shown bind to the domain IV voltage sensor region of Nav channels, we propose this series binds in the same location. We describe the development of this series from a published starting point, highlighting key selectivity and potency data, and several studies designed to validate Nav1.3 as a target for pain.
Collapse
Affiliation(s)
- D C Pryde
- Worldwide Medicinal Chemistry , Pfizer Neuroscience and Pain Research Unit , Portway Building, Granta Park , Cambridge , CB21 6GS , UK .
| | - N A Swain
- Worldwide Medicinal Chemistry , Pfizer Neuroscience and Pain Research Unit , Portway Building, Granta Park , Cambridge , CB21 6GS , UK .
| | - P A Stupple
- Worldwide Medicinal Chemistry , Pfizer Neuroscience and Pain Research Unit , Portway Building, Granta Park , Cambridge , CB21 6GS , UK .
| | - C W West
- Pfizer Neuroscience and Pain Research Unit , 4222 Emperor Boulevard, Suite 350 , Durham , North Carolina NC27703 , USA
| | - B Marron
- Pfizer Neuroscience and Pain Research Unit , 4222 Emperor Boulevard, Suite 350 , Durham , North Carolina NC27703 , USA
| | - C J Markworth
- Pfizer Neuroscience and Pain Research Unit , 4222 Emperor Boulevard, Suite 350 , Durham , North Carolina NC27703 , USA
| | - D Printzenhoff
- Pfizer Neuroscience and Pain Research Unit , 4222 Emperor Boulevard, Suite 350 , Durham , North Carolina NC27703 , USA
| | - Z Lin
- Pfizer Neuroscience and Pain Research Unit , 4222 Emperor Boulevard, Suite 350 , Durham , North Carolina NC27703 , USA
| | - P J Cox
- Pfizer Neuroscience and Pain Research Unit , Portway Building, Granta Park , Cambridge , CB21 6GS , UK
| | - R Suzuki
- Pfizer Neuroscience and Pain Research Unit , Portway Building, Granta Park , Cambridge , CB21 6GS , UK
| | - S McMurray
- Pfizer Neuroscience and Pain Research Unit , Portway Building, Granta Park , Cambridge , CB21 6GS , UK
| | - G J Waldron
- Pfizer Neuroscience and Pain Research Unit , Portway Building, Granta Park , Cambridge , CB21 6GS , UK
| | - C E Payne
- Pfizer Neuroscience and Pain Research Unit , Portway Building, Granta Park , Cambridge , CB21 6GS , UK
| | - J S Warmus
- Worldwide Medicinal Chemistry , Pfizer Neuroscience and Pain Research Unit , Groton , CT , USA
| | - M L Chapman
- Pfizer Neuroscience and Pain Research Unit , 4222 Emperor Boulevard, Suite 350 , Durham , North Carolina NC27703 , USA
| |
Collapse
|
50
|
Jo S, Bean BP. Lacosamide Inhibition of Nav1.7 Voltage-Gated Sodium Channels: Slow Binding to Fast-Inactivated States. Mol Pharmacol 2017; 91:277-286. [PMID: 28119481 PMCID: PMC5363714 DOI: 10.1124/mol.116.106401] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 01/18/2017] [Indexed: 11/22/2022] Open
Abstract
Lacosamide is an antiseizure agent that targets voltage-dependent sodium channels. Previous experiments have suggested that lacosamide is unusual in binding selectively to the slow-inactivated state of sodium channels, in contrast to drugs like carbamazepine and phenytoin, which bind tightly to fast-inactivated states. Using heterologously expressed human Nav1.7 sodium channels, we examined the state-dependent effects of lacosamide. Lacosamide induced a reversible shift in the voltage dependence of fast inactivation studied with 100-millisecond prepulses, suggesting binding to fast-inactivated states. Using steady holding potentials, lacosamide block was very weak at -120 mV (3% inhibition by 100 µM lacosamide) but greatly enhanced at -80 mV (43% inhibition by 100 µM lacosamide), where there is partial fast inactivation but little or no slow inactivation. During long depolarizations, lacosamide slowly (over seconds) put channels into states that recovered availability slowly (hundreds of milliseconds) at -120 mV. This resembles enhancement of slow inactivation, but the effect was much more pronounced at -40 mV, where fast inactivation is complete, but slow inactivation is not, than at 0 mV, where slow inactivation is maximal, more consistent with slow binding to fast-inactivated states than selective binding to slow-inactivated states. Furthermore, inhibition by lacosamide was greatly reduced by pretreatment with 300 µM lidocaine or 300 µM carbamazepine, suggesting that lacosamide, lidocaine, and carbamazepine all bind to the same site. The results suggest that lacosamide binds to fast-inactivated states in a manner similar to other antiseizure agents but with slower kinetics of binding and unbinding.
Collapse
Affiliation(s)
- Sooyeon Jo
- Department of Neurobiology, Harvard Medical School, Boston Massachusetts
| | - Bruce P Bean
- Department of Neurobiology, Harvard Medical School, Boston Massachusetts
| |
Collapse
|