1
|
Li Z, Wu Q, Yan N. A structural atlas of druggable sites on Na v channels. Channels (Austin) 2024; 18:2287832. [PMID: 38033122 PMCID: PMC10732651 DOI: 10.1080/19336950.2023.2287832] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 11/20/2023] [Indexed: 12/02/2023] Open
Abstract
Voltage-gated sodium (Nav) channels govern membrane excitability by initiating and propagating action potentials. Consistent with their physiological significance, dysfunction, or mutations in these channels are associated with various channelopathies. Nav channels are thereby major targets for various clinical and investigational drugs. In addition, a large number of natural toxins, both small molecules and peptides, can bind to Nav channels and modulate their functions. Technological breakthrough in cryo-electron microscopy (cryo-EM) has enabled the determination of high-resolution structures of eukaryotic and eventually human Nav channels, alone or in complex with auxiliary subunits, toxins, and drugs. These studies have not only advanced our comprehension of channel architecture and working mechanisms but also afforded unprecedented clarity to the molecular basis for the binding and mechanism of action (MOA) of prototypical drugs and toxins. In this review, we will provide an overview of the recent advances in structural pharmacology of Nav channels, encompassing the structural map for ligand binding on Nav channels. These findings have established a vital groundwork for future drug development.
Collapse
Affiliation(s)
- Zhangqiang Li
- Beijing Frontier Research Center for Biological Structures, State Key Laboratory of Membrane Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Qiurong Wu
- Beijing Frontier Research Center for Biological Structures, State Key Laboratory of Membrane Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Nieng Yan
- Beijing Frontier Research Center for Biological Structures, State Key Laboratory of Membrane Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
- Shenzhen Medical Academy of Research and Translation, Shenzhen, Guangdong Province, China
| |
Collapse
|
2
|
Li Z, Luo J, Li C, Zhu H. Upregulation of Nav1.6 expression in the ventral posterolateral nucleus of thalamus contributes to hyperalgesia in a model of Parkinson's disease. Exp Neurol 2024; 383:115032. [PMID: 39490625 DOI: 10.1016/j.expneurol.2024.115032] [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: 08/15/2024] [Revised: 10/23/2024] [Accepted: 10/24/2024] [Indexed: 11/05/2024]
Abstract
Pain is the most common non-motor manifestation of Parkinson's disease (PD), affecting the quality of life for patients. Nav1.6 is the most abundant subtype of voltage-gated sodium channels (VGSCs) in the brain of adult mammals. Here we investigated the expression patterns of Nav1.6 in the ventral posterolateral (VPL) nucleus of the thalamus and its involvement in the development of hyperalgesia in 6-hydroxydopamine (6-OHDA)-lesioned rats. The results showed a significant increase in Nav1.6 expression in reactive astrocytes of the ipsilateral VPL in 6-OHDA-lesioned rats at 4 weeks post-injection. Moreover, 6-OHDA-lesioned rats exhibited mechanical hyperalgesia, but did not display thermal hyperalgesia in the ipsilateral paw at the same time point. The down-regulation of Nav1.6 in the ipsilateral VPL can reduce mechanical hyperalgesia and improve sensorimotor impairments in 6-OHDA- lesioned rats. Furthermore, the analysis of local field potentials (LFPs) revealed that the increased Nav1.6 may participate in abnormal synchronized oscillations within the thalamocortical loop in 6-OHDA-lesioned rats. These findings suggest that the altered expression of Nav1.6 in astrocytes of the VPL may play an important role in the abnormal processing of pain within the thalamocortical circuit, contributing to the formation of mechanical hyperalgesia in animal models of PD.
Collapse
Affiliation(s)
- Zhiwei Li
- School of Life Science, Shanghai University, China
| | - Jiamin Luo
- School of Life Science, Shanghai University, China
| | | | - Hongyan Zhu
- School of Life Science, Shanghai University, China.
| |
Collapse
|
3
|
Martina M, Banderali U, Yogi A, Arbabi Ghahroudi M, Liu H, Sulea T, Durocher Y, Hussack G, van Faassen H, Chakravarty B, Liu QY, Iqbal U, Ling B, Lessard E, Sheff J, Robotham A, Callaghan D, Moreno M, Comas T, Ly D, Stanimirovic D. A Novel Antigen Design Strategy to Isolate Single-Domain Antibodies that Target Human Nav1.7 and Reduce Pain in Animal Models. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2405432. [PMID: 39206821 PMCID: PMC11516162 DOI: 10.1002/advs.202405432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 08/01/2024] [Indexed: 09/04/2024]
Abstract
Genetic studies have identified the voltage-gated sodium channel 1.7 (Nav1.7) as pain target. Due to the ineffectiveness of small molecules and monoclonal antibodies as therapeutics for pain, single-domain antibodies (VHHs) are developed against the human Nav1.7 (hNav1.7) using a novel antigen presentation strategy. A 70 amino-acid peptide from the hNav1.7 protein is identified as a target antigen. A recombinant version of this peptide is grafted into the complementarity determining region 3 (CDR3) loop of an inert VHH in order to maintain the native 3D conformation of the peptide. This antigen is used to isolate one VHH able to i) bind hNav1.7, ii) slow the deactivation of hNav1.7, iii) reduce the ability of eliciting action potentials in nociceptors, and iv) reverse hyperalgesia in in vivo rat and mouse models. This VHH exhibits the potential to be developed as a therapeutic capable of suppressing pain. This novel antigen presentation strategy can be applied to develop biologics against other difficult targets such as ion channels, transporters and GPCRs.
Collapse
Affiliation(s)
- Marzia Martina
- Human Health Therapeutics Research CenterNational Research Council Canada1200 Montreal Road, Building M54OttawaONK1A 0R6Canada
| | - Umberto Banderali
- Human Health Therapeutics Research CenterNational Research Council Canada1200 Montreal Road, Building M54OttawaONK1A 0R6Canada
| | - Alvaro Yogi
- Human Health Therapeutics Research CenterNational Research Council Canada1200 Montreal Road, Building M54OttawaONK1A 0R6Canada
| | - Mehdi Arbabi Ghahroudi
- Human Health Therapeutics Research CentreNational Research Council Canada100 Sussex DriveOttawaONK1N 5A2Canada
| | - Hong Liu
- Human Health Therapeutics Research CenterNational Research Council Canada1200 Montreal Road, Building M54OttawaONK1A 0R6Canada
| | - Traian Sulea
- Human Health Therapeutics Research CentreNational Research Council Canada6100 Royalmount Avenue MontréalQuebecH4P 2R2Canada
| | - Yves Durocher
- Human Health Therapeutics Research CentreNational Research Council Canada6100 Royalmount Avenue MontréalQuebecH4P 2R2Canada
| | - Greg Hussack
- Human Health Therapeutics Research CentreNational Research Council Canada100 Sussex DriveOttawaONK1N 5A2Canada
| | - Henk van Faassen
- Human Health Therapeutics Research CentreNational Research Council Canada100 Sussex DriveOttawaONK1N 5A2Canada
| | - Balu Chakravarty
- Human Health Therapeutics Research CenterNational Research Council Canada1200 Montreal Road, Building M54OttawaONK1A 0R6Canada
| | - Qing Yan Liu
- Human Health Therapeutics Research CenterNational Research Council Canada1200 Montreal Road, Building M54OttawaONK1A 0R6Canada
| | - Umar Iqbal
- Human Health Therapeutics Research CenterNational Research Council Canada1200 Montreal Road, Building M54OttawaONK1A 0R6Canada
| | - Binbing Ling
- Human Health Therapeutics Research CenterNational Research Council Canada1200 Montreal Road, Building M54OttawaONK1A 0R6Canada
| | - Etienne Lessard
- Human Health Therapeutics Research CentreNational Research Council Canada6100 Royalmount Avenue MontréalQuebecH4P 2R2Canada
| | - Joey Sheff
- Human Health Therapeutics Research CentreNational Research Council Canada100 Sussex DriveOttawaONK1N 5A2Canada
| | - Anna Robotham
- Human Health Therapeutics Research CentreNational Research Council Canada100 Sussex DriveOttawaONK1N 5A2Canada
| | - Debbie Callaghan
- Human Health Therapeutics Research CentreNational Research Council Canada100 Sussex DriveOttawaONK1N 5A2Canada
| | - Maria Moreno
- Human Health Therapeutics Research CenterNational Research Council Canada1200 Montreal Road, Building M54OttawaONK1A 0R6Canada
| | - Tanya Comas
- Human Health Therapeutics Research CenterNational Research Council Canada1200 Montreal Road, Building M54OttawaONK1A 0R6Canada
| | - Dao Ly
- Human Health Therapeutics Research CenterNational Research Council Canada1200 Montreal Road, Building M54OttawaONK1A 0R6Canada
| | - Danica Stanimirovic
- Human Health Therapeutics Research CenterNational Research Council Canada1200 Montreal Road, Building M54OttawaONK1A 0R6Canada
| |
Collapse
|
4
|
Li Z, Wu Q, Huang G, Jin X, Li J, Pan X, Yan N. Dissection of the structure-function relationship of Na v channels. Proc Natl Acad Sci U S A 2024; 121:e2322899121. [PMID: 38381792 PMCID: PMC10907234 DOI: 10.1073/pnas.2322899121] [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: 12/28/2023] [Accepted: 01/17/2024] [Indexed: 02/23/2024] Open
Abstract
Voltage-gated sodium channels (Nav) undergo conformational shifts in response to membrane potential changes, a mechanism known as the electromechanical coupling. To delineate the structure-function relationship of human Nav channels, we have performed systematic structural analysis using human Nav1.7 as a prototype. Guided by the structural differences between wild-type (WT) Nav1.7 and an eleven mutation-containing variant, designated Nav1.7-M11, we generated three additional intermediate mutants and solved their structures at overall resolutions of 2.9-3.4 Å. The mutant with nine-point mutations in the pore domain (PD), named Nav1.7-M9, has a reduced cavity volume and a sealed gate, with all voltage-sensing domains (VSDs) remaining up. Structural comparison of WT and Nav1.7-M9 pinpoints two residues that may be critical to the tightening of the PD. However, the variant containing these two mutations, Nav1.7-M2, or even in combination with two additional mutations in the VSDs, named Nav1.7-M4, failed to tighten the PD. Our structural analysis reveals a tendency of PD contraction correlated with the right shift of the static inactivation I-V curves. We predict that the channel in the resting state should have a "tight" PD with down VSDs.
Collapse
Affiliation(s)
- Zhangqiang Li
- Beijing Frontier Research Center for Biological Structures, State Key Laboratory of Membrane Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing100084, China
| | - Qiurong Wu
- Beijing Frontier Research Center for Biological Structures, State Key Laboratory of Membrane Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing100084, China
| | - Gaoxingyu Huang
- Westlake Laboratory of Life Sciences and Biomedicine, Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou310024, China
| | - Xueqin Jin
- Beijing Frontier Research Center for Biological Structures, State Key Laboratory of Membrane Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing100084, China
| | - Jiaao Li
- Beijing Frontier Research Center for Biological Structures, State Key Laboratory of Membrane Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing100084, China
| | - Xiaojing Pan
- Beijing Frontier Research Center for Biological Structures, State Key Laboratory of Membrane Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing100084, China
- Institute of Bio-Architecture and Bio-Interactions, Shenzhen Medical Academy of Research and Translation, Shenzhen518107, China
| | - Nieng Yan
- Beijing Frontier Research Center for Biological Structures, State Key Laboratory of Membrane Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing100084, China
- Institute of Bio-Architecture and Bio-Interactions, Shenzhen Medical Academy of Research and Translation, Shenzhen518107, China
| |
Collapse
|
5
|
Ding K, Gong Y, Cheng C, Li X, Zhu Y, Gao X, Li Y, Yuan C, Liu Z, Jiang W, Chen C, Yao LH. Expression and electrophysiological characteristics of VGSC during mouse myoblasts differentiation. Cell Signal 2024; 113:110970. [PMID: 37967692 DOI: 10.1016/j.cellsig.2023.110970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 10/31/2023] [Accepted: 11/12/2023] [Indexed: 11/17/2023]
Abstract
Voltage-gated sodium channels (VGSC) are essential for triggering and relaying action potentials (AP), which perform critical functions in a variety of physiological processes, such as controlling muscle contractions and facilitating the release of neurotransmitters. In this study, we used a mouse C2C12 cell differentiation model to study the molecular expression and channel dynamics of VGSC and to investigate the exact role of VGSC in the development of muscle regeneration. Immunofluorescence, Real-time quantitative polymerase chain reaction, Western blot, and whole-cell patch clamp were employed for this purpose in mouse myoblasts. The findings revealed an increase in intracellular sodium concentration, NaV1.4 gene expression, and protein expression with the progress of differentiation (days 0, 1, 3, 5 and 7). Furthermore, VGSC dynamics exhibit the following characteristics: ① The increase of sodium current (INa); ② The decrease in the activation threshold and the voltage trigger maximum of INa; ③ A positive shift in the steady-state inactivation curve; ④ The recovery of INa during repolarization is delayed, the activity-dependent decay rate of INa was accelerated, and the proportionate amount of the fraction of activated channels was reduced. Based on these results, it is postulated that the activation threshold of AP could be decreased, and the refractory period could be extended with the extension of differentiation duration, which may contribute to muscle contraction. Taken together, VGSC provides a theoretical and empirical basis for exploring potential targets for neuromuscular diseases and other therapeutic muscle regeneration dysfunctions.
Collapse
Affiliation(s)
- Kaizhi Ding
- School of Life Science, Jiangxi Science & Technology Normal University, Nanchang, Jiangxi 330013, PR China
| | - Yanchun Gong
- School of Life Science, Jiangxi Science & Technology Normal University, Nanchang, Jiangxi 330013, PR China
| | - Chunfang Cheng
- School of Life Science, Jiangxi Science & Technology Normal University, Nanchang, Jiangxi 330013, PR China; School of Physical Education and Health, Jiangxi Science & Technology Normal University, Nanchang, Jiangxi 330013, PR China
| | - Xiaonuo Li
- School of Life Science, Jiangxi Science & Technology Normal University, Nanchang, Jiangxi 330013, PR China
| | - Yuanjie Zhu
- School of Life Science, Jiangxi Science & Technology Normal University, Nanchang, Jiangxi 330013, PR China
| | - Xiaofei Gao
- School of Life Science, Jiangxi Science & Technology Normal University, Nanchang, Jiangxi 330013, PR China
| | - Yuhua Li
- School of Life Science, Jiangxi Science & Technology Normal University, Nanchang, Jiangxi 330013, PR China
| | - Chunhua Yuan
- School of Physical Education and Health, Jiangxi Science & Technology Normal University, Nanchang, Jiangxi 330013, PR China
| | - Zhibing Liu
- School of Physical Education and Health, Jiangxi Science & Technology Normal University, Nanchang, Jiangxi 330013, PR China
| | - Wei Jiang
- Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi 330006, PR China
| | - Chong Chen
- School of Life Science, Jiangxi Science & Technology Normal University, Nanchang, Jiangxi 330013, PR China; School of Physical Education and Health, Jiangxi Science & Technology Normal University, Nanchang, Jiangxi 330013, PR China; NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, PR China.
| | - Li-Hua Yao
- School of Life Science, Jiangxi Science & Technology Normal University, Nanchang, Jiangxi 330013, PR China; School of Physical Education and Health, Jiangxi Science & Technology Normal University, Nanchang, Jiangxi 330013, PR China.
| |
Collapse
|
6
|
Thompson AC, Aizenman CD. Characterization of Na + currents regulating intrinsic excitability of optic tectal neurons. Life Sci Alliance 2024; 7:e202302232. [PMID: 37918964 PMCID: PMC10622587 DOI: 10.26508/lsa.202302232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 10/20/2023] [Accepted: 10/23/2023] [Indexed: 11/04/2023] Open
Abstract
Developing neurons adapt their intrinsic excitability to maintain stable output despite changing synaptic input. The mechanisms behind this process remain unclear. In this study, we examined Xenopus optic tectal neurons and found that the expressions of Nav1.1 and Nav1.6 voltage-gated Na+ channels are regulated during changes in intrinsic excitability, both during development and becsuse of changes in visual experience. Using whole-cell electrophysiology, we demonstrate the existence of distinct, fast, persistent, and resurgent Na+ currents in the tectum, and show that these Na+ currents are co-regulated with changes in Nav channel expression. Using antisense RNA to suppress the expression of specific Nav subunits, we found that up-regulation of Nav1.6 expression, but not Nav1.1, was necessary for experience-dependent increases in Na+ currents and intrinsic excitability. Furthermore, this regulation was also necessary for normal development of sensory guided behaviors. These data suggest that the regulation of Na+ currents through the modulation of Nav1.6 expression, and to a lesser extent Nav1.1, plays a crucial role in controlling the intrinsic excitability of tectal neurons and guiding normal development of the tectal circuitry.
Collapse
Affiliation(s)
- Adrian C Thompson
- https://ror.org/05gq02987 Department of Neuroscience, Brown University, Providence, RI, USA
| | - Carlos D Aizenman
- https://ror.org/05gq02987 Department of Neuroscience, Brown University, Providence, RI, USA
| |
Collapse
|
7
|
Liu Y, Xia D, Zhong L, Chen L, Zhang L, Ai M, Mei R, Pang R. Casein Kinase 2 Affects Epilepsy by Regulating Ion Channels: A Potential Mechanism. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:894-905. [PMID: 37350003 DOI: 10.2174/1871527322666230622124618] [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: 08/12/2022] [Revised: 03/31/2023] [Accepted: 04/10/2023] [Indexed: 06/24/2023]
Abstract
Epilepsy, characterized by recurrent seizures and abnormal brain discharges, is the third most common chronic disorder of the Central Nervous System (CNS). Although significant progress has been made in the research on antiepileptic drugs (AEDs), approximately one-third of patients with epilepsy are refractory to these drugs. Thus, research on the pathogenesis of epilepsy is ongoing to find more effective treatments. Many pathological mechanisms are involved in epilepsy, including neuronal apoptosis, mossy fiber sprouting, neuroinflammation, and dysfunction of neuronal ion channels, leading to abnormal neuronal excitatory networks in the brain. CK2 (Casein kinase 2), which plays a critical role in modulating neuronal excitability and synaptic transmission, has been shown to be associated with epilepsy. However, there is limited research on the mechanisms involved. Recent studies have suggested that CK2 is involved in regulating the function of neuronal ion channels by directly phosphorylating them or their binding partners. Therefore, in this review, we will summarize recent research advances regarding the potential role of CK2 regulating ion channels in epilepsy, aiming to provide more evidence for future studies.
Collapse
Affiliation(s)
- Yan Liu
- Department of Neurology, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
| | - Di Xia
- Department of Neurology, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
| | - Lianmei Zhong
- Department of Neurology, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
| | - Ling Chen
- Department of Neurology, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
- Yunnan Provincial Clinical Research Center for Neurological Disease, Kunming, Yunnan, 650032, China
| | - Linming Zhang
- Department of Neurology, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
| | - Mingda Ai
- Department of Neurology, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
| | - Rong Mei
- Department of Neurology, the First People's Hospital of Yunnan Province, Kunming, Yunnan, 650034, China
| | - Ruijing Pang
- Department of Neurology, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
| |
Collapse
|
8
|
Banderali U, Moreno M, Martina M. The elusive Na v1.7: From pain to cancer. CURRENT TOPICS IN MEMBRANES 2023; 92:47-69. [PMID: 38007269 DOI: 10.1016/bs.ctm.2023.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2023]
Abstract
Voltage-gated sodium channels (Nav) are protein complexes that play fundamental roles in the transmission of signals in the nervous system, at the neuromuscular junction and in the heart. They are mainly present in excitable cells where they are responsible for triggering action potentials. Dysfunctions in Nav ion conduction give rise to a wide range of conditions, including neurological disorders, hypertension, arrhythmia, pain and cancer. Nav family 1 is composed of nine members, named numerically from 1 to 9. A Nax family also exists and is involved in body-fluid homeostasis. Of particular interest is Nav1.7 which is highly expressed in the sensory neurons of the dorsal root ganglions, where it is involved in the propagation of pain sensation. Gain-of-function mutations in Nav1.7 cause pathologies associated with increased pain sensitivity, while loss-of-function mutations cause reduced sensitivity to pain. The last decade has seen considerable effort in developing highly specific Nav1.7 blockers as pain medications, nonetheless, sufficient efficacy has yet to be achieved. Evidence is now conclusively showing that Navs are also present in many types of cancer cells, where they are involved in cell migration and invasiveness. Nav1.7 is anomalously expressed in endometrial, ovarian and lung cancers. Nav1.7 is also involved in Chemotherapy Induced Peripheral Neuropathy (CIPN). We propose that the knowledge and tools developed to study the role of Nav1.7 in pain can be exploited to develop novel cancer therapies. In this chapter, we illustrate the various aspects of Nav1.7 function in pain, cancer and CIPN, and outline therapeutic approaches.
Collapse
Affiliation(s)
- Umberto Banderali
- Human Health Therapeutics Research Centre, National Research Council of Canada, Montreal road, Ottawa, ON, Canada.
| | - Maria Moreno
- Human Health Therapeutics Research Centre, National Research Council of Canada, Montreal road, Ottawa, ON, Canada
| | - Marzia Martina
- Human Health Therapeutics Research Centre, National Research Council of Canada, Montreal road, Ottawa, ON, Canada
| |
Collapse
|
9
|
Brito A, Pinto M, Moreira Z, Lages N. Resistance to local anesthetics: Truth or myth? REVISTA ESPANOLA DE ANESTESIOLOGIA Y REANIMACION 2023; 70:297-299. [PMID: 36934844 DOI: 10.1016/j.redare.2022.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Accepted: 07/11/2022] [Indexed: 03/19/2023]
Abstract
The report of anesthetic technique failure is crucial and the etiology of the problem should be determined. We describe a case of locoregional anesthesia failure, in which, after excluding its most common causes, the resistance to local anesthetics was considered as the most probable clinical hypothesis. For this reason, a genetic test was performed, as well as the efficacy of other local anesthetics was evaluated, constituting a different approach in the cases of locoregional anesthesia failure. True resistance to local anesthetics is difficult to diagnose so information about this is scarce in the literature. One of the proposed causes is a mutation of sodium channels where local anesthetics bind. If not recognized, the application of locorregional anesthesia in this patient's condition can lead to unpleasant experiences and unnecessary risks, related to toxic levels of local anesthetics. For this reason, the resistance to local anesthetics should be always precluded in cases of strong clinical suspicion. This approach could be applied in similar cases.
Collapse
Affiliation(s)
- A Brito
- Anestesiología, Centro Hospitalar Universitário do Porto, Porto, Portugal.
| | - M Pinto
- Anestesiología, Centro Hospitalar Universitário do Porto, Porto, Portugal
| | - Z Moreira
- Anestesiología, Centro Hospitalar Universitário do Porto, Porto, Portugal
| | - N Lages
- Anestesiología, Centro Hospitalar Universitário do Porto, Porto, Portugal
| |
Collapse
|
10
|
Leslie TK, Brackenbury WJ. Sodium channels and the ionic microenvironment of breast tumours. J Physiol 2023; 601:1543-1553. [PMID: 36183245 PMCID: PMC10953337 DOI: 10.1113/jp282306] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 08/11/2022] [Indexed: 11/08/2022] Open
Abstract
Cancers of epithelial origin such as breast, prostate, cervical, gastric, colon and lung cancer account for a large proportion of deaths worldwide. Better treatment of metastasis, the main cause of cancer deaths, is therefore urgently required. Several of these tumours have been shown to have an abnormally high concentration of Na+ ([Na+ ]) and emerging evidence points to this accumulation being due to elevated intracellular [Na+ ]. This poses intriguing questions about the cellular mechanisms underlying Na+ dysregulation in cancer, and its pathophysiological significance. Elevated intracellular [Na+ ] may be due to alterations in activity of the Na+ /K+ -ATPase, and/or increased influx via Na+ channels and Na+ -linked transporters. Maintenance of the electrochemical Na+ gradient across the plasma membrane is vital to power many cellular processes that are highly active in cancer cells, including glucose and glutamine import. Na+ channels are also upregulated in cancer cells, which in turn promotes tumour growth and metastasis. For example, ENaC and ASICs are overexpressed in cancers, increasing invasion and proliferation. In addition, voltage-gated Na+ channels are also upregulated in a range of tumour types, where they promote metastatic cell behaviours via various mechanisms, including membrane potential depolarisation and altered pH regulation. Together, recent findings relating to elevated Na+ in the tumour microenvironment and how this may be regulated by several classes of Na+ channels provide a link between altered Na+ handling and poor clinical outcome. There are new opportunities to leverage this altered Na+ microenvironment for therapeutic benefit, as exemplified by several ongoing clinical trials.
Collapse
Affiliation(s)
- Theresa K. Leslie
- Department of BiologyUniversity of YorkHeslingtonYorkUK
- York Biomedical Research InstituteUniversity of YorkHeslingtonYorkUK
| | - William J. Brackenbury
- Department of BiologyUniversity of YorkHeslingtonYorkUK
- York Biomedical Research InstituteUniversity of YorkHeslingtonYorkUK
| |
Collapse
|
11
|
Oz M, Lorke DE, Howarth FC. Transient receptor potential vanilloid 1 (TRPV1)-independent actions of capsaicin on cellular excitability and ion transport. Med Res Rev 2023. [PMID: 36916676 DOI: 10.1002/med.21945] [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: 06/14/2022] [Revised: 01/17/2023] [Accepted: 02/26/2023] [Indexed: 03/15/2023]
Abstract
Capsaicin is a naturally occurring alkaloid derived from chili pepper that is responsible for its hot pungent taste. Capsaicin is known to exert multiple pharmacological actions, including analgesia, anticancer, anti-inflammatory, antiobesity, and antioxidant effects. The transient receptor potential vanilloid subfamily member 1 (TRPV1) is the main receptor mediating the majority of the capsaicin effects. However, numerous studies suggest that the TRPV1 receptor is not the only target for capsaicin. An increasing number of studies indicates that capsaicin, at low to mid µM ranges, not only indirectly through TRPV1-mediated Ca2+ increases, but also directly modulates the functions of voltage-gated Na+ , K+ , and Ca2+ channels, as well as ligand-gated ion channels and other ion transporters and enzymes involved in cellular excitability. These TRPV1-independent effects are mediated by alterations of the biophysical properties of the lipid membrane and subsequent modulation of the functional properties of ion channels and by direct binding of capsaicin to the channels. The present study, for the first time, systematically categorizes this diverse range of non-TRPV1 targets and discusses cellular and molecular mechanisms mediating TRPV1-independent effects of capsaicin in excitable, as well as nonexcitable cells.
Collapse
Affiliation(s)
- Murat Oz
- Department of Pharmacology and Therapeutics, Faculty of Pharmacy, Kuwait University, Safat, Kuwait
| | - Dietrich E Lorke
- Department of Anatomy and Cellular Biology, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, United Arab Emirates.,Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Frank C Howarth
- Department of Physiology, College of Medicine and Health Sciences, UAE University, Al Ain, United Arab Emirates
| |
Collapse
|
12
|
Bigiani A, Tirindelli R, Bigiani L, Mapelli J. Changes of the biophysical properties of voltage-gated Na + currents during maturation of the sodium-taste cells in rat fungiform papillae. J Physiol 2022; 600:5119-5144. [PMID: 36250254 DOI: 10.1113/jp283636] [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: 07/22/2022] [Accepted: 10/13/2022] [Indexed: 01/05/2023] Open
Abstract
Taste cells are a heterogeneous population of sensory receptors that undergo continuous turnover. Different chemo-sensitive cell lines rely on action potentials to release the neurotransmitter onto nerve endings. The electrical excitability is due to the presence of a tetrodotoxin-sensitive, voltage-gated sodium current (INa ) similar to that found in neurons. Since the biophysical properties of neuronal INa change during development, we wondered whether the same also occurred in taste cells. Here, we used the patch-clamp recording technique to study INa in salt-sensing cells (sodium cells) of rat fungiform papillae. We identified these cells by exploiting the known blocking effect of amiloride on ENaC, the sodium (salt) receptor. Based on the amplitude of INa , which is known to increase during development, we subdivided sodium cells into two groups: cells with small sodium current (SSC cells; INa < 1 nA) and cells with large sodium current (LSC cells; INa > 1 nA). We found that: the voltage dependence of activation and inactivation significantly differed between these subsets; a slowly inactivating sodium current was more prominent in LSC cells; membrane capacitance in SSC cells was larger than in LSC cells. mRNA expression analysis of the α-subunits of voltage-gated sodium channels in fungiform taste buds supported the functional data. Lucifer Yellow labelling of recorded cells revealed that our electrophysiological criterion for distinguishing two broad groups of taste cells was in good agreement with morphological observations for cell maturity. Thus, all these findings are consistent with developmental changes in the voltage-dependent properties of sodium-taste cells. KEY POINTS: Taste cells are sensory receptors that undergo continuous turnover while they detect food chemicals and communicate with afferent nerve fibres. The voltage-gated sodium current (INa ) is a key ion current for generating action potentials in fully differentiated and chemo-sensitive taste cells, which use electrical signalling to release neurotransmitters. Here we show that, during the maturation of rat taste cells involved in salt detection (sodium cells), the biophysical properties of INa , such as voltage dependence of activation and inactivation, change significantly. Our results help reveal how taste cells gain electrical excitability during turnover, a property critical to their operation as chemical detectors that relay sensory information to nerve fibres.
Collapse
Affiliation(s)
- Albertino Bigiani
- Dipartimento di Scienze Biomediche, Metaboliche e Neuroscienze, Università di Modena e Reggio Emilia, Modena, Italy
| | - Roberto Tirindelli
- Dipartimento di Medicina e Chirurgia, SMart Laboratory, Università di Parma, Parma, Italy
| | | | - Jonathan Mapelli
- Dipartimento di Scienze Biomediche, Metaboliche e Neuroscienze, Università di Modena e Reggio Emilia, Modena, Italy
| |
Collapse
|
13
|
Fahy JF, Emerling EW, Sterni LM. Perioperative Management and Considerations for Patients With Voltage-Gated Sodium Channel Mutations: A Pediatric Case Report. A A Pract 2022; 16:e01637. [PMID: 36599048 DOI: 10.1213/xaa.0000000000001637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
A 13-year-old girl with a voltage-gated sodium channel mutation (SCN8A)-associated intractable epilepsy presented for bilateral mastectomy for painful juvenile fibroadenomatosis. Sodium channel mutations are more frequently diagnosed with continued advances in genetic testing. Understanding the effects of sodium channel mutations is important to provide safe anesthetic care to these patients. In this article, we discuss what is known regarding the physiology of SCN8A channels and the anesthetic considerations when caring for patients with an SCN8A mutation.
Collapse
Affiliation(s)
- John F Fahy
- From the Department of Anesthesiology, Naval Medical Center San Diego, San Diego, California
| | | | | |
Collapse
|
14
|
Kelkar S, Nailwal N, Bhatia NY, Doshi G, Sathaye S, Godad AP. An Update On Proficiency of Voltage-gated Ion Channel Blockers in the Treatment of Inflammation-associated Diseases. Curr Drug Targets 2022; 23:1290-1303. [PMID: 35996239 DOI: 10.2174/1389450123666220819141827] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 05/24/2022] [Accepted: 06/21/2022] [Indexed: 01/25/2023]
Abstract
Inflammation is the body's mechanism to trigger the immune system, thereby preventing bacteria and viruses from manifesting their toxic effect. Inflammation plays a vital role in regulating inflammatory mediator levels to initiate the wound healing process depending on the nature of the stimuli. This process occurs due to chemical release from white blood cells by elevating blood flow to the site of action, leading to redness and increased body temperature. Currently, there are numerous Non-steroidal anti-inflammatory drugs (NSAIDs) available, but these drugs are reported with adverse effects such as gastric bleeding, progressive kidney damage, and increased risk of heart attacks when prolonged use. For such instances, alternative options need to be adopted. The introduction of voltage-gated ion channel blockers can be a substantial alternative to mask the side effects of these currently available drugs. Chronic inflammatory disorders such as rheumatoid and osteoarthritis, cancer and migraine, etc., can cause dreadful pain, which is often debilitating for the patient. The underlying mechanism for both acute and chronic inflammation involves various complex receptors, different types of cells, receptors, and proteins. The working of voltage-gated sodium and calcium channels is closely linked to both inflammatory and neuropathic pain. Certain drugs such as carbamazepine and gabapentin, which are ion channel blockers, have greater pharmacotherapeutic activity for sodium and calcium channel blockers for the treatment of chronic inflammatory pain states. This review intends to provide brief information on the mechanism of action, latest clinical trials, and applications of these blockers in treating inflammatory conditions.
Collapse
Affiliation(s)
- Siddesh Kelkar
- MET Institute of Pharmacy, Bhujbal Knowledge City, Reclamation, Bandra West, Mumbai, Maharashtra 400050, India
| | - Namrata Nailwal
- SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mithibai College Campus, Vaikunthlal Mehta Rd, Vile Parle West, Mumbai, Maharashtra 400056, India
| | - Nirav Yogesh Bhatia
- SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mithibai College Campus, Vaikunthlal Mehta Rd, Vile Parle West, Mumbai, Maharashtra 400056, India
| | - Gaurav Doshi
- SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mithibai College Campus, Vaikunthlal Mehta Rd, Vile Parle West, Mumbai, Maharashtra 400056, India
| | - Sadhana Sathaye
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai, India
| | - Angel Pavalu Godad
- SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mithibai College Campus, Vaikunthlal Mehta Rd, Vile Parle West, Mumbai, Maharashtra 400056, India.,Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai, India
| |
Collapse
|
15
|
Ion Channels in Endometrial Cancer. Cancers (Basel) 2022; 14:cancers14194733. [PMID: 36230654 PMCID: PMC9564232 DOI: 10.3390/cancers14194733] [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: 08/19/2022] [Revised: 09/22/2022] [Accepted: 09/23/2022] [Indexed: 11/17/2022] Open
Abstract
Simple Summary Uterine or endometrial cancer is one of the most common types of cancer among the female population. Different alterations of molecules are related to many types of cancer. Some molecules called ion channels have been described as involved in the development of cancer, including endometrial cancer. We review the scientific evidence about the involvement of the ion channels in endometrial cancer and how some treatments can be developed with these molecules as a target. Even though they are involved in the progression of endometrial cancer, since they are present throughout the whole body, some possible treatments based on these could be studied. Abstract Uterine or endometrial cancer (EC) is the sixth most common neoplasia among women worldwide. Cancer can originate from a myriad of causes, and increasing evidence suggests that ion channels (IC) play an important role in the process of carcinogenesis, taking part in many pathways such as self-sufficiency in growth signals, proliferation, evasion of programmed cell death (apoptosis), angiogenesis, cell differentiation, migration, adhesion, and metastasis. Hormones and growth factors are well-known to be involved in the development and/or progression of many cancers and can also regulate some ion channels and pumps. Since the endometrium is responsive and regulated by these factors, the ICs could make an important contribution to the development and progression of endometrial cancer. In this review, we explore what is beyond (ion) flow regulation by investigating the role of the main families of ICs in EC, including as possible targets for EC treatment.
Collapse
|
16
|
Gessner G, Jamili M, Tomczyk P, Menche D, Schönherr R, Hoshi T, Heinemann SH. Extracellular hemin is a reverse use-dependent gating modifier of cardiac voltage-gated Na + channels. Biol Chem 2022; 403:1067-1081. [PMID: 36038266 DOI: 10.1515/hsz-2022-0194] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 08/10/2022] [Indexed: 02/07/2023]
Abstract
Heme (Fe2+-protoporphyrin IX) is a well-known protein prosthetic group; however, heme and hemin (Fe3+-protoporphyrin IX) are also increasingly viewed as signaling molecules. Among the signaling targets are numerous ion channels, with intracellular-facing heme-binding sites modulated by heme and hemin in the sub-µM range. Much less is known about extracellular hemin, which is expected to be more abundant, in particular after hemolytic insults. Here we show that the human cardiac voltage-gated sodium channel hNaV1.5 is potently inhibited by extracellular hemin (IC 50 ≈ 80 nM), while heme, dimethylhemin, and protoporphyrin IX are ineffective. Hemin is selective for hNaV1.5 channels: hNaV1.2, hNaV1.4, hNaV1.7, and hNaV1.8 are insensitive to 1 µM hemin. Using domain chimeras of hNaV1.5 and rat rNaV1.2, domain II was identified as the critical determinant. Mutation N803G in the domain II S3/S4 linker largely diminished the impact of hemin on the cardiac channel. This profile is reminiscent of the interaction of some peptide voltage-sensor toxins with NaV channels. In line with a mechanism of select gating modifiers, the impact of hemin on NaV1.5 channels is reversely use dependent, compatible with an interaction of hemin and the voltage sensor of domain II. Extracellular hemin thus has potential to modulate the cardiac function.
Collapse
Affiliation(s)
- Guido Gessner
- Department of Biophysics, Center for Molecular Biomedicine, Friedrich Schiller University Jena and Jena University Hospital, Hans-Knöll-Straße 2, D-07745 Jena, Germany
| | - Mahdi Jamili
- Department of Biophysics, Center for Molecular Biomedicine, Friedrich Schiller University Jena and Jena University Hospital, Hans-Knöll-Straße 2, D-07745 Jena, Germany
| | - Pascal Tomczyk
- Kekulé-Institute for Organic Chemistry and Biochemistry, University of Bonn, Gerhard-Domagk-Straße 1, D-53121 Bonn, Germany
| | - Dirk Menche
- Kekulé-Institute for Organic Chemistry and Biochemistry, University of Bonn, Gerhard-Domagk-Straße 1, D-53121 Bonn, Germany
| | - Roland Schönherr
- Department of Biophysics, Center for Molecular Biomedicine, Friedrich Schiller University Jena and Jena University Hospital, Hans-Knöll-Straße 2, D-07745 Jena, Germany
| | - Toshinori Hoshi
- Department of Physiology, University of Pennsylvania, Philadelphia, PA 19104-6085, USA
| | - Stefan H Heinemann
- Department of Biophysics, Center for Molecular Biomedicine, Friedrich Schiller University Jena and Jena University Hospital, Hans-Knöll-Straße 2, D-07745 Jena, Germany
| |
Collapse
|
17
|
Zhang J, Yuan H, Yao X, Chen S. Endogenous ion channels expressed in human embryonic kidney (HEK-293) cells. Pflugers Arch 2022; 474:665-680. [PMID: 35567642 DOI: 10.1007/s00424-022-02700-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 04/25/2022] [Accepted: 04/30/2022] [Indexed: 12/21/2022]
Abstract
Mammalian expression systems, particularly the human embryonic kidney (HEK-293) cells, combined with electrophysiological studies, have greatly benefited our understanding of the function, characteristic, and regulation of various ion channels. It was previously assumed that the existence of endogenous ion channels in native HEK-293 cells could be negligible. Still, more and more ion channels are gradually reported in native HEK-293 cells, which should draw our attention. In this regard, we summarize the different ion channels that are endogenously expressed in HEK-293 cells, including voltage-gated Na+ channels, Ca2+ channels, K+ channels, Cl- channels, nonselective cation channels, TRP channels, acid-sensitive ion channels, and Piezo channels, which may complicate the recording of the heterogeneously expressed ion channels to a certain degree. We noted that the expression patterns and channel profiles varied with different studies, which may be due to the distinct originality of the cells, cell culture conditions, passage numbers, and different recording protocols. Therefore, a better knowledge of endogenous ion channels may help minimize potential problems in characterizing heterologously expressed ion channels. Based on this, it is recommended that HEK-293 cells from unknown sources should be examined before transfection for the characterization of their functional profile, especially when the expression level of exogenous ion channels does not overwhelm the endogenous ion channels largely, or the current amplitude is not significantly higher than the native currents.
Collapse
Affiliation(s)
- Jun Zhang
- School of Biomedical Sciences and Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, Hong Kong, China
| | - Huikai Yuan
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiaoqiang Yao
- School of Biomedical Sciences and Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, Hong Kong, China
| | - Shuo Chen
- Department of Biopharmaceutical Sciences, School of Pharmacy, Harbin Medical University at Daqing, No. 39 Xinyang Rd, High-tech District, Daqing, 163319, Heilongjiang Province, China.
| |
Collapse
|
18
|
Ngum NM, Aziz MYA, Latif ML, Wall RJ, Duce IR, Mellor IR. Non-canonical endogenous expression of voltage-gated sodium channel NaV1.7 subtype by the TE671 rhabdomyosarcoma cell line. J Physiol 2022; 600:2499-2513. [PMID: 35413129 PMCID: PMC9325523 DOI: 10.1113/jp283055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 04/05/2022] [Indexed: 11/29/2022] Open
Abstract
Abstract The human TE671 cell line was originally used as a model of medulloblastoma but has since been reassigned as rhabdomyosarcoma. Despite the characterised endogenous expression of voltage‐sensitive sodium currents in these cells, the specific voltage‐gated sodium channel (VGSC) subtype underlying these currents remains unknown. To profile the VGSC subtype in undifferentiated TE671 cells, endpoint and quantitative reverse transcription–PCR (qRT‐PCR), western blot and whole‐cell patch clamp electrophysiology were performed. qRT‐PCR profiling revealed that expression of the SCN9A gene was ∼215‐fold greater than the SCN4A gene and over 400‐fold greater than any of the other VGSC genes, while western blot confirmed that the dominant SCN9A RNA was translated to a protein with a molecular mass of ∼250 kDa. Elicited sodium currents had a mean amplitude of 2.6 ± 0.7 nA with activation and fast inactivation V50 values of −31.9 ± 1.1 and −69.6 ± 1.0 mV, respectively. The currents were completely and reversibly blocked by tetrodotoxin at concentrations greater than 100 nm (IC50 = 22.3 nm). They were also very susceptible to the NaV1.7 specific blockers Huwentoxin‐IV and Protoxin‐II with IC50 values of 14.6 nm and 0.8 nm, respectively, characteristic of those previously determined for NaV1.7. Combined, the results revealed the non‐canonical and highly dominant expression of NaV1.7 in the human TE671 rhabdomyosarcoma cell line. We show that the TE671 cell line is an easy to maintain and cost‐effective model for the study of NaV1.7, a major target for the development of analgesic drugs and more generally for the study of pain. Key points Undifferentiated TE671 cells produce a voltage‐sensitive sodium current when depolarised. The voltage‐gated sodium channel isoform expressed in undifferentiated TE671 cells was previously unknown.
Through qRT‐PCR, western blot and toxin pharmacology, it is shown that undifferentiated TE671 cells dominantly (>99.5%) express the NaV1.7 isoform that is strongly associated with pain.
The TE671 cell line is, therefore, a very easy to maintain and cost‐effective model to study NaV1.7‐targeting drugs.
Collapse
Affiliation(s)
- Neville M Ngum
- School of Life Sciences, University of Nottingham, University Park, Nottingham, NG7 2RD, UK
| | - Muhammad Y A Aziz
- School of Life Sciences, University of Nottingham, University Park, Nottingham, NG7 2RD, UK
| | - M Liaque Latif
- School of Life Sciences, University of Nottingham, University Park, Nottingham, NG7 2RD, UK
| | - Richard J Wall
- School of Life Sciences, University of Nottingham, University Park, Nottingham, NG7 2RD, UK
| | - Ian R Duce
- School of Life Sciences, University of Nottingham, University Park, Nottingham, NG7 2RD, UK
| | - Ian R Mellor
- School of Life Sciences, University of Nottingham, University Park, Nottingham, NG7 2RD, UK
| |
Collapse
|
19
|
Scorpion Venom peptide, AGAP inhibits TRPV1 and potentiates the analgesic effect of lidocaine. Heliyon 2021; 7:e08560. [PMID: 35005265 PMCID: PMC8715296 DOI: 10.1016/j.heliyon.2021.e08560] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/04/2021] [Accepted: 12/01/2021] [Indexed: 11/20/2022] Open
Abstract
The current study was designed to test the hypothesis that BmK AGAP (AGAP) potentiates the analgesic effect of lidocaine. The chronic constrictive injury was performed on 72 rats to induce a rapid onset and long-lasting pain. The rats were randomly assigned to one of six groups; Group A (n = 12) received an intrathecal administration of saline, Group B (n = 12) received an intrathecal injection of lidocaine, Group C (n = 12) received an intrathecal administration of AGAP, Group D, E, and F (n = 12 each) received an intrathecal administration of lidocaine 0.005 mg/ml + AGAP 25, 50, 100 μg/kg respectively. The von Frey filaments were used to assess mechanical allodynia. Nav1.7 and TRPV1 currents were recorded by the whole-cell aspiration patch-clamp technique, and KCNQ2/3 currents were recorded by the whole-cell drilling patch-clamp technique. The whole-cell aspiration patch-clamp technique showed that AGAP inhibited TRPV1and KCNQ2/3 currents and increased the analgesic effect of lidocaine. AGAP may have a synergistic effect with lidocaine which demonstrates a potential therapeutic approach for optimizing post-operative analgesia.
Collapse
|
20
|
Mexiletine Can Be Used to Prevent and Reduce Facial Wrinkles. J Craniofac Surg 2021; 33:645-646. [PMID: 34608011 DOI: 10.1097/scs.0000000000008262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
21
|
Singh AK, Dvorak NM, Tapia CM, Mosebarger A, Ali SR, Bullock Z, Chen H, Zhou J, Laezza F. Differential Modulation of the Voltage-Gated Na + Channel 1.6 by Peptides Derived From Fibroblast Growth Factor 14. Front Mol Biosci 2021; 8:742903. [PMID: 34557523 PMCID: PMC8452925 DOI: 10.3389/fmolb.2021.742903] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 08/23/2021] [Indexed: 12/15/2022] Open
Abstract
The voltage-gated Na+ (Nav) channel is a primary molecular determinant of the initiation and propagation of the action potential. Despite the central role of the pore-forming α subunit in conferring this functionality, protein:protein interactions (PPI) between the α subunit and auxiliary proteins are necessary for the full physiological activity of Nav channels. In the central nervous system (CNS), one such PPI occurs between the C-terminal domain of the Nav1.6 channel and fibroblast growth factor 14 (FGF14). Given the primacy of this PPI in regulating the excitability of neurons in clinically relevant brain regions, peptides targeting the FGF14:Nav1.6 PPI interface could be of pre-clinical value. In this work, we pharmacologically evaluated peptides derived from FGF14 that correspond to residues that are at FGF14's PPI interface with the CTD of Nav1.6. These peptides, Pro-Leu-Glu-Val (PLEV) and Glu-Tyr-Tyr-Val (EYYV), which correspond to residues of the β12 sheet and β8-β9 loop of FGF14, respectively, were shown to inhibit FGF14:Nav1.6 complex assembly. In functional studies using whole-cell patch-clamp electrophysiology, PLEV and EYYV were shown to confer differential modulation of Nav1.6-mediated currents through mechanisms dependent upon the presence of FGF14. Crucially, these FGF14-dependent effects of PLEV and EYYV on Nav1.6-mediated currents were further shown to be dependent on the N-terminal domain of FGF14. Overall, these data suggest that the PLEV and EYYV peptides represent scaffolds to interrogate the Nav1.6 channel macromolecular complex in an effort to develop targeted pharmacological modulators.
Collapse
Affiliation(s)
- Aditya K Singh
- Department of Pharmacology and Toxicology, Galveston, TX, United States
| | - Nolan M Dvorak
- Department of Pharmacology and Toxicology, Galveston, TX, United States.,Pharmacology and Toxicology Graduate Program, Galveston, TX, United States.,Presidential Scholarship Program, University of Texas Medical Branch, Galveston, TX, United States
| | - Cynthia M Tapia
- Department of Pharmacology and Toxicology, Galveston, TX, United States.,Presidential Scholarship Program, University of Texas Medical Branch, Galveston, TX, United States
| | - Angela Mosebarger
- Department of Pharmacology and Toxicology, Galveston, TX, United States.,Pharmacology and Toxicology Graduate Program, Galveston, TX, United States.,Presidential Scholarship Program, University of Texas Medical Branch, Galveston, TX, United States
| | - Syed R Ali
- Department of Pharmacology and Toxicology, Galveston, TX, United States
| | - Zaniqua Bullock
- Department of Pharmacology and Toxicology, Galveston, TX, United States
| | - Haiying Chen
- Department of Pharmacology and Toxicology, Galveston, TX, United States
| | - Jia Zhou
- Department of Pharmacology and Toxicology, Galveston, TX, United States
| | - Fernanda Laezza
- Department of Pharmacology and Toxicology, Galveston, TX, United States
| |
Collapse
|
22
|
Murtadha AH, Azahar IIM, Sharudin NA, Has ATC, Mokhtar NF. Influence of nNav1.5 on MHC class I expression in breast cancer. J Biosci 2021. [DOI: 10.1007/s12038-021-00196-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
23
|
Alrashdi B, Dawod B, Tacke S, Kuerten S, Côté PD, Marshall JS. Mice Heterozygous for the Sodium Channel Scn8a (Nav1.6) Have Reduced Inflammatory Responses During EAE and Following LPS Challenge. Front Immunol 2021; 12:533423. [PMID: 33815353 PMCID: PMC8017164 DOI: 10.3389/fimmu.2021.533423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Accepted: 03/03/2021] [Indexed: 11/16/2022] Open
Abstract
Voltage gated sodium (Nav) channels contribute to axonal damage following demyelination in experimental autoimmune encephalomyelitis (EAE), a rodent model of multiple sclerosis (MS). The Nav1.6 isoform has been implicated as a primary contributor in this process. However, the role of Nav1.6 in immune processes, critical to the pathology of both MS and EAE, has not been extensively studied. EAE was induced with myelin oligodendrocyte (MOG35-55) peptide in Scn8admu/+ mice, which have reduced Nav1.6 levels. Scn8admu/+ mice demonstrated improved motor capacity during the recovery and early chronic phases of EAE relative to wild-type animals. In the optic nerve, myeloid cell infiltration and the effects of EAE on the axonal ultrastructure were also significantly reduced in Scn8admu/+ mice. Analysis of innate immune parameters revealed reduced plasma IL-6 levels and decreased percentages of Gr-1high/CD11b+ and Gr-1int/CD11b+ myeloid cells in the blood during the chronic phase of EAE in Scn8admu/+ mice. Elevated levels of the anti-inflammatory cytokines IL-10, IL-13, and TGF-β1 were also observed in the brains of untreated Scn8admu/+ mice. A lipopolysaccharide (LPS) model was used to further evaluate inflammatory responses. Scn8admu/+ mice displayed reduced inflammation in response to LPS challenge. To further evaluate if this was an immune cell-intrinsic difference or the result of changes in the immune or hormonal environment, mast cells were derived from the bone marrow of Scn8admu/+ mice. These mast cells also produced lower levels of IL-6, in response to LPS, compared with those from wild type mice. Our results demonstrate that in addition to its recognized impact on axonal damage, Nav1.6 impacts multiple aspects of the innate inflammatory response.
Collapse
Affiliation(s)
- Barakat Alrashdi
- Department of Biology, Dalhousie University, Halifax, NS, Canada
| | - Bassel Dawod
- Department of Pathology, Dalhousie University, Halifax, NS, Canada
| | - Sabine Tacke
- Department of Anatomy and Cell Biology, Institute of Anatomy, Friedrich-Alexander-University Erlangen-Nuremberg (FAU), Erlangen, Germany
| | - Stefanie Kuerten
- Department of Anatomy and Cell Biology, Institute of Anatomy, Friedrich-Alexander-University Erlangen-Nuremberg (FAU), Erlangen, Germany
| | - Patrice D. Côté
- Department of Biology, Dalhousie University, Halifax, NS, Canada
- Department of Ophthalmology and Visual Sciences, Dalhousie University, Halifax, NS, Canada
| | - Jean S. Marshall
- Department of Pathology, Dalhousie University, Halifax, NS, Canada
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|
24
|
Smith LB, Silva JJ, Chen C, Harrington LC, Scott JG. Fitness costs of individual and combined pyrethroid resistance mechanisms, kdr and CYP-mediated detoxification, in Aedes aegypti. PLoS Negl Trop Dis 2021; 15:e0009271. [PMID: 33760828 PMCID: PMC7990171 DOI: 10.1371/journal.pntd.0009271] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 02/25/2021] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Aedes aegypti is an important vector of many human diseases and a serious threat to human health due to its wide geographic distribution and preference for human hosts. A. aegypti also has evolved widespread resistance to pyrethroids due to the extensive use of this insecticide class over the past decades. Mutations that cause insecticide resistance result in fitness costs in the absence of insecticides. The fitness costs of pyrethroid resistance mutations in A. aegypti are still poorly understood despite their implications for arbovirus transmission. METHODOLOGY/PRINCIPLE FINDINGS We evaluated fitness based both on allele-competition and by measuring specific fitness components (i.e. life table and mating competition) to determine the costs of the different resistance mechanisms individually and in combination. We used four congenic A. aegypti strains: Rockefeller (ROCK) is susceptible to insecticides; KDR:ROCK (KR) contains only voltage-sensitive sodium channel (Vssc) mutations S989P+V1016G (kdr); CYP:ROCK (CR) contains only CYP-mediated resistance; and CYP+KDR:ROCK (CKR) contains both CYP-mediated resistance and kdr. The kdr allele frequency decreased over nine generations in the allele-competition study regardless of the presence of CYP-mediated resistance. Specific fitness costs were variable by strain and component measured. CR and CKR had a lower net reproductive rate (R0) than ROCK or KR, and KR was not different than ROCK. There was no correlation between the level of permethrin resistance conferred by the different mechanisms and their fitness cost ratio. We also found that CKR males had a reduced mating success relative to ROCK males when attempting to mate with ROCK females. CONCLUSIONS/SIGNIFICANCE Both kdr and CYP-mediated resistance have a fitness cost affecting different physiological aspects of the mosquito. CYP-mediated resistance negatively affected adult longevity and mating competition, whereas the specific fitness costs of kdr remains elusive. Understanding fitness costs helps us determine whether and how quickly resistance will be lost after pesticide application has ceased.
Collapse
Affiliation(s)
- Letícia B. Smith
- Department of Entomology, Comstock Hall, Cornell University, Ithaca, New York, United States of America
| | - Juan J. Silva
- Department of Entomology, Comstock Hall, Cornell University, Ithaca, New York, United States of America
| | - Connie Chen
- Department of Entomology, Comstock Hall, Cornell University, Ithaca, New York, United States of America
| | - Laura C. Harrington
- Department of Entomology, Comstock Hall, Cornell University, Ithaca, New York, United States of America
| | - Jeffrey G. Scott
- Department of Entomology, Comstock Hall, Cornell University, Ithaca, New York, United States of America
| |
Collapse
|
25
|
Nathan S, Gabelli SB, Yoder JB, Srinivasan L, Aldrich RW, Tomaselli GF, Ben-Johny M, Amzel LM. Structural basis of cytoplasmic NaV1.5 and NaV1.4 regulation. J Gen Physiol 2020; 153:211587. [PMID: 33306788 PMCID: PMC7953540 DOI: 10.1085/jgp.202012722] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 11/06/2020] [Indexed: 12/14/2022] Open
Abstract
Voltage-gated sodium channels (NaVs) are membrane proteins responsible for the rapid upstroke of the action potential in excitable cells. There are nine human voltage-sensitive NaV1 isoforms that, in addition to their sequence differences, differ in tissue distribution and specific function. This review focuses on isoforms NaV1.4 and NaV1.5, which are primarily expressed in skeletal and cardiac muscle cells, respectively. The determination of the structures of several eukaryotic NaVs by single-particle cryo-electron microscopy (cryo-EM) has brought new perspective to the study of the channels. Alignment of the cryo-EM structure of the transmembrane channel pore with x-ray crystallographic structures of the cytoplasmic domains illustrates the complementary nature of the techniques and highlights the intricate cellular mechanisms that modulate these channels. Here, we review structural insights into the cytoplasmic C-terminal regulation of NaV1.4 and NaV1.5 with special attention to Ca2+ sensing by calmodulin, implications for disease, and putative channel dimerization.
Collapse
Affiliation(s)
- Sara Nathan
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Sandra B Gabelli
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD.,Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD.,Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Jesse B Yoder
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Lakshmi Srinivasan
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Richard W Aldrich
- Department of Neuroscience, University of Texas at Austin, Austin, TX
| | - Gordon F Tomaselli
- Division of Cardiology, Department of Medicine, Albert Einstein College of Medicine, Bronx, NY
| | - Manu Ben-Johny
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY
| | - L Mario Amzel
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD.,Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
26
|
Sun P, Ji Z, Li Z, Pan B. Prevention of scar hyperplasia in the skin by conotoxin: A prospective review. J Cosmet Dermatol 2020; 20:1885-1888. [PMID: 33025725 DOI: 10.1111/jocd.13761] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 09/05/2020] [Accepted: 09/28/2020] [Indexed: 02/05/2023]
Abstract
Scars are often considered to be skin problems that affect beauty. The tension acting on the edge of the wound is the main factor causing the scar hyperplasia. At present, the clinical use of botulinum toxin A (BTX-A) around the wound to cause transient muscle paralysis reduce the muscle movement around the wound and wound tension to prevent scar hyperplasia during wound healing. But the use of BTX-A to prevent scarring requires the use of a syringe. The syringe can cause trauma and pain when it pricks the skin for BTX-A injection. The conotoxin which is secreted by the poison glands on the inside of the venom tube and capsule of the snail provides a simple and effective way to prevent skin scar hyperplasia. We reviewed the classification of conotoxin, the conotoxin's mechanism of preventing scar hyperplasia, and the research direction of conotoxin in the future and provided reference for promoting the application of conotoxin in preventing skin scar hyperplasia.
Collapse
Affiliation(s)
- Pengfei Sun
- Department of Auricular Reconstruction, Plastic Surgery Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Zhonglei Ji
- Department of Plastic Surgery, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Zhengyong Li
- Department of Plastic Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Bo Pan
- Department of Auricular Reconstruction, Plastic Surgery Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
27
|
Montenarh M, Götz C. Protein kinase CK2 and ion channels (Review). Biomed Rep 2020; 13:55. [PMID: 33082952 PMCID: PMC7560519 DOI: 10.3892/br.2020.1362] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 07/28/2020] [Indexed: 12/18/2022] Open
Abstract
Protein kinase CK2 appears as a tetramer or higher molecular weight oligomer composed of catalytic CK2α, CK2α' subunits and non-catalytic regulatory CK2β subunits or as individual subunits. It is implicated in a variety of different regulatory processes, such as Akt signalling, splicing and DNA repair within eukaryotic cells. The present review evaluates the influence of CK2 on ion channels in the plasma membrane. CK2 phosphorylates platform proteins such as calmodulin and ankyrin G, which bind to channel proteins for a physiological transport to and positioning into the membrane. In addition, CK2 directly phosphorylates a variety of channel proteins directly to regulate opening and closing of the channels. Thus, modulation of CK2 activities by specific inhibitors, by siRNA technology or by CRISPR/Cas technology has an influence on intracellular ion concentrations and thereby on cellular signalling. The physiological regulation of the intracellular ion concentration is important for cell survival and correct intracellular signalling. Disturbance of this regulation results in a variety of different diseases including epilepsy, heart failure, cystic fibrosis and diabetes. Therefore, these effects should be considered when using CK2 inhibition as a treatment option for cancer.
Collapse
Affiliation(s)
- Mathias Montenarh
- Medical Biochemistry and Molecular Biology, Saarland University, D-66424 Homburg, Saarland, Germany
| | - Claudia Götz
- Medical Biochemistry and Molecular Biology, Saarland University, D-66424 Homburg, Saarland, Germany
| |
Collapse
|
28
|
Witty DR, Alvaro G, Derjean D, Giblin GMP, Gunn K, Large C, Macpherson DT, Morisset V, Owen D, Palmer J, Rugiero F, Tate S, Hinckley CA, Naik H. Discovery of Vixotrigine: A Novel Use-Dependent Sodium Channel Blocker for the Treatment of Trigeminal Neuralgia. ACS Med Chem Lett 2020; 11:1678-1687. [PMID: 32945812 PMCID: PMC7488392 DOI: 10.1021/acsmedchemlett.0c00263] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Accepted: 07/16/2020] [Indexed: 12/19/2022] Open
Abstract
Drugs that block voltage-gated sodium channels (NaVs) have utility in treating conditions including pain, epilepsy, and cardiac arrhythmias and as anesthetics (Lancet Neurol.20109413424; Expert Opin. Ther. Pat.201020755779). The identification of compounds with improved efficacy and safety is a key aim for the discovery of improved NaV blocking drugs (Comprehensive Medicinal Chemistry III; (Elsevier, 2017; pp 131-175). We report the identification of a novel class of brain penetrant and voltage-gated sodium channel blockers, leading to the discovery of vixotrigine, a use-dependent sodium channel blocker with activity in in vivo models of pain. Vixotrigine has excellent physiocochemical properties for drug development, and both preclinical and clinical data support a safety profile suitable for potential use in neuropathic pain and other conditions. It has shown efficacy in a Phase II study for pain associated with trigeminal neuralgia.
Collapse
Affiliation(s)
- David R. Witty
- Convergence
Pharmaceuticals Ltd., a Biogen Company, Babraham Research
Campus, Cambridge CB22 3AT,
U.K.
| | - Giuseppe Alvaro
- Convergence
Pharmaceuticals Ltd., a Biogen Company, Babraham Research
Campus, Cambridge CB22 3AT,
U.K.
| | - Dominique Derjean
- Convergence
Pharmaceuticals Ltd., a Biogen Company, Babraham Research
Campus, Cambridge CB22 3AT,
U.K.
| | - Gerard M. P. Giblin
- Convergence
Pharmaceuticals Ltd., a Biogen Company, Babraham Research
Campus, Cambridge CB22 3AT,
U.K.
| | - Kevin Gunn
- Convergence
Pharmaceuticals Ltd., a Biogen Company, Babraham Research
Campus, Cambridge CB22 3AT,
U.K.
| | - Charles Large
- Convergence
Pharmaceuticals Ltd., a Biogen Company, Babraham Research
Campus, Cambridge CB22 3AT,
U.K.
| | - David T. Macpherson
- Convergence
Pharmaceuticals Ltd., a Biogen Company, Babraham Research
Campus, Cambridge CB22 3AT,
U.K.
| | - Valerie Morisset
- Convergence
Pharmaceuticals Ltd., a Biogen Company, Babraham Research
Campus, Cambridge CB22 3AT,
U.K.
| | - Davina Owen
- Convergence
Pharmaceuticals Ltd., a Biogen Company, Babraham Research
Campus, Cambridge CB22 3AT,
U.K.
| | - Joanne Palmer
- Convergence
Pharmaceuticals Ltd., a Biogen Company, Babraham Research
Campus, Cambridge CB22 3AT,
U.K.
| | - Francois Rugiero
- Convergence
Pharmaceuticals Ltd., a Biogen Company, Babraham Research
Campus, Cambridge CB22 3AT,
U.K.
| | - Simon Tate
- Convergence
Pharmaceuticals Ltd., a Biogen Company, Babraham Research
Campus, Cambridge CB22 3AT,
U.K.
| | | | - Himanshu Naik
- Biogen
Inc., 225 Binney Street, Cambridge, Massachusetts 02142,
United States
| |
Collapse
|
29
|
Ebbinghaus M, Tuchscherr L, Segond von Banchet G, Liebmann L, Adams V, Gajda M, Hübner CA, Kurth I, Schaible HG. Gain-of-function mutation in SCN11A causes itch and affects neurogenic inflammation and muscle function in Scn11a+/L799P mice. PLoS One 2020; 15:e0237101. [PMID: 32817686 PMCID: PMC7440628 DOI: 10.1371/journal.pone.0237101] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 07/20/2020] [Indexed: 12/22/2022] Open
Abstract
Mutations in the genes encoding for voltage-gated sodium channels cause profound sensory disturbances and other symptoms dependent on the distribution of a particular channel subtype in different organs. Humans with the gain-of-function mutation p.Leu811Pro in SCN11A (encoding for the voltage-gated Nav1.9 channel) exhibit congenital insensitivity to pain, pruritus, self-inflicted injuries, slow healing wounds, muscle weakness, Charcot-like arthropathies, and intestinal dysmotility. As already shown, knock-in mice (Scn11a+/L799P) carrying the orthologous mutation p.Leu799Pro replicate reduced pain sensitivity and show frequent tissue lesions. In the present study we explored whether Scn11a+/L799P mice develop also pruritus, muscle weakness, and changes in gastrointestinal transit time. Furthermore, we analyzed morphological and functional differences in nerves, skeletal muscle, joints and small intestine from Scn11a+/L799P and Scn11a+/+ wild type mice. Compared to Scn11a+/+ mice, Scn11a+/L799P mice showed enhanced scratching bouts before skin lesions developed, indicating pruritus. Scn11a+/L799P mice exhibited reduced grip strength, but no disturbances in motor coordination. Skeletal muscle fiber types and joint architecture were unaltered in Scn11a+/L799P mice. Their gastrointestinal transit time was unaltered. The small intestine from Scn11a+/L799P showed a small shift towards less frequent peristaltic movements. Similar proportions of lumbar dorsal root ganglion neurons from Scn11a+/L799P and Scn11a+/+ mice were calcitonin gene-related peptide (CGRP-) positive, but isolated sciatic nerves from Scn11a+/L799P mice exhibited a significant reduction of the capsaicin-evoked release of CGRP indicating reduced neurogenic inflammation. These data indicate important Nav1.9 channel functions in several organs in both humans and mice. They support the pathophysiological relevance of increased basal activity of Nav1.9 channels for sensory abnormalities (pain and itch) and suggest resulting malfunctions of the motor system and of the gastrointestinal tract. Scn11a+/L799P mice are suitable to investigate the role of Nav1.9, and to explore the pathophysiological changes and mechanisms which develop as a consequence of Nav1.9 hyperactivity.
Collapse
Affiliation(s)
- Matthias Ebbinghaus
- Institute of Physiology 1/Neurophysiology, University Hospital - Friedrich Schiller University Jena, Jena, Germany
| | - Lorena Tuchscherr
- Institute of Medical Microbiology, University Hospital - Friedrich Schiller University Jena, Jena, Germany
| | - Gisela Segond von Banchet
- Institute of Physiology 1/Neurophysiology, University Hospital - Friedrich Schiller University Jena, Jena, Germany
| | - Lutz Liebmann
- Institute of Human Genetics, University Hospital - Friedrich Schiller University Jena, Jena, Germany
| | - Volker Adams
- Laboratory of Molecular and Experimental Cardiology, Heart Center Dresden, Technische Universität Dresden, Dresden, Germany
| | - Mieczyslaw Gajda
- Institute of Pathology, University Hospital - Friedrich Schiller University Jena, Jena, Germany
| | - Christian A. Hübner
- Institute of Human Genetics, University Hospital - Friedrich Schiller University Jena, Jena, Germany
| | - Ingo Kurth
- Institute of Human Genetics, Medical Faculty - RWTH Aachen University, Aachen, Germany
| | - Hans-Georg Schaible
- Institute of Physiology 1/Neurophysiology, University Hospital - Friedrich Schiller University Jena, Jena, Germany
| |
Collapse
|
30
|
Singh AK, Wadsworth PA, Tapia CM, Aceto G, Ali SR, Chen H, D'Ascenzo M, Zhou J, Laezza F. Mapping of the FGF14:Nav1.6 complex interface reveals FLPK as a functionally active peptide modulating excitability. Physiol Rep 2020; 8:e14505. [PMID: 32671946 PMCID: PMC7363588 DOI: 10.14814/phy2.14505] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 06/15/2020] [Accepted: 06/16/2020] [Indexed: 02/07/2023] Open
Abstract
The voltage-gated sodium (Nav) channel complex is comprised of pore-forming α subunits (Nav1.1-1.9) and accessory regulatory proteins such as the intracellular fibroblast growth factor 14 (FGF14). The cytosolic Nav1.6 C-terminal tail binds directly to FGF14 and this interaction modifies Nav1.6-mediated currents with effects on intrinsic excitability in the brain. Previous studies have identified the FGF14V160 residue within the FGF14 core domain as a hotspot for the FGF14:Nav1.6 complex formation. Here, we used three short amino acid peptides around FGF14V160 to probe for the FGF14 interaction with the Nav1.6 C-terminal tail and to evaluate the activity of the peptide on Nav1.6-mediated currents. In silico docking predicts FLPK to bind to FGF14V160 with the expectation of interfering with the FGF14:Nav1.6 complex formation, a phenotype that was confirmed by the split-luciferase assay (LCA) and surface plasmon resonance (SPR), respectively. Whole-cell patch-clamp electrophysiology studies demonstrate that FLPK is able to prevent previously reported FGF14-dependent phenotypes of Nav1.6 currents, but that its activity requires the FGF14 N-terminal tail, a domain that has been shown to contribute to Nav1.6 inactivation independently from the FGF14 core domain. In medium spiny neurons in the nucleus accumbens, where both FGF14 and Nav1.6 are abundantly expressed, FLPK significantly increased firing frequency by a mechanism consistent with the ability of the tetrapeptide to interfere with Nav1.6 inactivation and potentiate persistent Na+ currents. Taken together, these results indicate that FLPK might serve as a probe for characterizing molecular determinants of neuronal excitability and a peptide scaffold to develop allosteric modulators of Nav channels.
Collapse
Affiliation(s)
- Aditya K. Singh
- Department of Pharmacology & ToxicologyUniversità Cattolica del Sacro CuoreRomeItaly
| | - Paul A. Wadsworth
- Department of Pharmacology & ToxicologyUniversità Cattolica del Sacro CuoreRomeItaly
- M.D.‐Ph.D. Combined Degree ProgramUniversità Cattolica del Sacro CuoreRomeItaly
- Biochemistry and Molecular Biology Graduate ProgramUniversità Cattolica del Sacro CuoreRomeItaly
| | - Cynthia M. Tapia
- Department of Pharmacology & ToxicologyUniversità Cattolica del Sacro CuoreRomeItaly
- NIEHS Environmental Toxicology Training ProgramUniversità Cattolica del Sacro CuoreRomeItaly
| | - Giuseppe Aceto
- Institute of Human PhysiologyUniversità Cattolica del Sacro CuoreRomeItaly
- Department of NeuroscienceUniversità Cattolica del Sacro CuoreRomeItaly
- Fondazione Policlinico Universitario A. GemelliIRCCSRomeItaly
| | - Syed R. Ali
- Department of Pharmacology & ToxicologyUniversità Cattolica del Sacro CuoreRomeItaly
| | - Haiying Chen
- Department of Pharmacology & ToxicologyUniversità Cattolica del Sacro CuoreRomeItaly
| | - Marcello D'Ascenzo
- Institute of Human PhysiologyUniversità Cattolica del Sacro CuoreRomeItaly
- Department of NeuroscienceUniversità Cattolica del Sacro CuoreRomeItaly
- Fondazione Policlinico Universitario A. GemelliIRCCSRomeItaly
| | - Jia Zhou
- Department of Pharmacology & ToxicologyUniversità Cattolica del Sacro CuoreRomeItaly
- Center for Addiction ResearchUniversity of Texas Medical BranchGalvestonTXUSA
| | - Fernanda Laezza
- Department of Pharmacology & ToxicologyUniversità Cattolica del Sacro CuoreRomeItaly
- Center for Addiction ResearchUniversity of Texas Medical BranchGalvestonTXUSA
- Center for Neurodegenerative DiseasesUniversity of Texas Medical BranchGalvestonTXUSA
| |
Collapse
|
31
|
Imai R, Horita S, Ono Y, Hagihara K, Shimizu M, Maejima Y, Shimomura K. Goshajinkigan, a Traditional Japanese Medicine, Suppresses Voltage-Gated Sodium Channel Nav1.4 Currents in C2C12 Cells. Biores Open Access 2020; 9:116-120. [PMID: 32368413 PMCID: PMC7194311 DOI: 10.1089/biores.2019.0034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Goshajinkigan (GJG) is a traditional Japanese Kampo medicine used clinically to treat muscle pain in Japan. However, its underlying mechanism remains unclear. Since voltage-gated sodium channel (Nav) 1.4 is involved in skeletal muscle contraction, we investigated the possibility that GJG may affect Nav1.4 currents. By using an electrophysiological technique on skeletal muscle cell line C2C12, we found that GJG suppresses Nav1.4 currents in C2C12 cells. It is suggested that GJG may improve skeletal muscle stiffness or cramps by inhibiting abnormal Nav1.4 excitation. GJG may act as a Nav1.4 blocker and may be useful to treat muscle stiffness and clamps as well as easing the pain.
Collapse
Affiliation(s)
- Ryota Imai
- Department of Bioregulation and Pharmacological Medicine, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Shoichiro Horita
- Department of Bioregulation and Pharmacological Medicine, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Yuko Ono
- Department of Bioregulation and Pharmacological Medicine, Fukushima Medical University School of Medicine, Fukushima, Japan.,Department of Disaster and Emergency Medicine, Graduate School of Medicine, Kobe University, Kobe, Japan
| | - Keisuke Hagihara
- Department of Kampo Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Masaru Shimizu
- Department of Bioregulation and Pharmacological Medicine, Fukushima Medical University School of Medicine, Fukushima, Japan.,Department of Neurology, Matsumura General Hospital, Fukushima, Japan
| | - Yuko Maejima
- Department of Bioregulation and Pharmacological Medicine, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Kenju Shimomura
- Department of Bioregulation and Pharmacological Medicine, Fukushima Medical University School of Medicine, Fukushima, Japan
| |
Collapse
|
32
|
Abstract
Analgesics, particularly opioids, have been routinely used in the emergency treatment of ischemic chest pain for a long time. In the past two decades; however, several studies have raised the possibility of the harmful effects of opioid administration. In 2014, the American Heart Association (AHA)/American College of Cardiology Foundation (ACCF) changed the guidelines regarding the use of opioids from class IC to class IIb for non-ST elevation acute coronary syndrome. And in 2015, the European Society of Cardiology (ESC) guidelines incidentally noted the side effects of opioids. In ST-segment elevation myocardial infarction, both ESC and AHA/ACCF still recommend the use of opioids. Given the need for adequate pain relief in ischemic chest pain in the emergency setting, it is necessary to understand the adverse effects of analgesia, while still providing sufficiently potent options for analgesia. The primary purpose of this review is to quantify the effects of analgesics commonly used in the prehospital and emergency department in patients with ischemic chest pain.
Collapse
|
33
|
Johnson SR, Rikli HG. Aspartic Acid Isomerization Characterized by High Definition Mass Spectrometry Significantly Alters the Bioactivity of a Novel Toxin from Poecilotheria. Toxins (Basel) 2020; 12:E207. [PMID: 32218140 PMCID: PMC7232244 DOI: 10.3390/toxins12040207] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 03/20/2020] [Accepted: 03/23/2020] [Indexed: 11/25/2022] Open
Abstract
Research in toxinology has created a pharmacological paradox. With an estimated 220,000 venomous animals worldwide, the study of peptidyl toxins provides a vast number of effector molecules. However, due to the complexity of the protein-protein interactions, there are fewer than ten venom-derived molecules on the market. Structural characterization and identification of post-translational modifications are essential to develop biological lead structures into pharmaceuticals. Utilizing advancements in mass spectrometry, we have created a high definition approach that fuses conventional high-resolution MS-MS with ion mobility spectrometry (HDMSE) to elucidate these primary structure characteristics. We investigated venom from ten species of "tiger" spider (Genus: Poecilotheria) and discovered they contain isobaric conformers originating from non-enzymatic Asp isomerization. One conformer pair conserved in five of ten species examined, denominated PcaTX-1a and PcaTX-1b, was found to be a 36-residue peptide with a cysteine knot, an amidated C-terminus, and isoAsp33Asp substitution. Although the isomerization of Asp has been implicated in many pathologies, this is the first characterization of Asp isomerization in a toxin and demonstrates the isomerized product's diminished physiological effects. This study establishes the value of a HDMSE approach to toxin screening and characterization.
Collapse
Affiliation(s)
- Stephen R. Johnson
- Carbon Dynamics Institute LLC, Sherman, IL 62684, USA
- Chemistry Department, University of Illinois Springfield, Springfield, IL 62703, USA
| | - Hillary G. Rikli
- College of Liberal Arts & Sciences, University of Illinois Springfield, Springfield, IL 62703, USA;
| |
Collapse
|
34
|
Shimizu A, Zankov DP, Sato A, Komeno M, Toyoda F, Yamazaki S, Makita T, Noda T, Ikawa M, Asano Y, Miyashita Y, Takashima S, Morita H, Ishikawa T, Makita N, Hitosugi M, Matsuura H, Ohno S, Horie M, Ogita H. Identification of transmembrane protein 168 mutation in familial Brugada syndrome. FASEB J 2020; 34:6399-6417. [PMID: 32175648 DOI: 10.1096/fj.201902991r] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 02/19/2020] [Accepted: 03/02/2020] [Indexed: 12/30/2022]
Abstract
Brugada syndrome (BrS) is an inherited channelopathy responsible for almost 20% of sudden cardiac deaths in patients with nonstructural cardiac diseases. Approximately 70% of BrS patients, the causative gene mutation(s) remains unknown. In this study, we used whole exome sequencing to investigate candidate mutations in a family clinically diagnosed with BrS. A heterozygous 1616G>A substitution (R539Q mutation) was identified in the transmembrane protein 168 (TMEM168) gene of symptomatic individuals. Similar to endogenous TMEM168, both TMEM168 wild-type (WT) and mutant proteins that were ectopically induced in HL-1 cells showed nuclear membrane localization. A significant decrease in Na+ current and Nav 1.5 protein expression was observed in HL-1 cardiomyocytes expressing mutant TMEM168. Ventricular tachyarrhythmias and conduction disorders were induced in the heterozygous Tmem168 1616G>A knock-in mice by pharmacological stimulation, but not in WT mice. Na+ current was reduced in ventricular cardiomyocytes isolated from the Tmem168 knock-in heart, and Nav 1.5 expression was also impaired. This impairment was dependent on increased Nedd4-2 binding to Nav 1.5 and subsequent ubiquitination. Collectively, our results show an association between the TMEM168 1616G>A mutation and arrhythmogenesis in a family with BrS.
Collapse
Affiliation(s)
- Akio Shimizu
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular Biology, Shiga University of Medical Science, Otsu, Japan
| | - Dimitar P Zankov
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular Biology, Shiga University of Medical Science, Otsu, Japan.,Department of Bioscience and Genetics, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Akira Sato
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular Biology, Shiga University of Medical Science, Otsu, Japan
| | - Masahiro Komeno
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular Biology, Shiga University of Medical Science, Otsu, Japan
| | - Futoshi Toyoda
- Division of Cell Physiology, Department of Physiology, Shiga University of Medical Science, Otsu, Japan
| | - Satoru Yamazaki
- Department of Molecular Pharmacology, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Toshinori Makita
- Division of Cardiac Electrophysiology, Department of Cardiovascular Center, Osaka Red Cross Hospital, Osaka, Japan
| | - Taichi Noda
- Animal Resource Center for Infectious Diseases, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | - Masahito Ikawa
- Animal Resource Center for Infectious Diseases, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | - Yoshihiro Asano
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Yohei Miyashita
- Department of Legal Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Seiji Takashima
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine, Suita, Japan
| | - Hiroshi Morita
- Department of Cardiovascular Therapeutics, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Taisuke Ishikawa
- Omics Research Center, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Naomasa Makita
- Omics Research Center, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Masahito Hitosugi
- Department of Legal Medicine, Shiga University of Medical Science, Otsu, Japan
| | - Hiroshi Matsuura
- Division of Cell Physiology, Department of Physiology, Shiga University of Medical Science, Otsu, Japan
| | - Seiko Ohno
- Department of Bioscience and Genetics, National Cerebral and Cardiovascular Center, Suita, Japan.,Center for Epidemiologic Research in Asia, Shiga University of Medical Science, Otsu, Japan
| | - Minoru Horie
- Center for Epidemiologic Research in Asia, Shiga University of Medical Science, Otsu, Japan
| | - Hisakazu Ogita
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular Biology, Shiga University of Medical Science, Otsu, Japan
| |
Collapse
|
35
|
Pedrosa CDSG, Souza LRQ, Gomes TA, de Lima CVF, Ledur PF, Karmirian K, Barbeito-Andres J, Costa MDN, Higa LM, Rossi ÁD, Bellio M, Tanuri A, Prata-Barbosa A, Tovar-Moll F, Garcez PP, Lara FA, Molica RJR, Rehen SK. The cyanobacterial saxitoxin exacerbates neural cell death and brain malformations induced by Zika virus. PLoS Negl Trop Dis 2020; 14:e0008060. [PMID: 32163415 PMCID: PMC7067372 DOI: 10.1371/journal.pntd.0008060] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 01/15/2020] [Indexed: 12/17/2022] Open
Abstract
The northeast (NE) region of Brazil commonly goes through drought periods, which favor cyanobacterial blooms, capable of producing neurotoxins with implications for human and animal health. The most severe dry spell in the history of Brazil occurred between 2012 and 2016. Coincidently, the highest incidence of microcephaly associated with the Zika virus (ZIKV) outbreak took place in the NE region of Brazil during the same years. In this work, we tested the hypothesis that saxitoxin (STX), a neurotoxin produced in South America by the freshwater cyanobacteria Raphidiopsis raciborskii, could have contributed to the most severe Congenital Zika Syndrome (CZS) profile described worldwide. Quality surveillance showed higher cyanobacteria amounts and STX occurrence in human drinking water supplies of NE compared to other regions of Brazil. Experimentally, we described that STX doubled the quantity of ZIKV-induced neural cell death in progenitor areas of human brain organoids, while the chronic ingestion of water contaminated with STX before and during gestation caused brain abnormalities in offspring of ZIKV-infected immunocompetent C57BL/6J mice. Our data indicate that saxitoxin-producing cyanobacteria is overspread in water reservoirs of the NE and might have acted as a co-insult to ZIKV infection in Brazil. These results raise a public health concern regarding the consequences of arbovirus outbreaks happening in areas with droughts and/or frequent freshwater cyanobacterial blooms.
Collapse
Affiliation(s)
| | - Leticia R. Q. Souza
- D’Or Institute for Research and Education (IDOR), Rio de Janeiro, Rio de Janeiro, Brazil
| | - Tiago A. Gomes
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil
- Laboratory of Cellular Microbiology, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Caroline V. F. de Lima
- D’Or Institute for Research and Education (IDOR), Rio de Janeiro, Rio de Janeiro, Brazil
| | - Pitia F. Ledur
- D’Or Institute for Research and Education (IDOR), Rio de Janeiro, Rio de Janeiro, Brazil
| | - Karina Karmirian
- D’Or Institute for Research and Education (IDOR), Rio de Janeiro, Rio de Janeiro, Brazil
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Jimena Barbeito-Andres
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcelo do N. Costa
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luiza M. Higa
- Department of Genetics, Institute of Biology, Federal University of Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Átila D. Rossi
- Department of Genetics, Institute of Biology, Federal University of Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Maria Bellio
- Institute of Microbiology Paulo de Goes, Federal University of Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Amilcar Tanuri
- Department of Genetics, Institute of Biology, Federal University of Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Arnaldo Prata-Barbosa
- D’Or Institute for Research and Education (IDOR), Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fernanda Tovar-Moll
- D’Or Institute for Research and Education (IDOR), Rio de Janeiro, Rio de Janeiro, Brazil
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Patricia P. Garcez
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Flavio A. Lara
- Laboratory of Cellular Microbiology, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Renato J. R. Molica
- Academic Unit of Garanhuns, Federal Rural University of Pernambuco, Garanhuns, Pernambuco, Brazil
| | - Stevens K. Rehen
- D’Or Institute for Research and Education (IDOR), Rio de Janeiro, Rio de Janeiro, Brazil
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
36
|
Li Q, Zhai Z, Li J. Fibroblast growth factor homologous factors are potential ion channel modifiers associated with cardiac arrhythmias. Eur J Pharmacol 2020; 871:172920. [PMID: 31935396 DOI: 10.1016/j.ejphar.2020.172920] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Revised: 12/10/2019] [Accepted: 01/10/2020] [Indexed: 12/27/2022]
Abstract
Stable electrical activity in cardiac myocytes is the basis of maintaining normal myocardial systolic and diastolic function. Cardiac ionic currents and their associated regulatory proteins are crucial to myocyte excitability and heart function. Fibroblast growth factor homologous factors (FHFs) are intracellular noncanonical fibroblast growth factors (FGFs) that are incapable of activating FGF receptors. The main functions of FHFs are to regulate ion channels and influence excitability, which are processes involved in sustaining normal cardiac function. In addition to their regulatory effect on ion channels, FHFs can be regulators of cardiac hypertrophic signaling and alter signaling pathways, including the protein kinase, NF<kappa>B, and p53 pathways, which are related to the pathological processes of heart diseases. This review emphasizes FHF-mediated regulation of cardiac excitability and the association of FHFs with cardiac arrhythmias and explores the idea that abnormal FHFs may be an unrecognized cause of cardiac disorders.
Collapse
Affiliation(s)
- Qing Li
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Zhenyu Zhai
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Juxiang Li
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China.
| |
Collapse
|
37
|
Spider Knottin Pharmacology at Voltage-Gated Sodium Channels and Their Potential to Modulate Pain Pathways. Toxins (Basel) 2019; 11:toxins11110626. [PMID: 31671792 PMCID: PMC6891507 DOI: 10.3390/toxins11110626] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 10/24/2019] [Accepted: 10/24/2019] [Indexed: 12/15/2022] Open
Abstract
Voltage-gated sodium channels (NaVs) are a key determinant of neuronal signalling. Neurotoxins from diverse taxa that selectively activate or inhibit NaV channels have helped unravel the role of NaV channels in diseases, including chronic pain. Spider venoms contain the most diverse array of inhibitor cystine knot (ICK) toxins (knottins). This review provides an overview on how spider knottins modulate NaV channels and describes the structural features and molecular determinants that influence their affinity and subtype selectivity. Genetic and functional evidence support a major involvement of NaV subtypes in various chronic pain conditions. The exquisite inhibitory properties of spider knottins over key NaV subtypes make them the best lead molecules for the development of novel analgesics to treat chronic pain.
Collapse
|
38
|
Kazemi SM, Sabatier JM. Venoms of Iranian Scorpions (Arachnida, Scorpiones) and Their Potential for Drug Discovery. Molecules 2019; 24:molecules24142670. [PMID: 31340554 PMCID: PMC6680535 DOI: 10.3390/molecules24142670] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Revised: 07/16/2019] [Accepted: 07/20/2019] [Indexed: 12/19/2022] Open
Abstract
Scorpions, a characteristic group of arthropods, are among the earliest diverging arachnids, dating back almost 440 million years. One of the many interesting aspects of scorpions is that they have venom arsenals for capturing prey and defending against predators, which may play a critical role in their evolutionary success. Unfortunately, however, scorpion envenomation represents a serious health problem in several countries, including Iran. Iran is acknowledged as an area with a high richness of scorpion species and families. The diversity of the scorpion fauna in Iran is the subject of this review, in which we report a total of 78 species and subspecies in 19 genera and four families. We also list some of the toxins or genes studied from five species, including Androctonus crassicauda, Hottentotta zagrosensis, Mesobuthus phillipsi, Odontobuthus doriae, and Hemiscorpius lepturus, in the Buthidae and Hemiscorpiidae families. Lastly, we review the diverse functions of typical toxins from the Iranian scorpion species, including their medical applications.
Collapse
Affiliation(s)
- Seyed Mahdi Kazemi
- Zagros Herpetological Institute, No 12, Somayyeh 14 Avenue, 3715688415 Qom, Iran.
| | - Jean-Marc Sabatier
- Institute of NeuroPhysiopathology, UMR 7051, Faculté de Médecine Secteur Nord, 51, Boulevard Pierre Dramard-CS80011, 13344-Marseille Cedex 15, France
| |
Collapse
|
39
|
Hompoonsup S, Chambers D, Doherty P, Williams G. No transcriptional evidence for active Na v channels in two classes of cancer cell. Channels (Austin) 2019; 13:311-320. [PMID: 31329011 PMCID: PMC6682260 DOI: 10.1080/19336950.2019.1644858] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
Voltage-gated sodium channel (Nav) expression in non-excitable cells has raised questions regarding their non-canonical roles. Interestingly, a growing body of evidence also points towards the prevalence of aberrant Nav expression in malignant tumors, potentially opening a new therapeutic window. In this study, the transcriptional consequences of channel inhibition were investigated in non-small cell lung carcinoma H460 and neuroblastoma SH-SYSY cell lines, that both express Nav1.7. Channel activity was blocked by the application of both selective, ProTx-II, and non-selective, tetrodotoxin, inhibitors. Global gene expression profiling did not point to any statistically significant inhibition-associated perturbation of the transcriptome. A small subset of genes that showed relatively consistent changes across multiple treatments were further assayed in the context of a multiplex bead expression array which failed to recapitulate the changes seen in the global array. We conclude that there is no robust transcriptional signature associated with the inhibition of two sodium channel expressing cancer cell lines and consequently sodium channel inhibition will not lend itself to therapeutic approaches such as transcription-based drug repurposing.
Collapse
Affiliation(s)
- Supanida Hompoonsup
- a Wolfson Centre for Age-Related Diseases, King's College London , London , UK.,b Learning Institute, King Mongkut's University of Technology Thonburi , Bangkok , Thailand
| | - David Chambers
- a Wolfson Centre for Age-Related Diseases, King's College London , London , UK
| | - Patrick Doherty
- a Wolfson Centre for Age-Related Diseases, King's College London , London , UK
| | - Gareth Williams
- a Wolfson Centre for Age-Related Diseases, King's College London , London , UK
| |
Collapse
|
40
|
Fux JE, Mehta A, Moffat J, Spafford JD. Eukaryotic Voltage-Gated Sodium Channels: On Their Origins, Asymmetries, Losses, Diversification and Adaptations. Front Physiol 2018; 9:1406. [PMID: 30519187 PMCID: PMC6259924 DOI: 10.3389/fphys.2018.01406] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 09/14/2018] [Indexed: 12/19/2022] Open
Abstract
The appearance of voltage-gated, sodium-selective channels with rapid gating kinetics was a limiting factor in the evolution of nervous systems. Two rounds of domain duplications generated a common 24 transmembrane segment (4 × 6 TM) template that is shared amongst voltage-gated sodium (Nav1 and Nav2) and calcium channels (Cav1, Cav2, and Cav3) and leak channel (NALCN) plus homologs from yeast, different single-cell protists (heterokont and unikont) and algae (green and brown). A shared architecture in 4 × 6 TM channels include an asymmetrical arrangement of extended extracellular L5/L6 turrets containing a 4-0-2-2 pattern of cysteines, glycosylated residues, a universally short III-IV cytoplasmic linker and often a recognizable, C-terminal PDZ binding motif. Six intron splice junctions are conserved in the first domain, including a rare U12-type of the minor spliceosome provides support for a shared heritage for sodium and calcium channels, and a separate lineage for NALCN. The asymmetrically arranged pores of 4x6 TM channels allows for a changeable ion selectivity by means of a single lysine residue change in the high field strength site of the ion selectivity filter in Domains II or III. Multicellularity and the appearance of systems was an impetus for Nav1 channels to adapt to sodium ion selectivity and fast ion gating. A non-selective, and slowly gating Nav2 channel homolog in single cell eukaryotes, predate the diversification of Nav1 channels from a basal homolog in a common ancestor to extant cnidarians to the nine vertebrate Nav1.x channel genes plus Nax. A close kinship between Nav2 and Nav1 homologs is evident in the sharing of most (twenty) intron splice junctions. Different metazoan groups have lost their Nav1 channel genes altogether, while vertebrates rapidly expanded their gene numbers. The expansion in vertebrate Nav1 channel genes fills unique functional niches and generates overlapping properties contributing to redundancies. Specific nervous system adaptations include cytoplasmic linkers with phosphorylation sites and tethered elements to protein assemblies in First Initial Segments and nodes of Ranvier. Analogous accessory beta subunit appeared alongside Nav1 channels within different animal sub-phyla. Nav1 channels contribute to pace-making as persistent or resurgent currents, the former which is widespread across animals, while the latter is a likely vertebrate adaptation.
Collapse
Affiliation(s)
- Julia E Fux
- Department of Biology, University of Waterloo, Waterloo, ON, Canada
| | - Amrit Mehta
- Department of Biology, University of Waterloo, Waterloo, ON, Canada
| | - Jack Moffat
- Department of Biology, University of Waterloo, Waterloo, ON, Canada
| | - J David Spafford
- Department of Biology, University of Waterloo, Waterloo, ON, Canada
| |
Collapse
|
41
|
Verkerk AO, Amin AS, Remme CA. Disease Modifiers of Inherited SCN5A Channelopathy. Front Cardiovasc Med 2018; 5:137. [PMID: 30327767 PMCID: PMC6174200 DOI: 10.3389/fcvm.2018.00137] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 09/10/2018] [Indexed: 12/19/2022] Open
Abstract
To date, a large number of mutations in SCN5A, the gene encoding the pore-forming α-subunit of the primary cardiac Na+ channel (NaV1.5), have been found in patients presenting with a wide range of ECG abnormalities and cardiac syndromes. Although these mutations all affect the same NaV1.5 channel, the associated cardiac syndromes each display distinct phenotypical and biophysical characteristics. Variable disease expressivity has also been reported, where one particular mutation in SCN5A may lead to either one particular symptom, a range of various clinical signs, or no symptoms at all, even within one single family. Additionally, disease severity may vary considerably between patients carrying the same mutation. The exact reasons are unknown, but evidence is increasing that various cardiac and non-cardiac conditions can influence the expressivity and severity of inherited SCN5A channelopathies. In this review, we provide a summary of identified disease entities caused by SCN5A mutations, and give an overview of co-morbidities and other (non)-genetic factors which may modify SCN5A channelopathies. A comprehensive knowledge of these modulatory factors is not only essential for a complete understanding of the diverse clinical phenotypes associated with SCN5A mutations, but also for successful development of effective risk stratification and (alternative) treatment paradigms.
Collapse
Affiliation(s)
- Arie O Verkerk
- Department of Clinical and Experimental Cardiology, Heart Centre, Academic Medical Center, Amsterdam, Netherlands.,Department of Medical Biology, Academic Medical Center, Amsterdam, Netherlands
| | - Ahmad S Amin
- Department of Clinical and Experimental Cardiology, Heart Centre, Academic Medical Center, Amsterdam, Netherlands
| | - Carol Ann Remme
- Department of Clinical and Experimental Cardiology, Heart Centre, Academic Medical Center, Amsterdam, Netherlands
| |
Collapse
|
42
|
Smith RS, Kenny CJ, Ganesh V, Jang A, Borges-Monroy R, Partlow JN, Hill RS, Shin T, Chen AY, Doan RN, Anttonen AK, Ignatius J, Medne L, Bönnemann CG, Hecht JL, Salonen O, Barkovich AJ, Poduri A, Wilke M, de Wit MCY, Mancini GMS, Sztriha L, Im K, Amrom D, Andermann E, Paetau R, Lehesjoki AE, Walsh CA, Lehtinen MK. Sodium Channel SCN3A (Na V1.3) Regulation of Human Cerebral Cortical Folding and Oral Motor Development. Neuron 2018; 99:905-913.e7. [PMID: 30146301 DOI: 10.1016/j.neuron.2018.07.052] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 06/05/2018] [Accepted: 07/30/2018] [Indexed: 12/29/2022]
Abstract
Channelopathies are disorders caused by abnormal ion channel function in differentiated excitable tissues. We discovered a unique neurodevelopmental channelopathy resulting from pathogenic variants in SCN3A, a gene encoding the voltage-gated sodium channel NaV1.3. Pathogenic NaV1.3 channels showed altered biophysical properties including increased persistent current. Remarkably, affected individuals showed disrupted folding (polymicrogyria) of the perisylvian cortex of the brain but did not typically exhibit epilepsy; they presented with prominent speech and oral motor dysfunction, implicating SCN3A in prenatal development of human cortical language areas. The development of this disorder parallels SCN3A expression, which we observed to be highest early in fetal cortical development in progenitor cells of the outer subventricular zone and cortical plate neurons and decreased postnatally, when SCN1A (NaV1.1) expression increased. Disrupted cerebral cortical folding and neuronal migration were recapitulated in ferrets expressing the mutant channel, underscoring the unexpected role of SCN3A in progenitor cells and migrating neurons.
Collapse
Affiliation(s)
- Richard S Smith
- Division of Genetics and Genomics, Manton Center for Orphan Disease Research, and Howard Hughes Medical Institute, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Connor J Kenny
- Division of Genetics and Genomics, Manton Center for Orphan Disease Research, and Howard Hughes Medical Institute, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Vijay Ganesh
- Division of Genetics and Genomics, Manton Center for Orphan Disease Research, and Howard Hughes Medical Institute, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Ahram Jang
- Department of Pathology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Rebeca Borges-Monroy
- Division of Genetics and Genomics, Manton Center for Orphan Disease Research, and Howard Hughes Medical Institute, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Jennifer N Partlow
- Division of Genetics and Genomics, Manton Center for Orphan Disease Research, and Howard Hughes Medical Institute, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - R Sean Hill
- Division of Genetics and Genomics, Manton Center for Orphan Disease Research, and Howard Hughes Medical Institute, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Taehwan Shin
- Division of Genetics and Genomics, Manton Center for Orphan Disease Research, and Howard Hughes Medical Institute, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Allen Y Chen
- Division of Genetics and Genomics, Manton Center for Orphan Disease Research, and Howard Hughes Medical Institute, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Ryan N Doan
- Division of Genetics and Genomics, Manton Center for Orphan Disease Research, and Howard Hughes Medical Institute, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Anna-Kaisa Anttonen
- The Folkhälsan Institute of Genetics, 00290 Helsinki, Finland; Medical and Clinical Genetics, Neuroscience Center and Research Programs Unit, Molecular Neurology, 00014, University of Helsinki, Helsinki, Finland
| | - Jaakko Ignatius
- Department of Clinical Genetics, Turku University Hospital, Turku, 20521, Finland
| | - Livija Medne
- Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Carsten G Bönnemann
- Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Jonathan L Hecht
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA 02115, USA
| | - Oili Salonen
- Medical Imaging Center, Radiology, University of Helsinki and Helsinki University Hospital, 00029 HUS, Helsinki, Finland
| | - A James Barkovich
- Benioff Children's Hospital, Departments of Radiology, Pediatrics, Neurology, and Neurological Surgery, University of California San Francisco, San Francisco, CA 94117, USA
| | - Annapurna Poduri
- Department of Neurology, Boston Children's Hospital and Department of Neurology, Harvard Medical School, Boston, MA 02115, USA
| | - Martina Wilke
- Department of Clinical Genetics, Erasmus MC Rotterdam 3015CN, Netherlands
| | - Marie Claire Y de Wit
- Neurogenetics Joint Clinic in Sophia Children's Hospital, Erasmus MC Rotterdam 3015CN, Netherlands
| | - Grazia M S Mancini
- Department of Clinical Genetics, Erasmus MC Rotterdam 3015CN, Netherlands
| | - Laszlo Sztriha
- Department of Pediatrics, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
| | - Kiho Im
- Division of Newborn Medicine, Boston Children's Hospital and Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Dina Amrom
- Neurogenetics Unit and Epilepsy Research Group, Montreal Neurological Institute and Hospital; and the Departments of Neurology & Neurosurgery and Human Genetics, McGill University, Montreal, QC H3A 2B4, Canada
| | - Eva Andermann
- Neurogenetics Unit and Epilepsy Research Group, Montreal Neurological Institute and Hospital; and the Departments of Neurology & Neurosurgery and Human Genetics, McGill University, Montreal, QC H3A 2B4, Canada
| | - Ritva Paetau
- Children's Hospital, University of Helsinki and Helsinki University Hospital, 00029 HUS, Helsinki, Finland
| | - Anna-Elina Lehesjoki
- The Folkhälsan Institute of Genetics, 00290 Helsinki, Finland; Medical and Clinical Genetics, Neuroscience Center and Research Programs Unit, Molecular Neurology, 00014, University of Helsinki, Helsinki, Finland
| | - Christopher A Walsh
- Division of Genetics and Genomics, Manton Center for Orphan Disease Research, and Howard Hughes Medical Institute, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | - Maria K Lehtinen
- Department of Pathology, Boston Children's Hospital, Boston, MA 02115, USA.
| |
Collapse
|
43
|
Ali SR, Liu Z, Nenov MN, Folorunso O, Singh A, Scala F, Chen H, James TF, Alshammari M, Panova-Elektronova NI, White MA, Zhou J, Laezza F. Functional Modulation of Voltage-Gated Sodium Channels by a FGF14-Based Peptidomimetic. ACS Chem Neurosci 2018; 9:976-987. [PMID: 29359916 DOI: 10.1021/acschemneuro.7b00399] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Protein-protein interactions (PPI) offer unexploited opportunities for CNS drug discovery and neurochemical probe development. Here, we present ZL181, a novel peptidomimetic targeting the PPI interface of the voltage-gated Na+ channel Nav1.6 and its regulatory protein fibroblast growth factor 14 (FGF14). ZL181 binds to FGF14 and inhibits its interaction with the Nav1.6 channel C-tail. In HEK-Nav1.6 expressing cells, ZL181 acts synergistically with FGF14 to suppress Nav1.6 current density and to slow kinetics of fast inactivation, but antagonizes FGF14 modulation of steady-state inactivation that is regulated by the N-terminal tail of the protein. In medium spiny neurons in the nucleus accumbens, ZL181 suppresses excitability by a mechanism that is dependent upon expression of FGF14 and is consistent with a state-dependent inhibition of FGF14. Overall, ZL181 and derivatives could lay the ground for developing allosteric modulators of Nav channels that are of interest for a broad range of CNS disorders.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Musaad Alshammari
- King Saud University Graduate Studies Abroad Program, King Saud University, Riyadh, Saudi Arabia
| | | | | | | | | |
Collapse
|
44
|
Erickson A, Deiteren A, Harrington AM, Garcia‐Caraballo S, Castro J, Caldwell A, Grundy L, Brierley SM. Voltage-gated sodium channels: (Na V )igating the field to determine their contribution to visceral nociception. J Physiol 2018; 596:785-807. [PMID: 29318638 PMCID: PMC5830430 DOI: 10.1113/jp273461] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 01/02/2018] [Indexed: 12/19/2022] Open
Abstract
Chronic visceral pain, altered motility and bladder dysfunction are common, yet poorly managed symptoms of functional and inflammatory disorders of the gastrointestinal and urinary tracts. Recently, numerous human channelopathies of the voltage-gated sodium (NaV ) channel family have been identified, which induce either painful neuropathies, an insensitivity to pain, or alterations in smooth muscle function. The identification of these disorders, in addition to the recent utilisation of genetically modified NaV mice and specific NaV channel modulators, has shed new light on how NaV channels contribute to the function of neuronal and non-neuronal tissues within the gastrointestinal tract and bladder. Here we review the current pre-clinical and clinical evidence to reveal how the nine NaV channel family members (NaV 1.1-NaV 1.9) contribute to abdominal visceral function in normal and disease states.
Collapse
Affiliation(s)
- Andelain Erickson
- Visceral Pain Research Group, Human Physiology, Centre for Neuroscience, College of Medicine and Public HealthFlinders UniversityBedford ParkSouth Australia5042Australia
- Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, University of AdelaideSouth Australian Health and Medical Research Institute (SAHMRI)North TerraceAdelaideSouth Australia 5000Australia
| | - Annemie Deiteren
- Visceral Pain Research Group, Human Physiology, Centre for Neuroscience, College of Medicine and Public HealthFlinders UniversityBedford ParkSouth Australia5042Australia
- Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, University of AdelaideSouth Australian Health and Medical Research Institute (SAHMRI)North TerraceAdelaideSouth Australia 5000Australia
| | - Andrea M. Harrington
- Visceral Pain Research Group, Human Physiology, Centre for Neuroscience, College of Medicine and Public HealthFlinders UniversityBedford ParkSouth Australia5042Australia
- Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, University of AdelaideSouth Australian Health and Medical Research Institute (SAHMRI)North TerraceAdelaideSouth Australia 5000Australia
| | - Sonia Garcia‐Caraballo
- Visceral Pain Research Group, Human Physiology, Centre for Neuroscience, College of Medicine and Public HealthFlinders UniversityBedford ParkSouth Australia5042Australia
- Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, University of AdelaideSouth Australian Health and Medical Research Institute (SAHMRI)North TerraceAdelaideSouth Australia 5000Australia
| | - Joel Castro
- Visceral Pain Research Group, Human Physiology, Centre for Neuroscience, College of Medicine and Public HealthFlinders UniversityBedford ParkSouth Australia5042Australia
- Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, University of AdelaideSouth Australian Health and Medical Research Institute (SAHMRI)North TerraceAdelaideSouth Australia 5000Australia
| | - Ashlee Caldwell
- Visceral Pain Research Group, Human Physiology, Centre for Neuroscience, College of Medicine and Public HealthFlinders UniversityBedford ParkSouth Australia5042Australia
- Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, University of AdelaideSouth Australian Health and Medical Research Institute (SAHMRI)North TerraceAdelaideSouth Australia 5000Australia
| | - Luke Grundy
- Visceral Pain Research Group, Human Physiology, Centre for Neuroscience, College of Medicine and Public HealthFlinders UniversityBedford ParkSouth Australia5042Australia
- Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, University of AdelaideSouth Australian Health and Medical Research Institute (SAHMRI)North TerraceAdelaideSouth Australia 5000Australia
| | - Stuart M. Brierley
- Visceral Pain Research Group, Human Physiology, Centre for Neuroscience, College of Medicine and Public HealthFlinders UniversityBedford ParkSouth Australia5042Australia
- Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, University of AdelaideSouth Australian Health and Medical Research Institute (SAHMRI)North TerraceAdelaideSouth Australia 5000Australia
| |
Collapse
|
45
|
Wu Y, Ma H, Zhang F, Zhang C, Zou X, Cao Z. Selective Voltage-Gated Sodium Channel Peptide Toxins from Animal Venom: Pharmacological Probes and Analgesic Drug Development. ACS Chem Neurosci 2018; 9:187-197. [PMID: 29161016 DOI: 10.1021/acschemneuro.7b00406] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Voltage-gated sodium channels (Navs) play critical roles in action potential generation and propagation. Nav channelopathy as well as pathological sensitization contribute to allodynia and hyperalgesia. Recent evidence has demonstrated the significant roles of Nav subtypes (Nav1.3, 1.7, 1.8, and 1.9) in nociceptive transduction, and therefore these Navs may represent attractive targets for analgesic drug discovery. Animal toxins are structurally diverse peptides that are highly potent yet selective on ion channel subtypes and therefore represent valuable probes to elucidate the structures, gating properties, and cellular functions of ion channels. In this review, we summarize recent advances on peptide toxins from animal venom that selectively target Nav1.3, 1.7, 1.8, and 1.9, along with their potential in analgesic drug discovery.
Collapse
Affiliation(s)
- Ying Wu
- Jiangsu Provincial Key Laboratory for TCM Evaluation
and Translational Development, China Pharmaceutical University, Nanjing 211198, China
| | - Hui Ma
- Jiangsu Provincial Key Laboratory for TCM Evaluation
and Translational Development, China Pharmaceutical University, Nanjing 211198, China
| | - Fan Zhang
- Jiangsu Provincial Key Laboratory for TCM Evaluation
and Translational Development, China Pharmaceutical University, Nanjing 211198, China
| | - Chunlei Zhang
- Jiangsu Provincial Key Laboratory for TCM Evaluation
and Translational Development, China Pharmaceutical University, Nanjing 211198, China
| | - Xiaohan Zou
- Jiangsu Provincial Key Laboratory for TCM Evaluation
and Translational Development, China Pharmaceutical University, Nanjing 211198, China
| | - Zhengyu Cao
- Jiangsu Provincial Key Laboratory for TCM Evaluation
and Translational Development, China Pharmaceutical University, Nanjing 211198, China
| |
Collapse
|
46
|
Savio-Galimberti E, Argenziano M, Antzelevitch C. Cardiac Arrhythmias Related to Sodium Channel Dysfunction. Handb Exp Pharmacol 2018; 246:331-354. [PMID: 28965168 DOI: 10.1007/164_2017_43] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
The voltage-gated cardiac sodium channel (Nav1.5) is a mega-complex comprised of a pore-forming α subunit and 4 ancillary β-subunits together with numerous protein partners. Genetic defects in the form of rare variants in one or more sodium channel-related genes can cause a loss- or gain-of-function of sodium channel current (INa) leading to the manifestation of various disease phenotypes, including Brugada syndrome, long QT syndrome, progressive cardiac conduction disease, sick sinus syndrome, multifocal ectopic Purkinje-related premature contractions, and atrial fibrillation. Some sodium channelopathies have also been shown to be responsible for sudden infant death syndrome (SIDS). Although these genetic defects often present as pure electrical diseases, recent studies point to a contribution of structural abnormalities to the electrocardiographic and arrhythmic manifestation in some cases, such as dilated cardiomyopathy. The same rare variants in SCN5A or related genes may present with different clinical phenotypes in different individuals and sometimes in members of the same family. Genetic background and epigenetic and environmental factors contribute to the expression of these overlap syndromes. Our goal in this chapter is to review and discuss what is known about the clinical phenotype and genotype of each cardiac sodium channelopathy, and to briefly discuss the underlying mechanisms.
Collapse
Affiliation(s)
| | - Mariana Argenziano
- Lankenau Institute for Medical Research, 100 E. Lancaster Avenue, Wynnewood, PA, 19096, USA
| | - Charles Antzelevitch
- Lankenau Institute for Medical Research, 100 E. Lancaster Avenue, Wynnewood, PA, 19096, USA.
| |
Collapse
|
47
|
Chen X, Sun Y, Huang H, Liu W, Hu P, Huang X, Zou F, Liu J. Uncovering the proteome response of murine neuroblastoma cells against low-dose exposure to saxitoxin. Toxicol Mech Methods 2017; 28:335-344. [DOI: 10.1080/15376516.2017.1411413] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Xiao Chen
- Key Laboratory of Modern Toxicology of Shenzhen, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
- Department of Occupational Health and Occupational Medicine, School of Public Health, Southern Medical University, Guangzhou, China
| | - Ye Sun
- Key Laboratory of Modern Toxicology of Shenzhen, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Haiyan Huang
- Key Laboratory of Modern Toxicology of Shenzhen, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Wei Liu
- Key Laboratory of Modern Toxicology of Shenzhen, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Panpan Hu
- Key Laboratory of Modern Toxicology of Shenzhen, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Xinfeng Huang
- Key Laboratory of Modern Toxicology of Shenzhen, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Fei Zou
- Department of Occupational Health and Occupational Medicine, School of Public Health, Southern Medical University, Guangzhou, China
| | - Jianjun Liu
- Key Laboratory of Modern Toxicology of Shenzhen, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| |
Collapse
|
48
|
Estebe JP. Intravenous lidocaine. Best Pract Res Clin Anaesthesiol 2017; 31:513-521. [DOI: 10.1016/j.bpa.2017.05.005] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 05/13/2017] [Accepted: 05/23/2017] [Indexed: 12/14/2022]
|
49
|
Oz M, El Nebrisi EG, Yang KHS, Howarth FC, Al Kury LT. Cellular and Molecular Targets of Menthol Actions. Front Pharmacol 2017; 8:472. [PMID: 28769802 PMCID: PMC5513973 DOI: 10.3389/fphar.2017.00472] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 07/03/2017] [Indexed: 02/04/2023] Open
Abstract
Menthol belongs to monoterpene class of a structurally diverse group of phytochemicals found in plant-derived essential oils. Menthol is widely used in pharmaceuticals, confectionary, oral hygiene products, pesticides, cosmetics, and as a flavoring agent. In addition, menthol is known to have antioxidant, anti-inflammatory, and analgesic effects. Recently, there has been renewed awareness in comprehending the biological and pharmacological effects of menthol. TRP channels have been demonstrated to mediate the cooling actions of menthol. There has been new evidence demonstrating that menthol can significantly influence the functional characteristics of a number of different kinds of ligand and voltage-gated ion channels, indicating that at least some of the biological and pharmacological effects of menthol can be mediated by alterations in cellular excitability. In this article, we examine the results of earlier studies on the actions of menthol with voltage and ligand-gated ion channels.
Collapse
Affiliation(s)
- Murat Oz
- Department of Pharmacology, College of Medicine and Health Sciences, United Arab Emirates UniversityAl Ain, United Arab Emirates.,Department of Basic Medical Sciences, College of Medicine, Qatar UniversityDoha, Qatar
| | - Eslam G El Nebrisi
- Department of Pharmacology, College of Medicine and Health Sciences, United Arab Emirates UniversityAl Ain, United Arab Emirates
| | - Keun-Hang S Yang
- Department of Biological Sciences, Schmid College of Science and Technology, Chapman UniversityOrange, CA, United States
| | - Frank C Howarth
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates UniversityAl Ain, United Arab Emirates
| | - Lina T Al Kury
- Department of Health Sciences, College of Natural and Health Sciences, Zayed UniversityAbu Dhabi, United Arab Emirates
| |
Collapse
|
50
|
Duménieu M, Oulé M, Kreutz MR, Lopez-Rojas J. The Segregated Expression of Voltage-Gated Potassium and Sodium Channels in Neuronal Membranes: Functional Implications and Regulatory Mechanisms. Front Cell Neurosci 2017; 11:115. [PMID: 28484374 PMCID: PMC5403416 DOI: 10.3389/fncel.2017.00115] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 04/05/2017] [Indexed: 01/25/2023] Open
Abstract
Neurons are highly polarized cells with apparent functional and morphological differences between dendrites and axon. A critical determinant for the molecular and functional identity of axonal and dendritic segments is the restricted expression of voltage-gated ion channels (VGCs). Several studies show an uneven distribution of ion channels and their differential regulation within dendrites and axons, which is a prerequisite for an appropriate integration of synaptic inputs and the generation of adequate action potential (AP) firing patterns. This review article will focus on the signaling pathways leading to segmented expression of voltage-gated potassium and sodium ion channels at the neuronal plasma membrane and the regulatory mechanisms ensuring segregated functions. We will also discuss the relevance of proper ion channel targeting for neuronal physiology and how alterations in polarized distribution contribute to neuronal pathology.
Collapse
Affiliation(s)
- Maël Duménieu
- Research Group Neuroplasticity, Leibniz Institute for NeurobiologyMagdeburg, Germany
| | - Marie Oulé
- Research Group Neuroplasticity, Leibniz Institute for NeurobiologyMagdeburg, Germany
| | - Michael R Kreutz
- Research Group Neuroplasticity, Leibniz Institute for NeurobiologyMagdeburg, Germany.,Leibniz Group "Dendritic Organelles and Synaptic Function", University Medical Center Hamburg-Eppendorf, Center for Molecular Neurobiology (ZMNH)Hamburg, Germany
| | - Jeffrey Lopez-Rojas
- Research Group Neuroplasticity, Leibniz Institute for NeurobiologyMagdeburg, Germany
| |
Collapse
|