51
|
Gammon DW, Liu Z, Chandrasekaran A, El‐Naggar SF, Kuryshev YA, Jackson S. Pyrethroid neurotoxicity studies with bifenthrin indicate a mixed Type I/II mode of action. PEST MANAGEMENT SCIENCE 2019; 75:1190-1197. [PMID: 30548793 PMCID: PMC6590159 DOI: 10.1002/ps.5300] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 11/25/2018] [Accepted: 12/07/2018] [Indexed: 05/13/2023]
Abstract
BACKGROUND Bifenthrin is usually considered a Type I pyrethroid, because it lacks an α-CN group present in Type II pyrethroids, but some previous studies suggest a mixed Type I/II mode-of-action. Results are presented for bifenthrin in a rat developmental neurotoxicity (DNT) study along with effects on Na currents in human VGSC subtypes. Molecular modeling comparisons were also made for bifenthrin and other pyrethroids. RESULTS In a rat DNT study, bifenthrin produced tremors and clonic convulsions in dams and pups and slightly reduced acoustic startle response amplitude, and increased Tmax, at PND20 in females. Similar blood levels of bifenthrin were measured in dams and pups at each dose level i.e. no concentration in pups. In human VGSC experiments, using the Nav1.8 subtype, bifenthrin's effects on inactivation were slight, as for Type II pyrethroids, but without large prolongation of the tail current (deactivation) seen with Type II. Molecular modeling of bifenthrin indicates that the o-Me group may occupy a similar space to the α-CN group of cypermethrin and fenpropathrin. CONCLUSION In a DNT study and on human Nav1.8 tail currents bifenthrin showed Type I and II effects, similar to some published studies. Overall, bifenthrin acts as a mixed Type I/II pyrethroid. © 2018 The Authors. Pest Management Science published by John Wiley & Sons Ltd on behalf of Society of Chemical Industry.
Collapse
Affiliation(s)
- Derek W Gammon
- FMC Agricultural SolutionsGlobal Regulatory SciencesEwingNJUSA
| | - Zhiwei Liu
- FMC Agricultural SolutionsGlobal Regulatory SciencesEwingNJUSA
| | | | | | - Yuri A Kuryshev
- FMC Agricultural SolutionsGlobal Regulatory SciencesEwingNJUSA
| | - Sharon Jackson
- FMC Agricultural SolutionsGlobal Regulatory SciencesEwingNJUSA
| |
Collapse
|
52
|
Maatuf Y, Geron M, Priel A. The Role of Toxins in the Pursuit for Novel Analgesics. Toxins (Basel) 2019; 11:toxins11020131. [PMID: 30813430 PMCID: PMC6409898 DOI: 10.3390/toxins11020131] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 02/17/2019] [Accepted: 02/20/2019] [Indexed: 12/19/2022] Open
Abstract
Chronic pain is a major medical issue which reduces the quality of life of millions and inflicts a significant burden on health authorities worldwide. Currently, management of chronic pain includes first-line pharmacological therapies that are inadequately effective, as in just a portion of patients pain relief is obtained. Furthermore, most analgesics in use produce severe or intolerable adverse effects that impose dose restrictions and reduce compliance. As the majority of analgesic agents act on the central nervous system (CNS), it is possible that blocking pain at its source by targeting nociceptors would prove more efficient with minimal CNS-related side effects. The development of such analgesics requires the identification of appropriate molecular targets and thorough understanding of their structural and functional features. To this end, plant and animal toxins can be employed as they affect ion channels with high potency and selectivity. Moreover, elucidation of the toxin-bound ion channel structure could generate pharmacophores for rational drug design while favorable safety and analgesic profiles could highlight toxins as leads or even as valuable therapeutic compounds themselves. Here, we discuss the use of plant and animal toxins in the characterization of peripherally expressed ion channels which are implicated in pain.
Collapse
Affiliation(s)
- Yossi Maatuf
- The Institute for Drug Research (IDR), School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel.
| | - Matan Geron
- The Institute for Drug Research (IDR), School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel.
| | - Avi Priel
- The Institute for Drug Research (IDR), School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel.
| |
Collapse
|
53
|
Zhou C, Johnson KW, Herold KF, Hemmings HC. Differential Inhibition of Neuronal Sodium Channel Subtypes by the General Anesthetic Isoflurane. J Pharmacol Exp Ther 2019; 369:200-211. [PMID: 30792243 DOI: 10.1124/jpet.118.254938] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Accepted: 02/19/2019] [Indexed: 02/05/2023] Open
Abstract
Volatile anesthetics depress neurotransmitter release in a brain region- and neurotransmitter-selective manner by unclear mechanisms. Voltage-gated sodium channels (Navs), which are coupled to synaptic vesicle exocytosis, are inhibited by volatile anesthetics through reduction of peak current and modulation of gating. Subtype-selective effects of anesthetics on Nav might contribute to observed neurotransmitter-selective anesthetic effects on release. We analyzed anesthetic effects on Na+ currents mediated by the principal neuronal Nav subtypes Nav1.1, Nav1.2, and Nav1.6 heterologously expressed in ND7/23 neuroblastoma cells using whole-cell patch-clamp electrophysiology. Isoflurane at clinically relevant concentrations induced a hyperpolarizing shift in the voltage dependence of steady-state inactivation and slowed recovery from fast inactivation in all three Nav subtypes, with the voltage of half-maximal steady-state inactivation significantly more positive for Nav1.1 (-49.7 ± 3.9 mV) than for Nav1.2 (-57.5 ± 1.2 mV) or Nav1.6 (-58.0 ± 3.8 mV). Isoflurane significantly inhibited peak Na+ current (I Na) in a voltage-dependent manner: at a physiologically relevant holding potential of -70 mV, isoflurane inhibited peak I Na of Nav1.2 (16.5% ± 5.5%) and Nav1.6 (18.0% ± 7.8%), but not of Nav1.1 (1.2% ± 0.8%). Since Nav subtypes are differentially expressed both between neuronal types and within neurons, greater inhibition of Nav1.2 and Nav1.6 compared with Nav1.1 could contribute to neurotransmitter-selective effects of isoflurane on synaptic transmission.
Collapse
Affiliation(s)
- Cheng Zhou
- Departments of Anesthesiology (C.Z., K.W.J., K.F.H., H.C.H.) and Pharmacology (H.C.H.), Weill Cornell Medicine, New York, New York; and Laboratory of Anesthesia and Critical Care Medicine, Translational Neuroscience Center, West China Hospital of Sichuan University, Chengdu, Sichuan, People's Republic of China (C.Z.)
| | - Kenneth W Johnson
- Departments of Anesthesiology (C.Z., K.W.J., K.F.H., H.C.H.) and Pharmacology (H.C.H.), Weill Cornell Medicine, New York, New York; and Laboratory of Anesthesia and Critical Care Medicine, Translational Neuroscience Center, West China Hospital of Sichuan University, Chengdu, Sichuan, People's Republic of China (C.Z.)
| | - Karl F Herold
- Departments of Anesthesiology (C.Z., K.W.J., K.F.H., H.C.H.) and Pharmacology (H.C.H.), Weill Cornell Medicine, New York, New York; and Laboratory of Anesthesia and Critical Care Medicine, Translational Neuroscience Center, West China Hospital of Sichuan University, Chengdu, Sichuan, People's Republic of China (C.Z.)
| | - Hugh C Hemmings
- Departments of Anesthesiology (C.Z., K.W.J., K.F.H., H.C.H.) and Pharmacology (H.C.H.), Weill Cornell Medicine, New York, New York; and Laboratory of Anesthesia and Critical Care Medicine, Translational Neuroscience Center, West China Hospital of Sichuan University, Chengdu, Sichuan, People's Republic of China (C.Z.)
| |
Collapse
|
54
|
Aiba I, Noebels JL. Adrenergic agonist induces rhythmic firing in quiescent cardiac preganglionic neurons in nucleus ambiguous via activation of intrinsic membrane excitability. J Neurophysiol 2019; 121:1266-1278. [PMID: 30699052 PMCID: PMC6485744 DOI: 10.1152/jn.00761.2018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Cholinergic vagal nerves projecting from neurons in the brain stem nucleus ambiguus (NAm) play a predominant role in cardiac parasympathetic pacemaking control. Central adrenergic signaling modulates the tone of this vagal output; however, the exact excitability mechanisms are not fully understood. We investigated responses of NAm neurons to adrenergic agonists using in vitro mouse brain stem slices. Preganglionic NAm neurons were identified by ChAT-tdTomato fluorescence in young adult transgenic mice, and their cardiac projection was confirmed by retrograde dye tracing. Juxtacellular recordings detected sparse or absent spontaneous action potentials (AP) in NAm neurons. However, bath application of epinephrine or norepinephrine strongly and reversibly activated most NAm neurons regardless of their basal firing rate. Epinephrine was more potent than norepinephrine, and this activation largely depends on α1-adrenoceptors. Interestingly, adrenergic activation of NAm neurons does not require an ionotropic synaptic mechanism, because postsynaptic excitatory or inhibitory receptor blockade did not occlude the excitatory effect, and bath-applied adrenergic agonists did not alter excitatory or inhibitory synaptic transmission. Instead, adrenergic agonists significantly elevated intrinsic membrane excitability to facilitate generation of recurrent action potentials. T-type calcium current and hyperpolarization-activated current are involved in this excitation pattern, although not required for spontaneous AP induction by epinephrine. In contrast, pharmacological blockade of persistent sodium current significantly inhibited the adrenergic effects. Our results demonstrate that central adrenergic signaling enhances the intrinsic excitability of NAm neurons and that persistent sodium current is required for this effect. This central balancing mechanism may counteract excessive peripheral cardiac excitation during increased sympathetic tone. NEW & NOTEWORTHY Cardiac preganglionic cholinergic neurons in the nucleus ambiguus (NAm) are responsible for slowing cardiac pacemaking. This study identified that adrenergic agonists can induce rhythmic action potentials in otherwise quiescent cholinergic NAm preganglionic neurons in brain stem slice preparation. The modulatory influence of adrenaline on central parasympathetic outflow may contribute to both physiological and deleterious cardiovascular regulation.
Collapse
Affiliation(s)
- Isamu Aiba
- Department of Neurology, Baylor College of Medicine , Houston, Texas
| | - Jeffrey L Noebels
- Department of Neurology, Baylor College of Medicine , Houston, Texas
| |
Collapse
|
55
|
Lin JW, Yu F, Müller WS, Ehnholm G, Okada Y. Focused ultrasound transiently increases membrane conductance in isolated crayfish axon. J Neurophysiol 2018; 121:480-489. [PMID: 30565960 DOI: 10.1152/jn.00541.2018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
We report a novel phenomenon produced by focused ultrasound (US) that may be important for understanding its effects on cell membranes. When a US burst (2.1 MHz, 1-mm focal diameter, 0.1-1 MPa) was focused on a motor axon of the crayfish neuromuscular junction, it consistently produced a fast hyperpolarization, which was followed or superseded by subthreshold depolarizations or action potentials in a stochastic manner. The depolarization persisted in the presence of voltage-gated channel blockers [1 µM TTX ( INa), 50 µM ZD7288 ( Ih), and 200 µM 4-aminopyridine ( IK)] and typically started shortly after the onset of a 5-ms US burst, with a mean latency of 3.35 ± 0.53 ms (SE). The duration and amplitude of depolarizations averaged 2.13 ± 0.87 s and 10.1 ± 2.09 mV, with a maximum of 200 s and 60 mV, respectively. The US-induced depolarization was always associated with a decrease in membrane resistance. By measuring membrane potential and resistance during the US-induced depolarization, the reversal potential of US-induced conductance ( gus) was estimated to be -8.4 ± 2.3 mV, suggesting a nonselective conductance. The increase in gus was 10-100 times larger than the leak conductance; thus it could significantly influence neuronal activity. This change in conductance may be due to stimulation of mechanoreceptors. Alternatively, US may perturb the lateral motion of phospholipids and produce nanopores, which then increase gus. These results may be important for understanding mechanisms underlying US-mediated modulation of neuronal activity and brain function. NEW & NOTEWORTHY We report a specific increase in membrane conductance produced by ultrasound (US) on neuronal membrane. When a 5-ms US tone burst was focused on a crayfish motor axon, it stochastically triggered either depolarization or a spike train. The depolarization was up to 60 mV in amplitude and 200 s in duration and therefore could significantly influence neuronal activity. Depolarization was still evoked by US burst in the presence of Na+ and Ca2+ channel blockers and had a reversal potential of -8.4 ± 2.3 mV, suggesting a nonselective permeability. US can be applied noninvasively in the form of a focused beam to deep brain areas through the skull and has been shown to modulate brain activity. Understanding the depolarization reported here should be helpful for improving the use of US for noninvasive modulation and stimulation in brain-related disease.
Collapse
Affiliation(s)
- Jen-Wei Lin
- Department of Biology, Boston University , Boston, Massachusetts
| | - Feiyuan Yu
- Department of Biology, Boston University , Boston, Massachusetts
| | - Wolfgang S Müller
- Division of Newborn Medicine, Department of Medicine, Boston Children's Hospital and Harvard Medical School , Boston, Massachusetts
| | - Gösta Ehnholm
- Department of Neuroscience and Biomedical Engineering, Aalto University, Aalto, Finland
| | - Yoshio Okada
- Division of Newborn Medicine, Department of Medicine, Boston Children's Hospital and Harvard Medical School , Boston, Massachusetts
| |
Collapse
|
56
|
Meredith FL, Rennie KJ. Regional and Developmental Differences in Na + Currents in Vestibular Primary Afferent Neurons. Front Cell Neurosci 2018; 12:423. [PMID: 30487736 PMCID: PMC6246661 DOI: 10.3389/fncel.2018.00423] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 10/29/2018] [Indexed: 02/04/2023] Open
Abstract
The vestibular system relays information about head position via afferent nerve fibers to the brain in the form of action potentials. Voltage-gated Na+ channels in vestibular afferents drive the initiation and propagation of action potentials, but their expression during postnatal development and their contributions to firing in diverse mature afferent populations are unknown. Electrophysiological techniques were used to determine Na+ channel subunit types in vestibular calyx-bearing afferents at different stages of postnatal development. We used whole cell patch clamp recordings in thin slices of gerbil crista neuroepithelium to investigate Na+ channels and firing patterns in central zone (CZ) and peripheral zone (PZ) afferents. PZ afferents are exclusively dimorphic, innervating type I and type II hair cells, whereas CZ afferents can form dimorphs or calyx-only terminals which innervate type I hair cells alone. All afferents expressed tetrodotoxin (TTX)-sensitive Na+ currents, but TTX-sensitivity varied with age. During the fourth postnatal week, 200–300 nM TTX completely blocked sodium currents in PZ and CZ calyces. By contrast, in immature calyces [postnatal day (P) 5–11], a small component of peak sodium current remained in 200 nM TTX. Application of 1 μM TTX, or Jingzhaotoxin-III plus 200 nM TTX, abolished sodium current in immature calyces, suggesting the transient expression of voltage-gated sodium channel 1.5 (Nav1.5) during development. A similar TTX-insensitive current was found in early postnatal crista hair cells (P5–9) and constituted approximately one third of the total sodium current. The Nav1.6 channel blocker, 4,9-anhydrotetrodotoxin, reduced a component of sodium current in immature and mature calyces. At 100 nM 4,9-anhydrotetrodotoxin, peak sodium current was reduced on average by 20% in P5–14 calyces, by 37% in mature dimorphic PZ calyces, but by less than 15% in mature CZ calyx-only terminals. In mature PZ calyces, action potentials became shorter and broader in the presence of 4,9-anhydrotetrodotoxin implicating a role for Nav1.6 channels in firing in dimorphic afferents.
Collapse
Affiliation(s)
- Frances L Meredith
- Department of Otolaryngology, University of Colorado School of Medicine, Aurora, CO, United States
| | - Katherine J Rennie
- Department of Otolaryngology, University of Colorado School of Medicine, Aurora, CO, United States.,Department of Physiology & Biophysics, University of Colorado School of Medicine, Aurora, CO, United States
| |
Collapse
|
57
|
Gonçalves TC, Benoit E, Partiseti M, Servent D. The Na V1.7 Channel Subtype as an Antinociceptive Target for Spider Toxins in Adult Dorsal Root Ganglia Neurons. Front Pharmacol 2018; 9:1000. [PMID: 30233376 PMCID: PMC6131673 DOI: 10.3389/fphar.2018.01000] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 08/14/2018] [Indexed: 12/11/2022] Open
Abstract
Although necessary for human survival, pain may sometimes become pathologic if long-lasting and associated with alterations in its signaling pathway. Opioid painkillers are officially used to treat moderate to severe, and even mild, pain. However, the consequent strong and not so rare complications that occur, including addiction and overdose, combined with pain management costs, remain an important societal and economic concern. In this context, animal venom toxins represent an original source of antinociceptive peptides that mainly target ion channels (such as ASICs as well as TRP, CaV, KV and NaV channels) involved in pain transmission. The present review aims to highlight the NaV1.7 channel subtype as an antinociceptive target for spider toxins in adult dorsal root ganglia neurons. It will detail (i) the characteristics of these primary sensory neurons, the first ones in contact with pain stimulus and conveying the nociceptive message, (ii) the electrophysiological properties of the different NaV channel subtypes expressed in these neurons, with a particular attention on the NaV1.7 subtype, an antinociceptive target of choice that has been validated by human genetic evidence, and (iii) the features of spider venom toxins, shaped of inhibitory cysteine knot motif, that present high affinity for the NaV1.7 subtype associated with evidenced analgesic efficacy in animal models.
Collapse
Affiliation(s)
- Tânia C Gonçalves
- Sanofi R&D, Integrated Drug Discovery - High Content Biology, Paris, France.,Service d'Ingénierie Moléculaire des Protéines, CEA de Saclay, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Evelyne Benoit
- Service d'Ingénierie Moléculaire des Protéines, CEA de Saclay, Université Paris-Saclay, Gif-sur-Yvette, France.,Institut des Neurosciences Paris-Saclay, UMR CNRS/Université Paris-Sud 9197, Gif-sur-Yvette, France
| | - Michel Partiseti
- Sanofi R&D, Integrated Drug Discovery - High Content Biology, Paris, France
| | - Denis Servent
- Service d'Ingénierie Moléculaire des Protéines, CEA de Saclay, Université Paris-Saclay, Gif-sur-Yvette, France
| |
Collapse
|
58
|
Mata DOD, Tibery DV, Campos LA, Camargos TS, Peigneur S, Tytgat J, Schwartz EF. Subtype Specificity of β-Toxin Tf1a from Tityus fasciolatus in Voltage Gated Sodium Channels. Toxins (Basel) 2018; 10:toxins10090339. [PMID: 30131471 PMCID: PMC6162530 DOI: 10.3390/toxins10090339] [Citation(s) in RCA: 2] [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: 07/13/2018] [Revised: 08/10/2018] [Accepted: 08/16/2018] [Indexed: 11/25/2022] Open
Abstract
Scorpion venoms are a complex mixture of components. Among them the most important are peptides, which presents the capacity to interact and modulate several ion channel subtypes, including voltage-gated sodium channels (NaV). Screening the activity of scorpion toxins on different subtypes of NaV reveals the scope of modulatory activity and, in most cases, low channel selectivity. Until now there are approximately 60 scorpion toxins experimentally assayed on NaV channels. However, the molecular bases of interaction between scorpion toxins and NaV channels are not fully elucidated. The activity description of new scorpion toxins is crucial to enhance the predictive strength of the structural–function correlations of these NaV modulatory molecules. In the present work a new scorpion toxin (Tf1a) was purified from Tityus fasciolatus venom by RP-HPLC, and characterized using electrophysiological experiments on different types of voltage-gated sodium channels. Tf1a was able to modify the normal function of NaV tested, showing to be a typical β-NaScTx. Tf1a also demonstrated an unusual capability to alter the kinetics of NaV1.5.
Collapse
Affiliation(s)
- Daniel Oliveira da Mata
- Laboratório de Neurofarmacologia, Departamento de Ciências Biológicas, Universidade de Brasília, Brasília 70910-900, Distrito Federal, Brazil.
| | - Diogo Vieira Tibery
- Laboratório de Neurofarmacologia, Departamento de Ciências Biológicas, Universidade de Brasília, Brasília 70910-900, Distrito Federal, Brazil.
| | - Leandro Ambrósio Campos
- Laboratório de Neurofarmacologia, Departamento de Ciências Biológicas, Universidade de Brasília, Brasília 70910-900, Distrito Federal, Brazil.
| | - Thalita Soares Camargos
- Departamento de Ciências da Saúde, Centro Universitário UDF, Brasília 70390-045, Distrito Federal, Brazil.
| | - Steve Peigneur
- Toxicology and Pharmacology, Department of Pharmaceutical and Pharmacological Sciences, University of Leuven (KU Leuven), P.O. Box 922, Herestraat 49, 3000 Leuven, Belgium.
| | - Jan Tytgat
- Toxicology and Pharmacology, Department of Pharmaceutical and Pharmacological Sciences, University of Leuven (KU Leuven), P.O. Box 922, Herestraat 49, 3000 Leuven, Belgium.
| | - Elisabeth Ferroni Schwartz
- Laboratório de Neurofarmacologia, Departamento de Ciências Biológicas, Universidade de Brasília, Brasília 70910-900, Distrito Federal, Brazil.
| |
Collapse
|
59
|
Galindo J, Contreras M, Maldonado P, Torrealba F, Lagos N, Valdés JL. Long-lasting, reversible and non-neurotoxic inactivation of hippocampus activity induced by neosaxitoxin. J Neurosci Methods 2018; 308:197-204. [PMID: 30107206 DOI: 10.1016/j.jneumeth.2018.08.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 08/06/2018] [Accepted: 08/10/2018] [Indexed: 11/29/2022]
Abstract
BACKGROUND Neosaxitoxin (NeoSTX) and related paralytics shellfish toxins has been successfully used as local anesthetic and muscle relaxants to treat a variety of ailments. The primary mechanism of action of these toxins occurs by blocking voltage-gated sodium channels with compounds such as TTX, lidocaine, or derivatives. However, most of these non-classical sodium channel blockers act with a reduced time effect as well as ensuing neurotoxicity. NEW METHOD In this report, we show that the use of local NeoSTX injections inactivates the hippocampal neuronal activity reversibly with a by long-term dynamics, without neuronal damage. RESULTS A single 10 ng/μl injection of NeoSTX in the dorsal CA1 region abolished for up to 48 h memory capacities and neuronal activity measured by the neuronal marker c-fos. After 72 h of toxin injection, the animals fully recover their memory capacities and hippocampal neuronal activity. The histological inspection of NeoSTX injected brain regions revealed no damage to the tissue or reactive gliosis, similar to vehicle injection. Acute electrophysiological recording in vivo shows, also, minimal spreading of the NeoSTX in the cerebral tissue. COMPARISON WITH EXISTING METHODS Intracerebral NeoSTX injection showed longer effects than other voltage sodium channel blocker, with minimal spreading and no neuronal damage. CONCLUSION NeoSTX is a new useful tool that reversibly inactivates different brains region for a long time, with minimal diffusion and without neuronal damage. Moreover, NeoSTX can be used as a valuable sodium channel blocker for many studies in vivo and with potential therapeutic uses.
Collapse
Affiliation(s)
- J Galindo
- Departamento de Neurociencia, Facultad de Medicina, Universidad de Chile, Chile; Biomedical Neuroscience Institute (BNI), Chile
| | - M Contreras
- Departamento de Neurociencia, Facultad de Medicina, Universidad de Chile, Chile; Biomedical Neuroscience Institute (BNI), Chile
| | - P Maldonado
- Departamento de Neurociencia, Facultad de Medicina, Universidad de Chile, Chile; Biomedical Neuroscience Institute (BNI), Chile
| | - F Torrealba
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Chile
| | - N Lagos
- Membrane Biochemistry Laboratory, Department of Physiology and Biophysics, Faculty of Medicine, University of Chile, Chile
| | - J L Valdés
- Departamento de Neurociencia, Facultad de Medicina, Universidad de Chile, Chile; Biomedical Neuroscience Institute (BNI), Chile.
| |
Collapse
|
60
|
Nakajima K, Takakura H, Shimizu Y, Ogawa M. Changes in plasma membrane damage inducing cell death after treatment with near-infrared photoimmunotherapy. Cancer Sci 2018; 109:2889-2896. [PMID: 29949672 PMCID: PMC6125438 DOI: 10.1111/cas.13713] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 06/25/2018] [Indexed: 12/12/2022] Open
Abstract
Near‐infrared photoimmunotherapy (NIR‐PIT) is a new cancer phototherapy modality using an antibody conjugated to a photosensitizer, IRDye700DX. When the conjugate binds to the plasma membrane and is exposed to NIR light, NIR‐PIT‐treated cells undergo swelling, and target‐selective necrotic/immunogenic cell death is induced. However, the cytotoxic mechanism of NIR‐PIT has not been elucidated. In order to understand the mechanism, it is important to elucidate how the damage to the plasma membrane induced by NIR light irradiation changes over time. Thus, in the present study, we investigated the changes in plasma membrane permeability using ions and molecules of various sizes. Na+ flowed into cells immediately after NIR light irradiation, even when the function of transporters or channels was blocked. Subsequently, fluorescent molecules larger than Na+ entered the cells, but the damage was not large enough for dextran to pass through at early time points. To assess these phenomena quantitatively, membrane permeability was estimated using radiolabeled ions and molecules: 111InCl3, 111In‐DTPA, and 3H‐H2O, and comparable results were obtained. Although minute plasma membrane perforations usually do not induce cell death, our results suggest that the minute damage induced by NIR‐PIT was irreversibly extended with time. In conclusion, minute plasma membrane damage is a trigger for the increase in plasma membrane permeability, cell swelling, and necrotic/immunogenic cell death in NIR‐PIT. Our findings provide new insight into the cytotoxic mechanism of NIR‐PIT.
Collapse
Affiliation(s)
- Kohei Nakajima
- Laboratory of Bioanalysis and Molecular Imaging, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Hideo Takakura
- Laboratory of Bioanalysis and Molecular Imaging, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Yoichi Shimizu
- Laboratory of Bioanalysis and Molecular Imaging, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Mikako Ogawa
- Laboratory of Bioanalysis and Molecular Imaging, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan.,PRESTO, Japan Science and Technology Agency, Kawaguchi, Japan
| |
Collapse
|
61
|
The anti-parkinsonian drug zonisamide reduces neuroinflammation: Role of microglial Na v 1.6. Exp Neurol 2018; 308:111-119. [PMID: 30017881 DOI: 10.1016/j.expneurol.2018.07.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Revised: 06/22/2018] [Accepted: 07/11/2018] [Indexed: 11/23/2022]
Abstract
Parkinson's disease (PD), the second most common age-related progressive neurodegenerative disorder, is characterized by dopamine depletion and the loss of dopaminergic (DA) neurons with accompanying neuroinflammation. Zonisamide is an-anti-convulsant drug that has recently been shown to improve clinical symptoms of PD through its inhibition of monoamine oxidase B (MAO-B). However, zonisamide has additional targets, including voltage-gated sodium channels (Nav), which may contribute to its reported neuroprotective role in preclinical models of PD. Here, we report that Nav1.6 is highly expressed in microglia of post-mortem PD brain and of mice treated with the parkinsonism-inducing neurotoxin MPTP. Administration of zonisamide (20 mg/kg, i.p. every 4 h × 3) following a single injection of MPTP (12.5 mg/kg, s.c.) reduced microglial Nav 1.6 and microglial activation in the striatum, as indicated by Iba-1 staining and mRNA expression of F4/80. MPTP increased the levels of the pro-inflammatory cytokine TNF-α and gp91phox, and this was significantly reduced by zonisamide. Together, these findings suggest that zonisamide may reduce neuroinflammation through the down-regulation of microglial Nav 1.6. Thus, in addition to its effects on parkinsonian symptoms through inhibition of MAO-B, zonisamide may have disease modifying potential through the inhibition of Nav 1.6 and neuroinflammation.
Collapse
|
62
|
Djordjevic N, Jankovic SM, Milovanovic JR. Pharmacokinetics and Pharmacogenetics of Carbamazepine in Children. Eur J Drug Metab Pharmacokinet 2018; 42:729-744. [PMID: 28064419 DOI: 10.1007/s13318-016-0397-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Although carbamazepine is one of the oldest anticonvulsant drugs, it is still heavily utilized for treatment of epilepsy in children. The aim of this article was to review the current knowledge about pharmacokinetics and pharmacogenetics of carbamazepine in children. The literature for this review was systematically searched for in the MEDLINE and SCINDEKS databases. Oral bioavailability of carbamazepine in children is about 75-85%, and it is approximately 75-85% bound to plasma proteins. Apparent volume of distribution is 1.2-1.9 l/kg and total clearance between 0.05 and 0.1 l/h/kg. Pharmacokinetics of carbamazepine in children is age and body weight dependent and highly variable due to influence of dosing regimen and co-medication. The current evidence on the importance of pharmacogenetics for carbamazepine efficacy and safety in children supports the association of PXR*1B, HNF4a rs2071197, CYP1A2*1F, ABCC2 1249G>A, and PRRT2 c.649dupC with either pharmacokinetics or pharmacodynamics of carbamazepine. The importance of human leukocyte antigen (HLA) typing for prediction of adverse drug reactions to carbamazepine in children is also confirmed. Both genetic and environmental factors are responsible for shaping pharmacokinetics and pharmacodynamics of carbamazepine in children. To ensure safe and effective use of carbamazepine in this population, physicians should adjust dosing regimen according to existing pattern of genetic and environmental influences.
Collapse
Affiliation(s)
- Natasa Djordjevic
- Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovica Street, 69, 34000, Kragujevac, Serbia
| | - Slobodan M Jankovic
- Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovica Street, 69, 34000, Kragujevac, Serbia.
| | - Jasmina R Milovanovic
- Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovica Street, 69, 34000, Kragujevac, Serbia
| |
Collapse
|
63
|
Cortical and spinal conditioned media modify the inward ion currents and excitability and promote differentiation of human striatal primordium. J Chem Neuroanat 2018; 90:87-97. [DOI: 10.1016/j.jchemneu.2017.12.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 12/19/2017] [Accepted: 12/19/2017] [Indexed: 11/18/2022]
|
64
|
Ogawara H. Comparison of Strategies to Overcome Drug Resistance: Learning from Various Kingdoms. Molecules 2018; 23:E1476. [PMID: 29912169 PMCID: PMC6100412 DOI: 10.3390/molecules23061476] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 06/13/2018] [Accepted: 06/15/2018] [Indexed: 11/16/2022] Open
Abstract
Drug resistance, especially antibiotic resistance, is a growing threat to human health. To overcome this problem, it is significant to know precisely the mechanisms of drug resistance and/or self-resistance in various kingdoms, from bacteria through plants to animals, once more. This review compares the molecular mechanisms of the resistance against phycotoxins, toxins from marine and terrestrial animals, plants and fungi, and antibiotics. The results reveal that each kingdom possesses the characteristic features. The main mechanisms in each kingdom are transporters/efflux pumps in phycotoxins, mutation and modification of targets and sequestration in marine and terrestrial animal toxins, ABC transporters and sequestration in plant toxins, transporters in fungal toxins, and various or mixed mechanisms in antibiotics. Antibiotic producers in particular make tremendous efforts for avoiding suicide, and are more flexible and adaptable to the changes of environments. With these features in mind, potential alternative strategies to overcome these resistance problems are discussed. This paper will provide clues for solving the issues of drug resistance.
Collapse
Affiliation(s)
- Hiroshi Ogawara
- HO Bio Institute, Yushima-2, Bunkyo-ku, Tokyo 113-0034, Japan.
- Department of Biochemistry, Meiji Pharmaceutical University, Noshio-2, Kiyose, Tokyo 204-8588, Japan.
| |
Collapse
|
65
|
Sanders SJ, Campbell AJ, Cottrell JR, Moller RS, Wagner FF, Auldridge AL, Bernier RA, Catterall WA, Chung WK, Empfield JR, George AL, Hipp JF, Khwaja O, Kiskinis E, Lal D, Malhotra D, Millichap JJ, Otis TS, Petrou S, Pitt G, Schust LF, Taylor CM, Tjernagel J, Spiro JE, Bender KJ. Progress in Understanding and Treating SCN2A-Mediated Disorders. Trends Neurosci 2018; 41:442-456. [PMID: 29691040 DOI: 10.1016/j.tins.2018.03.011] [Citation(s) in RCA: 189] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 03/09/2018] [Accepted: 03/14/2018] [Indexed: 01/20/2023]
Abstract
Advances in gene discovery for neurodevelopmental disorders have identified SCN2A dysfunction as a leading cause of infantile seizures, autism spectrum disorder, and intellectual disability. SCN2A encodes the neuronal sodium channel NaV1.2. Functional assays demonstrate strong correlation between genotype and phenotype. This insight can help guide therapeutic decisions and raises the possibility that ligands that selectively enhance or diminish channel function may improve symptoms. The well-defined function of sodium channels makes SCN2A an important test case for investigating the neurobiology of neurodevelopmental disorders more generally. Here, we discuss the progress made, through the concerted efforts of a diverse group of academic and industry scientists as well as policy advocates, in understanding and treating SCN2A-related disorders.
Collapse
Affiliation(s)
- Stephan J Sanders
- Department of Psychiatry, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA.
| | - Arthur J Campbell
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, 75 Ames Street, Cambridge, MA 02142, USA
| | - Jeffrey R Cottrell
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, 75 Ames Street, Cambridge, MA 02142, USA
| | - Rikke S Moller
- The Danish Epilepsy Centre, Dianalund, Denmark; Institute for Regional Health Services, University of Southern Denmark, Campusvej 55, 5230 Odense, Denmark
| | - Florence F Wagner
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, 75 Ames Street, Cambridge, MA 02142, USA
| | - Angie L Auldridge
- FamilieSCN2a Foundation, P.O. Box 82, East Longmeadow, MA 01028, USA
| | - Raphael A Bernier
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA 98195, USA
| | - William A Catterall
- Department of Pharmacology, University of Washington, Seattle, WA 98195-7280, USA
| | - Wendy K Chung
- Simons Foundation, New York, NY 10010, USA; Department of Pediatrics and Medicine, Columbia University, New York, NY 10032, USA
| | - James R Empfield
- Xenon Pharmaceuticals Inc., 3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | - Alfred L George
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Joerg F Hipp
- Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Omar Khwaja
- Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Evangelos Kiskinis
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Dennis Lal
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, 75 Ames Street, Cambridge, MA 02142, USA
| | - Dheeraj Malhotra
- Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - John J Millichap
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Epilepsy Center and Division of Neurology, Ann & Robert H. Lurie Children's Hospital of Chicago, IL 60611, USA; Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Thomas S Otis
- Sainsbury Wellcome Centre for Neural Circuits and Behaviour, University College London, 25 Howland Street, London W1T 4JG, UK
| | - Steven Petrou
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Geoffrey Pitt
- Cardiovascular Research Institute, Weill Cornell Medical College, Cornell University, New York, NY 10065, USA
| | - Leah F Schust
- FamilieSCN2a Foundation, P.O. Box 82, East Longmeadow, MA 01028, USA
| | - Cora M Taylor
- Geisinger Health System, 100 North Academy Avenue, Danville, PA 17822, USA
| | | | | | - Kevin J Bender
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA.
| |
Collapse
|
66
|
Regulation of voltage-gated sodium channel expression, control of excitability and implications for seizure generation. CURRENT OPINION IN PHYSIOLOGY 2018. [DOI: 10.1016/j.cophys.2017.12.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
67
|
Zhao F, Jin W, Ma L, Zhang JY, Wang JL, Zhang JH, Song YB. Investigation of the selectivity of one type of small-molecule inhibitor for three Na v channel isoforms based on the method of computer simulation. J Biomol Struct Dyn 2018; 37:702-713. [PMID: 29448911 DOI: 10.1080/07391102.2018.1438921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Voltage-gated sodium (Nav) channels play a pivotal role for the changes in membrane potential and belong to large membrane proteins that compose four voltage sensor domains (VSD1-4). In this study, we describe the binding mode and selectivity of one of the aryl sulfonamide sodium channel inhibitors, PF-04856264, for the VSD4s in Nav1.4, Nav1.5 and Nav1.7, respectively, through molecular dynamics simulation and enhanced post-dynamics analyses. Our results show that there are three binding site regions (BSR1-3) in the combination of the ligand and receptors, of which BSR1 and BSR3 contribute to the selectivity and affinity of the ligand to the receptor. What's more, the 39th residue (Y39 in VSD4hNav1.4/ VSD4hNav1.7 and A39 in VSD4hNav1.5) and N42 in BSR1, the 84th residue (L84 in VSD4hNav1.4, T84 in VSD4hNav1.5, and M84 in VSD4hNav1.7) in BSR2 and the conserved positive charged residues in BSR3 have major contributions to the interaction between the ligand and receptor. Further analysis reveals that if the 39th residue has a benzene ring structure, the connection of BSR1 and the ligand would be much stronger through π-stacking interaction. On the other hand, the strength and number of the hydrogen bonds formed by the ligand and the conserved arginines on S4 determine the contribution of BSR3 to the total free binding energy. We anticipate this study pave the way for the design of more effective and safe treatment for pain that selectively target Nav1.7.
Collapse
Affiliation(s)
- Fan Zhao
- a School of Life Science and Bio-pharmaceutics , Shenyang Pharmaceutical University , 103 Wenhua Road, Shenyang 110016 , China
| | - Wei Jin
- a School of Life Science and Bio-pharmaceutics , Shenyang Pharmaceutical University , 103 Wenhua Road, Shenyang 110016 , China
| | - Lin Ma
- a School of Life Science and Bio-pharmaceutics , Shenyang Pharmaceutical University , 103 Wenhua Road, Shenyang 110016 , China
| | - Jian-Ye Zhang
- a School of Life Science and Bio-pharmaceutics , Shenyang Pharmaceutical University , 103 Wenhua Road, Shenyang 110016 , China
| | - Jin-Long Wang
- a School of Life Science and Bio-pharmaceutics , Shenyang Pharmaceutical University , 103 Wenhua Road, Shenyang 110016 , China
| | - Jing-Hai Zhang
- a School of Life Science and Bio-pharmaceutics , Shenyang Pharmaceutical University , 103 Wenhua Road, Shenyang 110016 , China
| | - Yong-Bo Song
- a School of Life Science and Bio-pharmaceutics , Shenyang Pharmaceutical University , 103 Wenhua Road, Shenyang 110016 , China
| |
Collapse
|
68
|
Takahara K, Yamamoto T, Uchida K, Zhu HL, Shibata A, Inai T, Noguchi M, Yotsu-Yamashita M, Teramoto N. Effects of 4,9-anhydrotetrodotoxin on voltage-gated Na + channels of mouse vas deferens myocytes and recombinant Na V1.6 channels. Naunyn Schmiedebergs Arch Pharmacol 2018; 391:489-499. [PMID: 29453527 DOI: 10.1007/s00210-018-1476-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 01/31/2018] [Indexed: 12/19/2022]
Abstract
Molecular investigations were performed in order to determine the major characteristics of voltage-gated Na+ channel β-subunits in mouse vas deferens. The use of real-time quantitative PCR showed that the expression of Scn1b was significantly higher than that of other β-subunit genes (Scn2b - Scn4b). Immunoreactivity of Scn1b proteins was also detected in the inner circular and outer longitudinal smooth muscle of mouse vas deferens. In whole-cell recordings, the actions of 4,9-anhydroTTX on voltage-gated Na+ current peak amplitude in myocytes (i.e., native INa) were compared with its inhibitory potency on recombinant NaV1.6 channels (expressed in HEK293 cells). A depolarizing rectangular voltage-pulse elicited a fast and transient inward native INa and recombinant NaV1.6 expressed in HEK293 cells (i.e., recombinant INa). The current decay of native INa was similar to the recombinant NaV1.6 current co-expressed with β1-subunits. The current-voltage (I-V) relationships of native INa were similar to those of recombinant NaV1.6 currents co-expressed with β1-subunits. Application of 4,9-anhydroTTX inhibited the peak amplitude of native INa (K i = 510 nM), recombinant INa (K i = 112 nM), and recombinant INa co-expressed with β1-subunits (K i = 92 nM). The half-maximal (Vhalf) activation and inactivation of native INa values were similar to those observed in recombinant INa co-expressed with β1-subunits. These results suggest that β1-subunit proteins are likely to be expressed mainly in the smooth muscle layers of murine vas deferens and that 4,9-anhydroTTX inhibited not only native INa but also recombinant INa and recombinant INa co-expressed with β1-subunits in a concentration-dependent manner.
Collapse
Affiliation(s)
- Kohei Takahara
- Department of Pharmacology, Faculty of Medicine, Saga University, Saga, 849-8501, Japan
| | - Tadashi Yamamoto
- Department of Pharmacology, Faculty of Medicine, Saga University, Saga, 849-8501, Japan
| | - Keiichiro Uchida
- Department of Pharmacology, Faculty of Medicine, Saga University, Saga, 849-8501, Japan
| | - Hai-Lei Zhu
- Department of Pharmacology, Faculty of Medicine, Saga University, Saga, 849-8501, Japan
| | - Atsushi Shibata
- Department of Pharmacology, Faculty of Medicine, Saga University, Saga, 849-8501, Japan
| | - Tetsuichiro Inai
- Department of Morphological Biology, Division of Biomedical Sciences, Fukuoka Dental College, Fukuoka, 814-0193, Japan
| | - Mitsuru Noguchi
- Department of Urology, Faculty of Medicine, Saga University, Saga, 849-8501, Japan
| | - Mari Yotsu-Yamashita
- Laboratory of Bioorganic Chemistry of Natural Products, Graduate School of Agricultural Science, Tohoku University, Sendai, 981-8555, Japan
| | - Noriyoshi Teramoto
- Department of Pharmacology, Faculty of Medicine, Saga University, Saga, 849-8501, Japan. .,Laboratory of Biomedical Engineering, Graduate School of Biomedical Engineering, Tohoku University, Sendai, 980-0845, Japan.
| |
Collapse
|
69
|
Bang S, Yoo J, Gong X, Liu D, Han Q, Luo X, Chang W, Chen G, Im ST, Kim YH, Strong JA, Zhang MZ, Zhang JM, Lee SY, Ji RR. Differential Inhibition of Nav1.7 and Neuropathic Pain by Hybridoma-Produced and Recombinant Monoclonal Antibodies that Target Nav1.7 : Differential activities of Nav1.7-targeting monoclonal antibodies. Neurosci Bull 2018; 34:22-41. [PMID: 29333591 PMCID: PMC5799132 DOI: 10.1007/s12264-018-0203-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Accepted: 12/18/2017] [Indexed: 12/21/2022] Open
Abstract
The voltage-gated Na+ channel subtype Nav1.7 is important for pain and itch in rodents and humans. We previously showed that a Nav1.7-targeting monoclonal antibody (SVmab) reduces Na+ currents and pain and itch responses in mice. Here, we investigated whether recombinant SVmab (rSVmab) binds to and blocks Nav1.7 similar to SVmab. ELISA tests revealed that SVmab was capable of binding to Nav1.7-expressing HEK293 cells, mouse DRG neurons, human nerve tissue, and the voltage-sensor domain II of Nav1.7. In contrast, rSVmab showed no or weak binding to Nav1.7 in these tests. Patch-clamp recordings showed that SVmab, but not rSVmab, markedly inhibited Na+ currents in Nav1.7-expressing HEK293 cells. Notably, electrical field stimulation increased the blocking activity of SVmab and rSVmab in Nav1.7-expressing HEK293 cells. SVmab was more effective than rSVmab in inhibiting paclitaxel-induced mechanical allodynia. SVmab also bound to human DRG neurons and inhibited their Na+ currents. Finally, potential reasons for the differential efficacy of SVmab and rSVmab and future directions are discussed.
Collapse
Affiliation(s)
- Sangsu Bang
- Department of Anesthesiology, Duke University Medical Center, 595 LaSalle St, Durham, NC, 27710, USA
| | - Jiho Yoo
- Department of Biochemistry, Duke University Medical Center, 303 Research Drive, Durham, NC, 27710, USA
| | - Xingrui Gong
- Pain Research Center, Department of Anesthesiology, University of Cincinnati, 231 Albert Sabin Way, Cincinnati, OH, 45267-0531, USA
- Department of Anesthesiology, Shanghai Children's Medical Center, Shanghai, 200127, China
| | - Di Liu
- Department of Anesthesiology, Duke University Medical Center, 595 LaSalle St, Durham, NC, 27710, USA
| | - Qingjian Han
- Department of Anesthesiology, Duke University Medical Center, 595 LaSalle St, Durham, NC, 27710, USA
| | - Xin Luo
- Department of Anesthesiology, Duke University Medical Center, 595 LaSalle St, Durham, NC, 27710, USA
| | - Wonseok Chang
- Department of Anesthesiology, Duke University Medical Center, 595 LaSalle St, Durham, NC, 27710, USA
- Department of Physiology and Biophysics, College of Medicine, Eulji University, 143-5 Yongdu-Dong, Jung-Gu, Daejeon, 34824, Korea
| | - Gang Chen
- Department of Anesthesiology, Duke University Medical Center, 595 LaSalle St, Durham, NC, 27710, USA
| | - Sang-Taek Im
- Department of Physiology, College of Medicine, Gachon University, Incheon, 21999, Korea
| | - Yong Ho Kim
- Department of Anesthesiology, Duke University Medical Center, 595 LaSalle St, Durham, NC, 27710, USA
- Department of Physiology, College of Medicine, Gachon University, Incheon, 21999, Korea
| | - Judith A Strong
- Pain Research Center, Department of Anesthesiology, University of Cincinnati, 231 Albert Sabin Way, Cincinnati, OH, 45267-0531, USA
| | - Ma-Zhong Zhang
- Department of Anesthesiology, Shanghai Children's Medical Center, Shanghai, 200127, China
| | - Jun-Ming Zhang
- Pain Research Center, Department of Anesthesiology, University of Cincinnati, 231 Albert Sabin Way, Cincinnati, OH, 45267-0531, USA.
| | - Seok-Yong Lee
- Department of Biochemistry, Duke University Medical Center, 303 Research Drive, Durham, NC, 27710, USA.
| | - Ru-Rong Ji
- Department of Anesthesiology, Duke University Medical Center, 595 LaSalle St, Durham, NC, 27710, USA.
| |
Collapse
|
70
|
Abstract
Every cell within living organisms actively maintains an intracellular Na+ concentration that is 10-12 times lower than the extracellular concentration. The cells then utilize this transmembrane Na+ concentration gradient as a driving force to produce electrical signals, sometimes in the form of action potentials. The protein family comprising voltage-gated sodium channels (NaVs) is essential for such signaling and enables cells to change their status in a regenerative manner and to rapidly communicate with one another. NaVs were first predicted in squid and were later identified through molecular biology in the electric eel. Since then, these proteins have been discovered in organisms ranging from bacteria to humans. Recent research has succeeded in decoding the amino acid sequences of a wide variety of NaV family members, as well as the three-dimensional structures of some. These studies and others have uncovered several of the major steps in the functional and structural transition of NaV proteins that has occurred along the course of the evolutionary history of organisms. Here we present an overview of the molecular evolutionary innovations that established present-day NaV α subunits and discuss their contribution to the evolutionary changes in animal bodies.
Collapse
Affiliation(s)
- Atsuo Nishino
- Department of Biology, Faculty of Agriculture and Life Science, Hirosaki University, Hirosaki, Aomori, Japan.
| | - Yasushi Okamura
- Integrative Physiology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| |
Collapse
|
71
|
Abstract
Voltage-gated sodium (Na+) channels are expressed in virtually all electrically excitable tissues and are essential for muscle contraction and the conduction of impulses within the peripheral and central nervous systems. Genetic disorders that disrupt the function of these channels produce an array of Na+ channelopathies resulting in neuronal impairment, chronic pain, neuromuscular pathologies, and cardiac arrhythmias. Because of their importance to the conduction of electrical signals, Na+ channels are the target of a wide variety of local anesthetic, antiarrhythmic, anticonvulsant, and antidepressant drugs. The voltage-gated family of Na+ channels is composed of α-subunits that encode for the voltage sensor domains and the Na+-selective permeation pore. In vivo, Na+ channel α-subunits are associated with one or more accessory β-subunits (β1-β4) that regulate gating properties, trafficking, and cell-surface expression of the channels. The permeation pore of Na+ channels is divided in two parts: the outer mouth of the pore is the site of the ion selectivity filter, while the inner cytoplasmic pore serves as the channel activation gate. The cytoplasmic lining of the permeation pore is formed by the S6 segments that include highly conserved aromatic amino acids important for drug binding. These residues are believed to undergo voltage-dependent conformational changes that alter drug binding as the channels cycle through the closed, open, and inactivated states. The purpose of this chapter is to broadly review the mechanisms of Na+ channel gating and the models used to describe drug binding and Na+ channel inhibition.
Collapse
Affiliation(s)
- M E O'Leary
- Cooper Medical School of Rowan University, Camden, NJ, USA
| | - M Chahine
- CERVO Brain Research Center, Institut universitaire en santé mentale de Québec, Quebec City, QC, Canada.
- Department of Medicine, Université Laval, Quebec City, QC, Canada.
| |
Collapse
|
72
|
Marakhonov AV, Varenikov GG, Skoblov MY. Sodium Channelopathies: From Molecular Physiology towards Medical Genetics. RUSS J GENET+ 2018. [DOI: 10.1134/s102279541801009x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
73
|
Riquelme G, Sepúlveda JM, Al Ghumgham Z, Del Campo M, Montero C, Lagos N. Neosaxitoxin, a Paralytic Shellfish Poison toxin, effectively manages bucked shins pain, as a local long-acting pain blocker in an equine model. Toxicon 2017; 141:15-17. [PMID: 29146176 DOI: 10.1016/j.toxicon.2017.11.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 11/09/2017] [Accepted: 11/11/2017] [Indexed: 12/19/2022]
Abstract
Local anesthesia is an effective method to control pain. Neosaxitoxin is a phycotoxin whose molecular mechanism includes a reversible inhibition of voltage-gated sodium channels at the axonal level, impeding nerve impulse propagation. The present study was designed to evaluate the clinical efficacy of Neosaxitoxin as a local long-acting pain blocker in horse bucked shins, and it was found to effectively control pain. While Neosaxitoxin and Gonyautoxin, another Paralytic Shellfish Poison (PSP) toxin, have been successfully used in humans as long-lasting pain blockers, this finding marks the first time a PSP has been shown to have an established effect in veterinary medicine.
Collapse
Affiliation(s)
- Gricel Riquelme
- Membrane Biochemistry Laboratory, Department of Physiology and Biophysics, Faculty of Medicine, University of Chile, Independencia 1027, 8380000, Santiago, Chile
| | - Joaquín M Sepúlveda
- Membrane Biochemistry Laboratory, Department of Physiology and Biophysics, Faculty of Medicine, University of Chile, Independencia 1027, 8380000, Santiago, Chile
| | - Zaki Al Ghumgham
- Membrane Biochemistry Laboratory, Department of Physiology and Biophysics, Faculty of Medicine, University of Chile, Independencia 1027, 8380000, Santiago, Chile
| | - Miguel Del Campo
- Membrane Biochemistry Laboratory, Department of Physiology and Biophysics, Faculty of Medicine, University of Chile, Independencia 1027, 8380000, Santiago, Chile
| | - Cecilia Montero
- Membrane Biochemistry Laboratory, Department of Physiology and Biophysics, Faculty of Medicine, University of Chile, Independencia 1027, 8380000, Santiago, Chile
| | - Nestor Lagos
- Membrane Biochemistry Laboratory, Department of Physiology and Biophysics, Faculty of Medicine, University of Chile, Independencia 1027, 8380000, Santiago, Chile.
| |
Collapse
|
74
|
Abstract
Water intake is one of the most basic physiological responses and is essential to sustain life. The perception of thirst has a critical role in controlling body fluid homeostasis and if neglected or dysregulated can lead to life-threatening pathologies. Clear evidence suggests that the perception of thirst occurs in higher-order centres, such as the anterior cingulate cortex (ACC) and insular cortex (IC), which receive information from midline thalamic relay nuclei. Multiple brain regions, notably circumventricular organs such as the organum vasculosum lamina terminalis (OVLT) and subfornical organ (SFO), monitor changes in blood osmolality, solute load and hormone circulation and are thought to orchestrate appropriate responses to maintain extracellular fluid near ideal set points by engaging the medial thalamic-ACC/IC network. Thirst has long been thought of as a negative homeostatic feedback response to increases in blood solute concentration or decreases in blood volume. However, emerging evidence suggests a clear role for thirst as a feedforward adaptive anticipatory response that precedes physiological challenges. These anticipatory responses are promoted by rises in core body temperature, food intake (prandial) and signals from the circadian clock. Feedforward signals are also important mediators of satiety, inhibiting thirst well before the physiological state is restored by fluid ingestion. In this Review, we discuss the importance of thirst for body fluid balance and outline our current understanding of the neural mechanisms that underlie the various types of homeostatic and anticipatory thirst.
Collapse
Affiliation(s)
- Claire Gizowski
- Centre for Research in Neuroscience, Research Institute of the McGill University Health Centre and Montreal General Hospital, 1650 Cedar Avenue, Montreal H3G1A4, Canada
| | - Charles W Bourque
- Centre for Research in Neuroscience, Research Institute of the McGill University Health Centre and Montreal General Hospital, 1650 Cedar Avenue, Montreal H3G1A4, Canada
| |
Collapse
|
75
|
Resch JM, Fenselau H, Madara JC, Wu C, Campbell JN, Lyubetskaya A, Dawes BA, Tsai LT, Li MM, Livneh Y, Ke Q, Kang PM, Fejes-Tóth G, Náray-Fejes-Tóth A, Geerling JC, Lowell BB. Aldosterone-Sensing Neurons in the NTS Exhibit State-Dependent Pacemaker Activity and Drive Sodium Appetite via Synergy with Angiotensin II Signaling. Neuron 2017; 96:190-206.e7. [PMID: 28957668 DOI: 10.1016/j.neuron.2017.09.014] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 08/10/2017] [Accepted: 09/11/2017] [Indexed: 02/06/2023]
Abstract
Sodium deficiency increases angiotensin II (ATII) and aldosterone, which synergistically stimulate sodium retention and consumption. Recently, ATII-responsive neurons in the subfornical organ (SFO) and aldosterone-sensitive neurons in the nucleus of the solitary tract (NTSHSD2 neurons) were shown to drive sodium appetite. Here we investigate the basis for NTSHSD2 neuron activation, identify the circuit by which NTSHSD2 neurons drive appetite, and uncover an interaction between the NTSHSD2 circuit and ATII signaling. NTSHSD2 neurons respond to sodium deficiency with spontaneous pacemaker-like activity-the consequence of "cardiac" HCN and Nav1.5 channels. Remarkably, NTSHSD2 neurons are necessary for sodium appetite, and with concurrent ATII signaling their activity is sufficient to produce rapid consumption. Importantly, NTSHSD2 neurons stimulate appetite via projections to the vlBNST, which is also the effector site for ATII-responsive SFO neurons. The interaction between angiotensin signaling and NTSHSD2 neurons provides a neuronal context for the long-standing "synergy hypothesis" of sodium appetite regulation.
Collapse
Affiliation(s)
- Jon M Resch
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Henning Fenselau
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Joseph C Madara
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Chen Wu
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - John N Campbell
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Anna Lyubetskaya
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Brian A Dawes
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Linus T Tsai
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Monica M Li
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Yoav Livneh
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Qingen Ke
- Cardiovascular Division, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Peter M Kang
- Cardiovascular Division, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Géza Fejes-Tóth
- Department of Physiology and Neurobiology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03755, USA
| | - Anikó Náray-Fejes-Tóth
- Department of Physiology and Neurobiology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03755, USA
| | - Joel C Geerling
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| | - Bradford B Lowell
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; Program in Neuroscience, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
76
|
Blakemore LJ, Trombley PQ. Zinc as a Neuromodulator in the Central Nervous System with a Focus on the Olfactory Bulb. Front Cell Neurosci 2017; 11:297. [PMID: 29033788 PMCID: PMC5627021 DOI: 10.3389/fncel.2017.00297] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 09/06/2017] [Indexed: 12/19/2022] Open
Abstract
The olfactory bulb (OB) is central to the sense of smell, as it is the site of the first synaptic relay involved in the processing of odor information. Odor sensations are first transduced by olfactory sensory neurons (OSNs) before being transmitted, by way of the OB, to higher olfactory centers that mediate olfactory discrimination and perception. Zinc is a common trace element, and it is highly concentrated in the synaptic vesicles of subsets of glutamatergic neurons in some brain regions including the hippocampus and OB. In addition, zinc is contained in the synaptic vesicles of some glycinergic and GABAergic neurons. Thus, zinc released from synaptic vesicles is available to modulate synaptic transmission mediated by excitatory (e.g., N-methyl-D aspartate (NMDA), alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA)) and inhibitory (e.g., gamma-aminobutyric acid (GABA), glycine) amino acid receptors. Furthermore, extracellular zinc can alter the excitability of neurons through effects on a variety of voltage-gated ion channels. Consistent with the notion that zinc acts as a regulator of neuronal activity, we and others have shown zinc modulation (inhibition and/or potentiation) of amino acid receptors and voltage-gated ion channels expressed by OB neurons. This review summarizes the locations and release of vesicular zinc in the central nervous system (CNS), including in the OB. It also summarizes the effects of zinc on various amino acid receptors and ion channels involved in regulating synaptic transmission and neuronal excitability, with a special emphasis on the actions of zinc as a neuromodulator in the OB. An understanding of how neuroactive substances such as zinc modulate receptors and ion channels expressed by OB neurons will increase our understanding of the roles that synaptic circuits in the OB play in odor information processing and transmission.
Collapse
Affiliation(s)
- Laura J Blakemore
- Program in Neuroscience, Florida State UniversityTallahassee, FL, United States.,Department of Biological Science, Florida State UniversityTallahassee, FL, United States
| | - Paul Q Trombley
- Program in Neuroscience, Florida State UniversityTallahassee, FL, United States.,Department of Biological Science, Florida State UniversityTallahassee, FL, United States
| |
Collapse
|
77
|
Berret E, Barron T, Xu J, Debner E, Kim EJ, Kim JH. Oligodendroglial excitability mediated by glutamatergic inputs and Nav1.2 activation. Nat Commun 2017; 8:557. [PMID: 28916793 PMCID: PMC5601459 DOI: 10.1038/s41467-017-00688-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 07/19/2017] [Indexed: 12/19/2022] Open
Abstract
Oligodendrocyte (OL) maturation and axon-glial communication are required for proper myelination in the developing brain. However, physiological properties of OLs remain largely uncharacterized in different brain regions. The roles of oligodendroglial voltage-activated Na+ channels (Nav) and electrical excitability in relation to maturation to the myelinating stage are controversial, although oligodendroglial excitability is potentially important for promoting axon myelination. Here we show spiking properties of OLs and their role in axon-glial communication in the auditory brainstem. A subpopulation of pre-myelinating OLs (pre-OLs) can generate Nav1.2-driven action potentials throughout postnatal development to early adulthood. In addition, excitable pre-OLs receive glutamatergic inputs from neighboring neurons that trigger pre-OL spikes. Knockdown of Nav1.2 channels in pre-OLs alters their morphology, reduces axon-OL interactions and impairs myelination. Our results suggest that Nav1.2-driven spiking of pre-OLs is an integral component of axon-glial communication and is required for the function and maturation of OLs to promote myelination.Axon-glial communication is important for myelination. Here the authors show that during postnatal development in rats, a subpopulation of pre-myelinating oligodendrocytes in the auditory brainstem receive excitatory inputs and can generate Nav 1.2-driven action potentials, and that such process promotes myelination.
Collapse
Affiliation(s)
- Emmanuelle Berret
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center, San Antonio, Texas, 78229, USA
| | - Tara Barron
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center, San Antonio, Texas, 78229, USA
| | - Jie Xu
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center, San Antonio, Texas, 78229, USA
| | - Emily Debner
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center, San Antonio, Texas, 78229, USA
| | - Eun Jung Kim
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center, San Antonio, Texas, 78229, USA
| | - Jun Hee Kim
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center, San Antonio, Texas, 78229, USA.
| |
Collapse
|
78
|
Catterall WA. Forty Years of Sodium Channels: Structure, Function, Pharmacology, and Epilepsy. Neurochem Res 2017; 42:2495-2504. [PMID: 28589518 PMCID: PMC5693772 DOI: 10.1007/s11064-017-2314-9] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 05/19/2017] [Accepted: 05/26/2017] [Indexed: 11/24/2022]
Abstract
Voltage-gated sodium channels initiate action potentials in brain neurons. In the 1970s, much was known about the function of sodium channels from measurements of ionic currents using the voltage clamp method, but there was no information about the sodium channel molecules themselves. As a postdoctoral fellow and staff scientist at the National Institutes of Health, I developed neurotoxins as molecular probes of sodium channels in cultured neuroblastoma cells. During those years, Bruce Ransom and I crossed paths as members of the laboratories of Marshall Nirenberg and Philip Nelson and shared insights about sodium channels in neuroblastoma cells from my work and electrical excitability and synaptic transmission in cultured spinal cord neurons from Bruce's pioneering electrophysiological studies. When I established my laboratory at the University of Washington in 1977, my colleagues and I used those neurotoxins to identify the protein subunits of sodium channels, purify them, and reconstitute their ion conductance activity in pure form. Subsequent studies identified the molecular basis for the main functions of sodium channels-voltage-dependent activation, rapid and selective ion conductance, and fast inactivation. Bruce Ransom and I re-connected in the 1990s, as ski buddies at the Winter Conference on Brain Research and as faculty colleagues at the University of Washington when Bruce became our founding Chair of Neurology and provided visionary leadership of that department. In the past decade my work on sodium channels has evolved into structural biology. Molecular modeling and X-ray crystallographic studies have given new views of sodium channel function at atomic resolution. Sodium channels are also the molecular targets for genetic diseases, including Dravet Syndrome, an intractable pediatric epilepsy disorder with major co-morbidities of cognitive deficit, autistic-like behaviors, and premature death that is caused by loss-of-function mutations in the brain sodium channel NaV1.1. Our work on a mouse genetic model of this disease has shown that its multi-faceted pathophysiology and co-morbidities derive from selective loss of electrical excitability and action potential firing in GABAergic inhibitory neurons, which disinhibits neural circuits throughout the brain and leads directly to the epilepsy, premature death and complex co-morbidities of this disease. It has been rewarding for me to use our developing knowledge of sodium channels to help understand the pathophysiology and to suggest potential therapeutic approaches for this devastating childhood disease.
Collapse
Affiliation(s)
- William A Catterall
- Department of Pharmacology, University of Washington, Seattle, WA, 98195-7280, USA.
| |
Collapse
|
79
|
Swain NA, Batchelor D, Beaudoin S, Bechle BM, Bradley PA, Brown AD, Brown B, Butcher KJ, Butt RP, Chapman ML, Denton S, Ellis D, Galan SRG, Gaulier SM, Greener BS, de Groot MJ, Glossop MS, Gurrell IK, Hannam J, Johnson MS, Lin Z, Markworth CJ, Marron BE, Millan DS, Nakagawa S, Pike A, Printzenhoff D, Rawson DJ, Ransley SJ, Reister SM, Sasaki K, Storer RI, Stupple PA, West CW. Discovery of Clinical Candidate 4-[2-(5-Amino-1H-pyrazol-4-yl)-4-chlorophenoxy]-5-chloro-2-fluoro-N-1,3-thiazol-4-ylbenzenesulfonamide (PF-05089771): Design and Optimization of Diaryl Ether Aryl Sulfonamides as Selective Inhibitors of NaV1.7. J Med Chem 2017; 60:7029-7042. [DOI: 10.1021/acs.jmedchem.7b00598] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
| | | | - Serge Beaudoin
- Icagen Inc., 4222 Emperor Blvd
no. 350, Durham, North Carolina 27703, United States
| | - Bruce M. Bechle
- Worldwide
Medicinal Chemistry, Pfizer Inc., Eastern Point Road, Groton, Connecticut 06340, United States
| | | | | | | | | | | | - Mark L. Chapman
- Icagen Inc., 4222 Emperor Blvd
no. 350, Durham, North Carolina 27703, United States
| | | | | | | | | | | | | | | | | | | | - Matthew S. Johnson
- Icagen Inc., 4222 Emperor Blvd
no. 350, Durham, North Carolina 27703, United States
| | - Zhixin Lin
- Icagen Inc., 4222 Emperor Blvd
no. 350, Durham, North Carolina 27703, United States
| | | | - Brian E. Marron
- Icagen Inc., 4222 Emperor Blvd
no. 350, Durham, North Carolina 27703, United States
| | | | | | | | - David Printzenhoff
- Icagen Inc., 4222 Emperor Blvd
no. 350, Durham, North Carolina 27703, United States
| | | | | | - Steven M. Reister
- Icagen Inc., 4222 Emperor Blvd
no. 350, Durham, North Carolina 27703, United States
| | | | | | | | - Christopher W. West
- Icagen Inc., 4222 Emperor Blvd
no. 350, Durham, North Carolina 27703, United States
| |
Collapse
|
80
|
Sharifi M. Computational approaches to understand the adverse drug effect on potassium, sodium and calcium channels for predicting TdP cardiac arrhythmias. J Mol Graph Model 2017; 76:152-160. [PMID: 28756335 DOI: 10.1016/j.jmgm.2017.06.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2017] [Revised: 06/08/2017] [Accepted: 06/10/2017] [Indexed: 02/08/2023]
Abstract
Ion channels play a crucial role in the cardiovascular system. Our understanding of cardiac ion channel function has improved since their first discoveries. The flow of potassium, sodium and calcium ions across cardiomyocytes is vital for regular cardiac rhythm. Blockage of these channels, delays cardiac repolarization or tend to shorten repolarization and may induce arrhythmia. Detection of drug risk by channel blockade is considered essential for drug regulators. Advanced computational models can be used as an early screen for torsadogenic potential in drug candidates. New drug candidates that are determined to not cause blockage are more likely to pass successfully through preclinical trials and not be withdrawn later from the marketplace by manufacturer. Several different approved drugs, however, can cause a distinctive polymorphic ventricular arrhythmia known as torsade de pointes (TdP), which may lead to sudden death. The objective of the present study is to review the mechanisms and computational models used to assess the risk that a drug may TdP. KEY POINTS There is strong evidence from multiple studies that blockage of the L-type calcium current reduces risk of TdP. Blockage of sodium channels slows cardiac action potential conduction, however, not all sodium channel blocking antiarrhythmic drugs produce a significant effect, while late sodium channel block reduces TdP. Interestingly, there are some drugs that block the hERG potassium channel and therefore cause QT prolongation, but they are not associated with TdP. Recent studies confirmed the necessity of studying multiple distinctionic ion channels which are responsible for cardiac related diseases or TdP, to obtain an improved clinical TdP risk prediction of compound interactions and also for designing drugs.
Collapse
Affiliation(s)
- Mohsen Sharifi
- Division of Systems Biology, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR 72079, USA.
| |
Collapse
|
81
|
Abstract
Headache, especially migraine, has long been associated with epilepsy, based on the common clinical features of these disorders. Both migraine and epilepsy have a genetic predisposition and share common pathophysiological mechanisms including an imbalance between excitatory and inhibitory factors that result in spells of altered brain function and autonomic symptoms. There are well-documented reports on the headache as a sole manifestation of epileptic seizure and headache is commonly associated with as preictal, ictal, and postictal symptoms in epilepsy patients. In addition, migraine and epilepsy are frequently described as highly comorbid conditions and several antiepileptic drugs are used for the patients with migraine as well as epilepsy. In the present review, we briefly discuss the connection between headache and epilepsy in various aspects, including classification, clinical features, epidemiology, genetics, pathophysiology, and treatment.
Collapse
Affiliation(s)
- Dong Wook Kim
- Department of Neurology, Konkuk University School of Medicine, Seoul, Korea
| | - Sang Kun Lee
- Department of Neurology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
82
|
James TF, Nenov MN, Tapia CM, Lecchi M, Koshy S, Green TA, Laezza F. Consequences of acute Na v1.1 exposure to deltamethrin. Neurotoxicology 2017; 60:150-160. [PMID: 28007400 PMCID: PMC5447465 DOI: 10.1016/j.neuro.2016.12.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2015] [Revised: 12/02/2016] [Accepted: 12/14/2016] [Indexed: 12/19/2022]
Abstract
BACKGROUND Pyrethroid insecticides are the most popular class of insecticides in the world, despite their near-ubiquity, their effects of delaying the onset of inactivation of voltage-gated sodium (Nav) channels have not been well-evaluated in all the mammalian Nav isoforms. OBJECTIVE Here we compare the well-studied Nav1.6 isoforms to the less-understood Nav1.1 in their responses to acute deltamethrin exposure. METHODS We used patch-clamp electrophysiology to record sodium currents encoded by either Nav1.1 or Nav1.6 channels stably expressed in HEK293 cells. Protocols evaluating both resting and use-dependent modification were employed. RESULTS We found that exposure of both isoforms to 10μM deltamethrin significantly potentiated persistent and tail current densities without affecting peak transient current densities, and only Nav1.1 maintained these significant effects at 1μM deltamethrin. Window currents increased for both as well, and while only Nav1.6 displayed changes in activation slope and V1/2 of steady-state inactivation for peak currents, V1/2 of persistent current activation was hyperpolarized of ∼10mV by deltamethrin in Nav1.1 cells. Evaluating use-dependence, we found that deltamethrin again potentiated persistent and tail current densities in both isoforms, but only Nav1.6 demonstrated use-dependent enhancement, indicating the primary deltamethrin-induced effects on Nav1.1 channels are not use-dependent. CONCLUSION Collectively, these data provide evidence that Nav1.1 is indeed vulnerable to deltamethrin modification at lower concentrations than Nav1.6, and this effect is primarily mediated during the resting state. GENERAL SIGNIFICANCE These findings identify Nav1.1 as a novel target of pyrethroid exposure, which has major implications for the etiology of neuropsychiatric disorders associated with loss of Nav1.1-expressing inhibitory neurons.
Collapse
Affiliation(s)
- T F James
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, USA; Neuroscience Graduate Program, University of Texas Medical Branch, USA
| | - Miroslav N Nenov
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, USA
| | - Cynthia M Tapia
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, USA
| | - Marzia Lecchi
- Department of Biotechnology and Bioscience, University of Milano-Bicocca, Italy
| | - Shyny Koshy
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, USA; Center for Addiction Research, University of Texas Medical Branch, USA
| | - Thomas A Green
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, USA; Center for Addiction Research, University of Texas Medical Branch, USA
| | - Fernanda Laezza
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, USA; Mitchell Center for Neurodegenerative Diseases, USA; Center for Environmental Toxicology, University of Texas Medical Branch, USA; Center for Addiction Research, University of Texas Medical Branch, USA.
| |
Collapse
|
83
|
Zhorov BS, Dong K. Elucidation of pyrethroid and DDT receptor sites in the voltage-gated sodium channel. Neurotoxicology 2017; 60:171-177. [PMID: 27567732 PMCID: PMC5730328 DOI: 10.1016/j.neuro.2016.08.013] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 08/20/2016] [Accepted: 08/23/2016] [Indexed: 12/19/2022]
Abstract
DDT and pyrethroid insecticides were among the earliest neurotoxins identified to act on voltage-gated sodium channels. In the 1960s, equipped with, at the time, new voltage-clamp techniques, Professor Narahashi and associates provided the initial evidence that DDT and allethrin (the first commercial pyrethroid insecticide) caused prolonged flow of sodium currents in lobster and squid giant axons. Over the next several decades, continued efforts by Prof. Narahashi's group as well as other laboratories led to a comprehensive understanding of the mechanism of action of DDT and pyrethroids on sodium channels. Fast forward to the 1990s, genetic, pharmacological and toxicological data all further confirmed voltage-gated sodium channels as the primary targets of DDT and pyrethroid insecticides. Modifications of the gating kinetics of sodium channels by these insecticides result in repetitive firing and/or membrane depolarization in the nervous system. This mini-review focuses on studies from Prof. Narahashi's pioneer work and more recent mutational and computational modeling analyses which collectively elucidated the elusive pyrethroid receptor sites as well as the molecular basis of differential sensitivities of insect and mammalian sodium channels to pyrethroids.
Collapse
Affiliation(s)
- Boris S Zhorov
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario L8N 3Z5, Canada; Sechenov Institute of Evolutionary Physiology & Biochemistry, Russian Academy of Sciences, St. Petersburg 194223, Russia
| | - Ke Dong
- Department of Entomology, Genetics and Neuroscience Programs, Michigan State University, East Lansing, MI 48824, USA.
| |
Collapse
|
84
|
Soderlund DM, Tan J, He B. Functional reconstitution of rat Na v1.6 sodium channels in vitro for studies of pyrethroid action. Neurotoxicology 2017; 60:142-149. [PMID: 27013268 PMCID: PMC5031521 DOI: 10.1016/j.neuro.2016.03.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 03/17/2016] [Accepted: 03/17/2016] [Indexed: 01/14/2023]
Abstract
The ability to reconstitute sodium channel function and pharmacology in vitro using cloned subunits of known structure has greatly enhanced our understanding of the action of pyrethroid insecticides at this target and the structural determinants of resistance and interspecies selectivity. However, the use of reconstituted channels raises three critical questions: (1) Which subunits and subunit combinations should be used? (2) Which heterologous expression system is preferred? (3) Which combination of subunits and expression system best represents the function of native neuronal channels in the organism of interest? This review considers these questions from the perspective of recent research in this laboratory on the action of pyrethroid insecticides on rat Nav1.6 sodium channels by comparing the effects of heteroligomeric complex composition on channel function and insecticide response when channels are expressed in either Xenopus oocytes or stably-transformed HEK293 cells. These comparisons provide new insight into the influence of cellular context on the functional and pharmacological properties of expressed channels, the modulatory effects of sodium channel auxiliary subunits on the action of pyrethroids, and the relative fidelity of the Xenopus oocyte and HEK293 cell expression systems as model systems for studying of channel function and pyrethroid action.
Collapse
Affiliation(s)
- David M Soderlund
- Department of Entomology, Cornell University, Geneva, NY 14456, USA.
| | | | - Bingjun He
- College of Life Sciences, Nankai University, Tianjin 300071, China
| |
Collapse
|
85
|
Weiss MM, Dineen TA, Marx IE, Altmann S, Boezio A, Bregman H, Chu-Moyer M, DiMauro EF, Feric Bojic E, Foti RS, Gao H, Graceffa R, Gunaydin H, Guzman-Perez A, Huang H, Huang L, Jarosh M, Kornecook T, Kreiman CR, Ligutti J, La DS, Lin MHJ, Liu D, Moyer BD, Nguyen HN, Peterson EA, Rose PE, Taborn K, Youngblood BD, Yu V, Fremeau RT. Sulfonamides as Selective NaV1.7 Inhibitors: Optimizing Potency and Pharmacokinetics While Mitigating Metabolic Liabilities. J Med Chem 2017; 60:5969-5989. [DOI: 10.1021/acs.jmedchem.6b01851] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Thomas Kornecook
- Department
of Neuroscience, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
| | | | - Joseph Ligutti
- Department
of Neuroscience, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
| | | | | | - Dong Liu
- Department
of Neuroscience, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Bryan D. Moyer
- Department
of Neuroscience, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
| | | | | | | | | | | | | | | |
Collapse
|
86
|
Ji X, Saha S, Gao G, Lasek AW, Homanics GE, Guildford M, Tapper AR, Martin GE. The Sodium Channel β4 Auxiliary Subunit Selectively Controls Long-Term Depression in Core Nucleus Accumbens Medium Spiny Neurons. Front Cell Neurosci 2017; 11:17. [PMID: 28243192 PMCID: PMC5303751 DOI: 10.3389/fncel.2017.00017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 01/19/2017] [Indexed: 12/19/2022] Open
Abstract
Voltage-gated sodium channels are essential for generating the initial rapid depolarization of neuronal membrane potential during action potentials (APs) that enable cell-to-cell communication, the propagation of signals throughout the brain, and the induction of synaptic plasticity. Although all brain neurons express one or several variants coding for the core pore-forming sodium channel α subunit, the expression of the β (β1–4) auxiliary subunits varies greatly. Of particular interest is the β4 subunit, encoded by the Scn4b gene, that is highly expressed in dorsal and ventral (i.e., nucleus accumbens – NAc) striata compared to other brain regions, and that endows sodium channels with unique gating properties. However, its role on neuronal activity, synaptic plasticity, and behaviors related to drugs of abuse remains poorly understood. Combining whole-cell patch-clamp recordings with two-photon calcium imaging in Scn4b knockout (KO) and knockdown mice, we found that Scn4b altered the properties of APs in core accumbens medium spiny neurons (MSNs). These alterations are associated with a reduction of the probability of MSNs to evoke spike-timing-dependent long-term depression (tLTD) and a reduced ability of backpropagating APs to evoke dendritic calcium transients. In contrast, long-term potentiation (tLTP) remained unaffected. Interestingly, we also showed that amphetamine-induced locomotor activity was significantly reduced in male Scn4b KO mice compared to wild-type controls. Taken together, these data indicate that the Scn4b subunit selectively controls tLTD by modulating dendritic calcium transients evoked by backpropagating APs.
Collapse
Affiliation(s)
- Xincai Ji
- Department of Psychiatry, The Brudnick Neuropsychiatric Research Institute, University of Massachusetts Medical School, Worcester MA, USA
| | - Sucharita Saha
- Department of Psychiatry, The Brudnick Neuropsychiatric Research Institute, University of Massachusetts Medical School, Worcester MA, USA
| | - Guangping Gao
- Gene Therapy Center, University of Massachusetts Medical School, Worcester MA, USA
| | - Amy W Lasek
- Department of Psychiatry, University of Illinois at Chicago, Chicago IL, USA
| | - Gregg E Homanics
- Department of Anesthesiology, University of Pittsburgh, Pittsburgh PA, USA
| | - Melissa Guildford
- Department of Psychiatry, The Brudnick Neuropsychiatric Research Institute, University of Massachusetts Medical School, Worcester MA, USA
| | - Andrew R Tapper
- Department of Psychiatry, The Brudnick Neuropsychiatric Research Institute, University of Massachusetts Medical School, Worcester MA, USA
| | - Gilles E Martin
- Department of Psychiatry, The Brudnick Neuropsychiatric Research Institute, University of Massachusetts Medical School, Worcester MA, USA
| |
Collapse
|
87
|
Structure-based assessment of disease-related mutations in human voltage-gated sodium channels. Protein Cell 2017; 8:401-438. [PMID: 28150151 PMCID: PMC5445024 DOI: 10.1007/s13238-017-0372-z] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 01/09/2017] [Indexed: 12/15/2022] Open
Abstract
Voltage-gated sodium (Nav) channels are essential for the rapid upstroke of action potentials and the propagation of electrical signals in nerves and muscles. Defects of Nav channels are associated with a variety of channelopathies. More than 1000 disease-related mutations have been identified in Nav channels, with Nav1.1 and Nav1.5 each harboring more than 400 mutations. Nav channels represent major targets for a wide array of neurotoxins and drugs. Atomic structures of Nav channels are required to understand their function and disease mechanisms. The recently determined atomic structure of the rabbit voltage-gated calcium (Cav) channel Cav1.1 provides a template for homology-based structural modeling of the evolutionarily related Nav channels. In this Resource article, we summarized all the reported disease-related mutations in human Nav channels, generated a homologous model of human Nav1.7, and structurally mapped disease-associated mutations. Before the determination of structures of human Nav channels, the analysis presented here serves as the base framework for mechanistic investigation of Nav channelopathies and for potential structure-based drug discovery.
Collapse
|
88
|
Carrasco DI, Vincent JA, Cope TC. Distribution of TTX-sensitive voltage-gated sodium channels in primary sensory endings of mammalian muscle spindles. J Neurophysiol 2017; 117:1690-1701. [PMID: 28123009 DOI: 10.1152/jn.00889.2016] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 01/24/2017] [Accepted: 01/24/2017] [Indexed: 12/19/2022] Open
Abstract
Knowledge of the molecular mechanisms underlying signaling of mechanical stimuli by muscle spindles remains incomplete. In particular, the ionic conductances that sustain tonic firing during static muscle stretch are unknown. We hypothesized that tonic firing by spindle afferents depends on sodium persistent inward current (INaP) and tested for the necessary presence of the appropriate voltage-gated sodium (NaV) channels in primary sensory endings. The NaV1.6 isoform was selected for both its capacity to produce INaP and for its presence in other mechanosensors that fire tonically. The present study shows that NaV1.6 immunoreactivity (IR) is concentrated in heminodes, presumably where tonic firing is generated, and we were surprised to find NaV1.6 IR strongly expressed also in the sensory terminals, where mechanotransduction occurs. This spatial pattern of NaV1.6 IR distribution was consistent for three mammalian species (rat, cat, and mouse), as was tonic firing by primary spindle afferents. These findings meet some of the conditions needed to establish participation of INaP in tonic firing by primary sensory endings. The study was extended to two additional NaV isoforms, selected for their sensitivity to TTX, excluding TTX-resistant NaV channels, which alone are insufficient to support firing by primary spindle endings. Positive immunoreactivity was found for NaV1.1, predominantly in sensory terminals together with NaV1.6 and for NaV1.7, mainly in preterminal axons. Differential distribution in primary sensory endings suggests specialized roles for these three NaV isoforms in the process of mechanosensory signaling by muscle spindles.NEW & NOTEWORTHY The molecular mechanisms underlying mechanosensory signaling responsible for proprioceptive functions are not completely elucidated. This study provides the first evidence that voltage-gated sodium channels (NaVs) are expressed in the spindle primary sensory ending, where NaVs are found at every site involved in transduction or encoding of muscle stretch. We propose that NaVs contribute to multiple steps in sensory signaling by muscle spindles as it does in other types of slowly adapting sensory neurons.
Collapse
Affiliation(s)
- Dario I Carrasco
- School of Biological Science, Georgia Institute of Technology, Atlanta Georgia
| | - Jacob A Vincent
- School of Biological Science, Georgia Institute of Technology, Atlanta Georgia
| | - Timothy C Cope
- School of Biological Science, Georgia Institute of Technology, Atlanta Georgia; .,Biomedical Engineering, Georgia Institute of Technology, Atlanta Georgia; and
| |
Collapse
|
89
|
Silver K, Dong K, Zhorov BS. Molecular Mechanism of Action and Selectivity of Sodium Ch annel Blocker Insecticides. Curr Med Chem 2017; 24:2912-2924. [PMID: 27993108 PMCID: PMC5730267 DOI: 10.2174/0929867323666161216143844] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 11/22/2016] [Accepted: 11/30/2016] [Indexed: 12/19/2022]
Abstract
Sodium channel blocker insecticides (SCBIs) are a relatively new class of insecticides that are represented by two commercially registered compounds, indoxacarb and metaflumizone. SCBIs, like pyrethroids and DDT, target voltage-gated sodium channels (VGSCs) to intoxicate insects. In contrast to pyrethroids, however, SCBIs inhibit VGSCs at a distinct receptor site that overlaps those of therapeutic inhibitors of sodium channels, such as local anesthetics, anticonvulsants and antiarrhythmics. This review will recount the development of the SCBI insecticide class from its roots as chitin synthesis inhibitors, discuss the symptoms of poisoning and evidence supporting inhibition of VGSCs as their mechanism of action, describe the current model for SCBI-induced inhibition of VGSCs, present a model for the receptor for SCBIs on VGSCs, and highlight differences between data collected from mammalian and insect experimental models.
Collapse
Affiliation(s)
- Kristopher Silver
- Department of Entomology, Kansas State University, Manhattan, KS 66506, USA
| | - Ke Dong
- Department of Entomology, Genetics and Neuroscience Programs, Michigan State University, East Lansing, MI 48824, USA
| | - Boris S. Zhorov
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada; Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St. Petersburg, Russia
| |
Collapse
|
90
|
Abstract
Voltage-gated sodium channels (VGSCs) are critical determinants of excitability. The properties of VGSCs are thought to be tightly controlled. However, VGSCs are also subjected to extensive modifications. Multiple posttranslational modifications that covalently modify VGSCs in neurons and muscle have been identified. These include, but are not limited to, phosphorylation, ubiquitination, palmitoylation, nitrosylation, glycosylation, and SUMOylation. Posttranslational modifications of VGSCs can have profound impact on cellular excitability, contributing to normal and abnormal physiology. Despite four decades of research, the complexity of VGSC modulation is still being determined. While some modifications have similar effects on the various VGSC isoforms, others have isoform-specific interactions. In addition, while much has been learned about how individual modifications can impact VGSC function, there is still more to be learned about how different modifications can interact. Here we review what is known about VGSC posttranslational modifications with a focus on the breadth and complexity of the regulatory mechanisms that impact VGSC properties.
Collapse
Affiliation(s)
- Zifan Pei
- Department of Biology, Indiana University - Purdue University Indianapolis, Indianapolis, IN, USA.,Department of Pharmacology and Toxicology, Indiana University - Purdue University Indianapolis, Indianapolis, IN, USA
| | - Yanling Pan
- Medical Neuroscience Graduate Program, Indiana University - Purdue University Indianapolis, Indianapolis, IN, USA
| | - Theodore R Cummins
- Department of Biology, Indiana University - Purdue University Indianapolis, Indianapolis, IN, USA. .,Department of Pharmacology and Toxicology, Indiana University - Purdue University Indianapolis, Indianapolis, IN, USA. .,Medical Neuroscience Graduate Program, Indiana University - Purdue University Indianapolis, Indianapolis, IN, USA.
| |
Collapse
|
91
|
Jiang XZ, Pei YX, Lei W, Wang KY, Shang F, Jiang HB, Wang JJ. Characterization of an insect heterodimeric voltage-gated sodium channel with unique alternative splicing mode. Comp Biochem Physiol B Biochem Mol Biol 2017; 203:149-158. [DOI: 10.1016/j.cbpb.2016.10.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2016] [Revised: 10/20/2016] [Accepted: 10/31/2016] [Indexed: 12/22/2022]
|
92
|
Park SB, Kiernan MC, Vucic S. Axonal Excitability in Amyotrophic Lateral Sclerosis : Axonal Excitability in ALS. Neurotherapeutics 2017; 14:78-90. [PMID: 27878516 PMCID: PMC5233634 DOI: 10.1007/s13311-016-0492-9] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Axonal excitability testing provides in vivo assessment of axonal ion channel function and membrane potential. Excitability techniques have provided insights into the pathophysiological mechanisms underlying the development of neurodegeneration and clinical features of amyotrophic lateral sclerosis (ALS) and related neuromuscular disorders. Specifically, abnormalities of Na+ and K+ conductances contribute to development of membrane hyperexcitability in ALS, thereby leading to symptom generation of muscle cramps and fasciculations, in addition to promoting a neurodegenerative cascade via Ca2+-mediated processes. Modulation of axonal ion channel function in ALS has resulted in significant symptomatic improvement that has been accompanied by stabilization of axonal excitability parameters. Separately, axonal ion channel dysfunction evolves with disease progression and correlates with survival, thereby serving as a potential therapeutic biomarker in ALS. The present review provides an overview of axonal excitability techniques and the physiological mechanisms underlying membrane excitability, with a focus on the role of axonal ion channel dysfunction in motor neuron disease and related neuromuscular diseases.
Collapse
Affiliation(s)
- Susanna B Park
- Brain and Mind Centre, University of Sydney, Sydney, Australia
| | | | - Steve Vucic
- Westmead Clinical School, University of Sydney, Sydney, Australia.
| |
Collapse
|
93
|
Inhibitory effects of hesperetin on Nav1.5 channels stably expressed in HEK 293 cells and on the voltage-gated cardiac sodium current in human atrial myocytes. Acta Pharmacol Sin 2016; 37:1563-1573. [PMID: 27694909 DOI: 10.1038/aps.2016.97] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 07/31/2016] [Indexed: 12/19/2022] Open
Abstract
AIM Voltage-gated sodium channels composed of a pore-forming α subunit and auxiliary β subunits are responsible for the upstroke of the action potential in cardiac myocytes. The pore-forming subunit of the cardiac sodium channel Nav1.5, which is encoded by SCN5A, is the main ion channel that conducts the voltage-gated cardiac sodium current (INa) in cardiac cells. The current study sought to investigate the inhibitory effects of hesperetin on human cardiac Nav1.5 channels stably expressed in human embryonic kidney 293 (HEK 293) cells and on the voltage-gated cardiac sodium current (INa) in human atrial myocytes. METHODS The effects of hesperetin on human cardiac Nav1.5 channels expressed in HEK 293 cells and on cardiac Na+ currents in human atrial myocytes were examined through whole-cell patch-clamp techniques. RESULTS Nav1.5 currents were potently and reversibly suppressed in a concentration- and voltage-dependent manner by hesperetin, which exhibited an IC50 of 62.99 μmol/L. Hesperetin significantly and negatively shifted the voltage-dependent activation and inactivation curves. Hesperetin also markedly decelerated Nav1.5 current inactivation and slowed the recovery from Nav1.5 channel inactivation. The hesperetin-dependent blockage of Nav1.5 currents was frequency-dependent. Hesperetin also potently and reversibly inhibited Na+ current (INa) in human atrial myocytes, consistently with its effects on Nav1.5 currents in HEK 293 cells. CONCLUSION Hesperetin is a potent inhibitor of INa in human atrial myocytes and Nav1.5 channels expressed in human embryonic kidney 293 cells. Hesperetin probably functions by blocking the open state and the inactivated state of these channels.
Collapse
|
94
|
Ching B, Woo JM, Hiong KC, Boo MV, Wong WP, Chew SF, Ip YK. Voltage-Gated Na+ Channel Isoforms and Their mRNA Expression Levels and Protein Abundance in Three Electric Organs and the Skeletal Muscle of the Electric Eel Electrophorus electricus. PLoS One 2016; 11:e0167589. [PMID: 27907137 PMCID: PMC5132174 DOI: 10.1371/journal.pone.0167589] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 11/16/2016] [Indexed: 11/18/2022] Open
Abstract
This study aimed to obtain the coding cDNA sequences of voltage-gated Na+ channel (scn) α-subunit (scna) and β-subunit (scnb) isoforms from, and to quantify their transcript levels in, the main electric organ (EO), Hunter's EO, Sach's EO and the skeletal muscle (SM) of the electric eel, Electrophorus electricus, which can generate both high and low voltage electric organ discharges (EODs). The full coding sequences of two scna (scn4aa and scn4ab) and three scnb (scn1b, scn2b and scn4b) were identified for the first time (except scn4aa) in E. electricus. In adult fish, the scn4aa transcript level was the highest in the main EO and the lowest in the Sach's EO, indicating that it might play an important role in generating high voltage EODs. For scn4ab/Scn4ab, the transcript and protein levels were unexpectedly high in the EOs, with expression levels in the main EO and the Hunter's EO comparable to those of scn4aa. As the key domains affecting the properties of the channel were mostly conserved between Scn4aa and Scn4ab, Scn4ab might play a role in electrogenesis. Concerning scnb, the transcript level of scn4b was much higher than those of scn1b and scn2b in the EOs and the SM. While the transcript level of scn4b was the highest in the main EO, protein abundance of Scn4b was the highest in the SM. Taken together, it is unlikely that Scna could function independently to generate EODs in the EOs as previously suggested. It is probable that different combinations of Scn4aa/Scn4ab and various Scnb isoforms in the three EOs account for the differences in EODs produced in E. electricus. In general, the transcript levels of various scn isoforms in the EOs and the SM were much higher in adult than in juvenile, and the three EOs of the juvenile fish could be functionally indistinct.
Collapse
Affiliation(s)
- Biyun Ching
- Department of Biological Sciences, National University of Singapore, Kent Ridge, Republic of Singapore
| | - Jia M. Woo
- Department of Biological Sciences, National University of Singapore, Kent Ridge, Republic of Singapore
| | - Kum C. Hiong
- Department of Biological Sciences, National University of Singapore, Kent Ridge, Republic of Singapore
| | - Mel V. Boo
- Department of Biological Sciences, National University of Singapore, Kent Ridge, Republic of Singapore
| | - Wai P. Wong
- Department of Biological Sciences, National University of Singapore, Kent Ridge, Republic of Singapore
| | - Shit F. Chew
- Natural Sciences and Science Education, National Institute of Education, Nanyang Technological University, Republic of Singapore
| | - Yuen K. Ip
- Department of Biological Sciences, National University of Singapore, Kent Ridge, Republic of Singapore
- The Tropical Marine Science Institute, National University of Singapore, Kent Ridge, Republic of Singapore
- * E-mail:
| |
Collapse
|
95
|
Smith BJ, Côté PD, Tremblay F. Contribution of Na v1.8 sodium channels to retinal function. Neuroscience 2016; 340:279-290. [PMID: 27984182 DOI: 10.1016/j.neuroscience.2016.10.054] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2016] [Revised: 10/07/2016] [Accepted: 10/21/2016] [Indexed: 12/28/2022]
Abstract
We examined the contribution of the sodium channel isoform Nav1.8 to retinal function using the specific blocker A803467. We found that A803467 has little influence on the electroretinogram (ERG) a- and b-waves, but significantly reduces the oscillatory potentials (OPs) to 40-60% of their original amplitude, with significant changes in implicit time in the rod-driven range. To date, only two cell types were found in mouse to express Nav1.8; the starburst amacrine cells (SBACs), and a subtype of retinal ganglion cells (RGCs). When we recorded light responses from ganglion cells using a multielectrode array we found significant and opposing changes in two physiological groups of RGCs. ON-sustained cells showed significant decreases while transient ON-OFF cells showed significant increases. The effects on ON-OFF transient cells but not ON-sustained cells disappeared in the presence of an inhibitory cocktail. We have previously shown that RGCs have only a minor contribution to the OPs (Smith et al., 2014), therefore suggesting that SBACs might be a significant contributor to this ERG component. Targeting SBACs with the cholinergic neurotoxin ethylcholine mustard aziridinium (AF64A) caused a reduction in the amplitude of the OPs similar to A803467. Our results, both using the ERG and MEA recordings from RGCs, suggest that Nav1.8 plays a role in modulating specific aspects of the retinal physiology and that SBACs are a fundamental cellular contributor to the OPs in mice, a clear demonstration of the dichotomy between ERG b-wave and OPs.
Collapse
Affiliation(s)
- Benjamin J Smith
- Department of Biology, Dalhousie University, 1355 Oxford St., PO Box 15000, Halifax, NS B3H 4R2, Canada.
| | - Patrice D Côté
- Department of Biology, Dalhousie University, 1355 Oxford St., PO Box 15000, Halifax, NS B3H 4R2, Canada; Department of Ophthalmology and Visual Sciences, Dalhousie University, 1276 South Park St., PO Box 15000, Halifax, NS B3H 4R2, Canada.
| | - François Tremblay
- Department of Ophthalmology and Visual Sciences, Dalhousie University, 1276 South Park St., PO Box 15000, Halifax, NS B3H 4R2, Canada; Department of Physiology and Biophysics, Dalhousie University, 5850 College Street, PO Box 15000, Halifax, NS B3H 4R2, Canada.
| |
Collapse
|
96
|
Shao H, Yang Y, Qi AP, Hong P, Zhu GX, Cao XY, Ji WG, Zhu ZR. Gastrodin Reduces the Severity of Status Epilepticus in the Rat Pilocarpine Model of Temporal Lobe Epilepsy by Inhibiting Nav1.6 Sodium Currents. Neurochem Res 2016; 42:360-374. [PMID: 27743286 DOI: 10.1007/s11064-016-2079-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Revised: 09/27/2016] [Accepted: 10/04/2016] [Indexed: 01/06/2023]
Abstract
Temporal lobe epilepsy (TLE) is one of the most refractory types of adult epilepsy, and treatment options remain unsatisfactory. Gastrodin (GAS), a phenolic glucoside used in Chinese herbal medicine and derived from Gastrodia elata Blume, has been shown to have remarkable anticonvulsant effects on various models of epilepsy in vivo. However, the mechanisms of GAS as an anticonvulsant drug remain to be established. By utilizing a combination of behavioral surveys, immunofluorescence and electrophysiological recordings, the present study characterized the anticonvulsant effect of GAS in a pilocarpine-induced status epilepticus (SE) rat model of TLE and explored the underlying cellular mechanisms. We found that GAS pretreatment effectively reduced the severity of SE in the acute phase of TLE. Moreover, GAS protected medial entorhinal cortex (mEC) layer III neurons from neuronal death and terminated the SE-induced bursting discharge of mEC layer II neurons from SE-experienced rats. Furthermore, the current study revealed that GAS prevented the pilocarpine-induced enhancement of Nav1.6 currents (persistent (INaP) and resurgent (INaR) currents), which were reported to play a critical role in the generation of bursting spikes. Consistent with this result, GAS treatment reversed the expression of Nav1.6 protein in SE-experienced EC neurons. These results suggest that the inhibition of Nav1.6 sodium currents may be the underlying mechanism of GAS's anticonvulsant properties.
Collapse
Affiliation(s)
- Hui Shao
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University, Gaotanyan Street 30, Chongqing, 400038, China
- Department of Physiology, Third Military Medical University, Chongqing, China
- The Fifth Camp of Cadet Brigade, Third Military Medical University, Chongqing, China
| | - Yang Yang
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University, Gaotanyan Street 30, Chongqing, 400038, China
| | - Ai-Ping Qi
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University, Gaotanyan Street 30, Chongqing, 400038, China
| | - Pian Hong
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University, Gaotanyan Street 30, Chongqing, 400038, China
| | - Guang-Xi Zhu
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University, Gaotanyan Street 30, Chongqing, 400038, China
| | - Xin-Yu Cao
- The Fifth Camp of Cadet Brigade, Third Military Medical University, Chongqing, China
| | - Wei-Gang Ji
- Department of Chemistry, Faculty of Pharmacy, Third Military Medical University, Chongqing, China
| | - Zhi-Ru Zhu
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University, Gaotanyan Street 30, Chongqing, 400038, China.
- Department of Physiology, Third Military Medical University, Chongqing, China.
| |
Collapse
|
97
|
DiMauro EF, Altmann S, Berry LM, Bregman H, Chakka N, Chu-Moyer M, Bojic EF, Foti RS, Fremeau R, Gao H, Gunaydin H, Guzman-Perez A, Hall BE, Huang H, Jarosh M, Kornecook T, Lee J, Ligutti J, Liu D, Moyer BD, Ortuno D, Rose PE, Schenkel LB, Taborn K, Wang J, Wang Y, Yu V, Weiss MM. Application of a Parallel Synthetic Strategy in the Discovery of Biaryl Acyl Sulfonamides as Efficient and Selective NaV1.7 Inhibitors. J Med Chem 2016; 59:7818-39. [DOI: 10.1021/acs.jmedchem.6b00425] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Affiliation(s)
| | | | | | | | | | | | | | | | - Robert Fremeau
- Department of Neuroscience, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
| | | | | | | | | | | | | | - Thomas Kornecook
- Department of Neuroscience, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
| | | | - Joseph Ligutti
- Department of Neuroscience, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Dong Liu
- Department of Neuroscience, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Bryan D. Moyer
- Department of Neuroscience, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
| | | | | | | | | | | | | | | | | |
Collapse
|
98
|
Winters JJ, Isom LL. Developmental and Regulatory Functions of Na(+) Channel Non-pore-forming β Subunits. CURRENT TOPICS IN MEMBRANES 2016; 78:315-51. [PMID: 27586289 DOI: 10.1016/bs.ctm.2016.07.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Voltage-gated Na(+) channels (VGSCs) isolated from mammalian neurons are heterotrimeric complexes containing one pore-forming α subunit and two non-pore-forming β subunits. In excitable cells, VGSCs are responsible for the initiation of action potentials. VGSC β subunits are type I topology glycoproteins, containing an extracellular amino-terminal immunoglobulin (Ig) domain with homology to many neural cell adhesion molecules (CAMs), a single transmembrane segment, and an intracellular carboxyl-terminal domain. VGSC β subunits are encoded by a gene family that is distinct from the α subunits. While α subunits are expressed in prokaryotes, β subunit orthologs did not arise until after the emergence of vertebrates. β subunits regulate the cell surface expression, subcellular localization, and gating properties of their associated α subunits. In addition, like many other Ig-CAMs, β subunits are involved in cell migration, neurite outgrowth, and axon pathfinding and may function in these roles in the absence of associated α subunits. In sum, these multifunctional proteins are critical for both channel regulation and central nervous system development.
Collapse
Affiliation(s)
- J J Winters
- University of Michigan Neuroscience Program, Ann Arbor, MI, United States
| | - L L Isom
- University of Michigan Neuroscience Program, Ann Arbor, MI, United States; University of Michigan Medical School, Ann Arbor, MI, United States
| |
Collapse
|
99
|
Ghovanloo MR, Aimar K, Ghadiry-Tavi R, Yu A, Ruben PC. Physiology and Pathophysiology of Sodium Channel Inactivation. CURRENT TOPICS IN MEMBRANES 2016; 78:479-509. [PMID: 27586293 DOI: 10.1016/bs.ctm.2016.04.001] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Voltage-gated sodium channels are present in different tissues within the human body, predominantly nerve, muscle, and heart. The sodium channel is composed of four similar domains, each containing six transmembrane segments. Each domain can be functionally organized into a voltage-sensing region and a pore region. The sodium channel may exist in resting, activated, fast inactivated, or slow inactivated states. Upon depolarization, when the channel opens, the fast inactivation gate is in its open state. Within the time frame of milliseconds, this gate closes and blocks the channel pore from conducting any more sodium ions. Repetitive or continuous stimulations of sodium channels result in a rate-dependent decrease of sodium current. This process may continue until the channel fully shuts down. This collapse is known as slow inactivation. This chapter reviews what is known to date regarding, sodium channel inactivation with a focus on various mutations within each NaV subtype and with clinical implications.
Collapse
Affiliation(s)
- M-R Ghovanloo
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
| | - K Aimar
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
| | - R Ghadiry-Tavi
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
| | - A Yu
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
| | - P C Ruben
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
| |
Collapse
|
100
|
Wu X, Huang Y, Kaas Q, Craik DJ. Cyclisation of Disulfide‐Rich Conotoxins in Drug Design Applications. European J Org Chem 2016. [DOI: 10.1002/ejoc.201600402] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Xiaosa Wu
- Institute for Molecular BioscienceThe University of Queensland306 Carmody Road (Building 80)4072BrisbaneAustralia
| | - Yen‐Hua Huang
- Institute for Molecular BioscienceThe University of Queensland306 Carmody Road (Building 80)4072BrisbaneAustralia
| | - Quentin Kaas
- Institute for Molecular BioscienceThe University of Queensland306 Carmody Road (Building 80)4072BrisbaneAustralia
| | - David J. Craik
- Institute for Molecular BioscienceThe University of Queensland306 Carmody Road (Building 80)4072BrisbaneAustralia
| |
Collapse
|