Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
J Ethnopharmacol ; 288: 114998, 2022 Apr 24.
Article in English | MEDLINE | ID: mdl-35063590

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Chronic pain management represents a serious healthcare problem worldwide. The use of opioid analgesics for pain has always been hampered by their side effects; in particular, the addictive liability associated with chronic use. Finding a morphine replacement has been a long-standing goal in the field of analgesia. In traditional Chinese medicine, processed Buthus martensii Karsch (BmK) scorpion has been used as a painkiller to treat chronic inflammatory arthritis and spondylitis, so called "Scorpio-analgesia". However, the molecular basis and the underline mechanism for the Scorpio-analgesia are still unclear. AIM OF THE STUDY: The study aims to investigate the molecular basis of "Scorpio analgesia" and identify novel analgesics from BmK scorpion. MATERIALS AND METHODS: In this study, the analgesic abilities were determined using formalin-, acetic acid- and complete Freund's adjuvant-induced pain models. The effect of BmK venom and processed BmK venom on Nav1.7 were detected by whole-cell voltage-clamp recordings on HEK293-hNav1.7 stable cell line. Action potentials in Dorsal root ganglion (DRG) neurons induced by Makatoxin-3-R58A were recorded in current-clamp mode. The content of Makatoxin-3 was detected using competitive enzyme-linked immunosorbent assay based on the Makatoxin-3 antibody. High performance liquid chromatography, western blot and circular dichroism spectroscopy were used to analysis the stability of Makatoxin-3. RESULTS: Here we demonstrate that Makatoxin-3, an α-like toxin in BmK scorpion venom targeting Nav1.7 is the critical component in Scorpio-analgesia. The analgesic effect of Makatoxin-3 could not be reversed by naloxone and is more potent than Nav1.7-selective inhibitors and non-steroidal anti-inflammatory drugs in inflammatory models. Moreover, a R58A mutant of Makatoxin-3 is capable of eliciting analgesia effect without inducing pain response. CONCLUSIONS: This study advances ion channel biology and proposes Nav1.7 agonists, rather than the presumed Nav1.7-only blockers, for non-narcotic relief of chronic pain.


Subject(s)
Analgesics/pharmacology , Inflammation/drug therapy , Pain/drug therapy , Scorpion Venoms/pharmacology , Action Potentials/drug effects , Analgesics/isolation & purification , Animals , Disease Models, Animal , Freund's Adjuvant , Ganglia, Spinal/drug effects , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , NAV1.7 Voltage-Gated Sodium Channel/drug effects , Neurons/drug effects , Pain/pathology , Voltage-Gated Sodium Channel Agonists/isolation & purification , Voltage-Gated Sodium Channel Agonists/pharmacology
2.
Neurosci Lett ; 753: 135844, 2021 05 14.
Article in English | MEDLINE | ID: mdl-33775738

ABSTRACT

The two voltage gated sodium channels Nav1.7 and Nav1.8 are expressed in the peripheral nervous system and involved in various pain conditions including inflammatory and neuropathic pain. Rodent models bearing deletions or mutations of the corresponding genes, Scn9a and Scn10a, were created in order to understand the role of these channels in the pathophysiological mechanism underlying pain symptoms. This review summarizes the pain behavior profiles reported in Scn9a and Scn10a rodent models. The complete loss-of-function or knockout (KO) of Scn9a or Scn10a and the conditional KO (cKO) of Scn9a in specific cell populations were shown to decrease sensitivity to various pain stimuli. The Possum mutant mice bearing a dominant hypermorphic mutation in Scn10a revealed higher sensitivity to noxious stimuli. Several gain-of-function mutations were identified in patients with painful small fiber neuropathy. Future knowledge obtained from preclinical models bearing these mutations will allow understanding how these mutations affect pain. In addition, the review gives perspectives for creating models that better mimic patients' pain symptoms in view to developing novel analgesic strategies.


Subject(s)
Analgesics/pharmacology , NAV1.7 Voltage-Gated Sodium Channel/genetics , NAV1.8 Voltage-Gated Sodium Channel/genetics , Pain/genetics , Small Fiber Neuropathy/complications , Analgesics/therapeutic use , Animals , Disease Models, Animal , Gain of Function Mutation , Humans , Loss of Function Mutation , Mice , Mice, Knockout , NAV1.7 Voltage-Gated Sodium Channel/metabolism , NAV1.8 Voltage-Gated Sodium Channel/metabolism , Nociception/drug effects , Pain/drug therapy , Rats , Rats, Transgenic , Small Fiber Neuropathy/drug therapy , Small Fiber Neuropathy/genetics , Voltage-Gated Sodium Channel Agonists/pharmacology , Voltage-Gated Sodium Channel Agonists/therapeutic use , Voltage-Gated Sodium Channel Blockers/pharmacology , Voltage-Gated Sodium Channel Blockers/therapeutic use
3.
Sci Rep ; 10(1): 10730, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32612253

ABSTRACT

Voltage-gated Na+ (NaV) channels regulate homeostasis in bacteria and control membrane electrical excitability in mammals. Compared to their mammalian counterparts, bacterial NaV channels possess a simpler, fourfold symmetric structure and have facilitated studies of the structural basis of channel gating. However, the pharmacology of bacterial NaV remains largely unexplored. Here we systematically screened 39 NaV modulators on a bacterial channel (NaChBac) and characterized a selection of compounds on NaChBac and a mammalian channel (human NaV1.7). We found that while many compounds interact with both channels, they exhibit distinct functional effects. For example, the local anesthetics ambroxol and lidocaine block both NaV1.7 and NaChBac but affect activation and inactivation of the two channels to different extents. The voltage-sensing domain targeting toxin BDS-I increases NaV1.7 but decreases NaChBac peak currents. The pore binding toxins aconitine and veratridine block peak currents of NaV1.7 and shift activation (aconitine) and inactivation (veratridine) respectively. In NaChBac, they block the peak current by binding to the pore residue F224. Nonetheless, aconitine has no effect on activation or inactivation, while veratridine only modulates activation of NaChBac. The conservation and divergence in the pharmacology of bacterial and mammalian NaV channels provide insights into the molecular basis of channel gating and will facilitate organism-specific drug discovery.


Subject(s)
Anesthetics, Local/pharmacology , Bacterial Proteins/metabolism , Drug Interactions , NAV1.7 Voltage-Gated Sodium Channel/metabolism , Small Molecule Libraries/metabolism , Sodium Channels/metabolism , Toxins, Biological/pharmacology , Aconitine/pharmacology , Bacterial Proteins/chemistry , Electrophysiological Phenomena , HEK293 Cells , Humans , Ion Channel Gating , NAV1.7 Voltage-Gated Sodium Channel/chemistry , Sodium Channels/chemistry , Veratridine/pharmacology , Voltage-Gated Sodium Channel Agonists/pharmacology
4.
Toxins (Basel) ; 12(5)2020 05 09.
Article in English | MEDLINE | ID: mdl-32397386

ABSTRACT

Ciguatoxins (CTXs) are a group of neurotoxins responsible for the syndrome ciguatera fish poisoning (CFP) as a result of the consumption of contaminated fish. The presence of these toxins has been detected around the Pacific, Caribbean and Indian coasts. Recent reports indicate the emergence of CFP in other geographic areas, in particular in European coasts, of the Canary Islands (Spain) and Madeira (Portugal). A neuroblastoma cell line of murine origin (N2a) has been applied to assay different groups of neurotoxins, acting on voltage-gated sodium channel (VGSC) of excitable cells, N2a-MTT. The great potential of N2a-MTT as a sensitive tool for the CTXs screening is clearly recognized, notably because it allows the detection of these toxins at levels below recommended as security levels. However, the complexity of the matrix is a critical point on the application of N2a-MTT, which needs to be evaluated. The aim of this work is to provide recommendations for an implemented N2a-MTT method for CTXs determination in fish that avoids matrix effects, particularly those related to high lipid content.


Subject(s)
Biological Assay , Ciguatoxins/analysis , Ciguatoxins/pharmacology , Fishes/metabolism , Neurons/drug effects , Voltage-Gated Sodium Channel Agonists/analysis , Voltage-Gated Sodium Channel Agonists/pharmacology , Voltage-Gated Sodium Channels/drug effects , Animals , Cell Line, Tumor , Cell Survival/drug effects , Membrane Potentials , Mice , Neurons/metabolism , Neurons/pathology , Ouabain/pharmacology , Veratridine/pharmacology , Voltage-Gated Sodium Channels/metabolism
5.
Toxins (Basel) ; 12(5)2020 04 27.
Article in English | MEDLINE | ID: mdl-32349302

ABSTRACT

The neuroblastoma cell-based assay (CBA-N2a) is widely used for the detection of marine biotoxins in seafood products, yet a consensus protocol is still lacking. In this study, six key parameters of CBA-N2a were revisited: cell seeding densities, cell layer viability after 26 h growth, MTT incubation time, Ouabain and Veratridine treatment and solvent and matrix effects. A step-by-step protocol was defined identifying five viability controls for the validation of CBA-N2a results. Specific detection of two voltage gated sodium channel activators, pacific ciguatoxin (P-CTX3C) and brevetoxin (PbTx3) and two inhibitors, saxitoxin (STX) and decarbamoylsaxitoxin (dc-STX) was achieved, with EC50 values of 1.7 ± 0.35 pg/mL, 5.8 ± 0.9 ng/mL, 3 ± 0.5 ng/mL and 15.8 ± 3 ng/mL, respectively. When applied to the detection of ciguatoxin (CTX)-like toxicity in fish samples, limit of detection (LOD) and limit of quantification (LOQ) values were 0.031 ± 0.008 and 0.064 ± 0.016 ng P-CTX3C eq/g of flesh, respectively. Intra and inter-assays comparisons of viability controls, LOD, LOQ and toxicity in fish samples gave coefficients of variation (CVs) ranging from 3% to 29%. This improved test adaptable to either high throughput screening or composite toxicity estimation is a useful starting point for a standardization of the CBA-N2a in the field of marine toxin detection.


Subject(s)
Biological Assay , Fishes/metabolism , Marine Toxins/analysis , Neurons/drug effects , Voltage-Gated Sodium Channel Agonists/analysis , Voltage-Gated Sodium Channels/drug effects , Animals , Cell Line, Tumor , Cell Proliferation , Cell Survival/drug effects , Ciguatoxins/analysis , Ciguatoxins/toxicity , Dose-Response Relationship, Drug , Limit of Detection , Marine Toxins/toxicity , Mice , Neuroblastoma , Neurons/metabolism , Neurons/pathology , Ouabain/pharmacology , Oxocins/analysis , Oxocins/toxicity , Reproducibility of Results , Saxitoxin/analysis , Saxitoxin/toxicity , Time Factors , Veratridine/pharmacology , Voltage-Gated Sodium Channel Agonists/toxicity , Voltage-Gated Sodium Channels/metabolism
6.
PLoS One ; 15(3): e0219106, 2020.
Article in English | MEDLINE | ID: mdl-32134913

ABSTRACT

Dravet syndrome is caused by dominant loss-of-function mutations in SCN1A which cause reduced activity of Nav1.1 leading to lack of neuronal inhibition. On the other hand, gain-of-function mutations in SCN8A can lead to a severe epileptic encephalopathy subtype by over activating NaV1.6 channels. These observations suggest that Nav1.1 and Nav1.6 represent two opposing sides of the neuronal balance between inhibition and activation. Here, we hypothesize that Dravet syndrome may be treated by either enhancing Nav1.1 or reducing Nav1.6 activity. To test this hypothesis we generated and characterized a novel DS zebrafish model and tested new compounds that selectively activate or inhibit the human NaV1.1 or NaV1.6 channel respectively. We used CRISPR/Cas9 to generate two separate Scn1Lab knockout lines as an alternative to previous zebrafish models generated by random mutagenesis or morpholino oligomers. Using an optimized locomotor assay, spontaneous burst movements were detected that were unique to Scn1Lab knockouts and disappear when introducing human SCN1A mRNA. Besides the behavioral phenotype, Scn1Lab knockouts show sudden, electrical discharges in the brain that indicate epileptic seizures in zebrafish. Scn1Lab knockouts showed increased sensitivity to the GABA antagonist pentylenetetrazole and a reduction in whole organism GABA levels. Drug screenings further validated a Dravet syndrome phenotype. We tested the NaV1.1 activator AA43279 and two novel NaV1.6 inhibitors MV1369 and MV1312 in the Scn1Lab knockouts. Both type of compounds significantly reduced the number of spontaneous burst movements and seizure activity. Our results show that selective inhibition of NaV1.6 could be just as efficient as selective activation of NaV1.1 and these approaches could prove to be novel potential treatment strategies for Dravet syndrome and other (genetic) epilepsies. Compounds tested in zebrafish however, should always be further validated in other model systems for efficacy in mammals and to screen for potential side effects.


Subject(s)
Epilepsies, Myoclonic/pathology , NAV1.1 Voltage-Gated Sodium Channel/metabolism , NAV1.6 Voltage-Gated Sodium Channel/metabolism , Zebrafish Proteins/metabolism , Animals , Anticonvulsants/pharmacology , Disease Models, Animal , Epilepsies, Myoclonic/metabolism , Humans , Locomotion/drug effects , Morpholinos/metabolism , Mutagenesis , NAV1.1 Voltage-Gated Sodium Channel/chemistry , NAV1.1 Voltage-Gated Sodium Channel/genetics , NAV1.6 Voltage-Gated Sodium Channel/chemistry , NAV1.6 Voltage-Gated Sodium Channel/genetics , Neurons/drug effects , Neurons/metabolism , Pentylenetetrazole/pharmacology , Phenotype , RNA, Guide, Kinetoplastida/metabolism , RNA, Messenger/metabolism , Voltage-Gated Sodium Channel Agonists/pharmacology , Voltage-Gated Sodium Channel Blockers/pharmacology , Zebrafish , Zebrafish Proteins/chemistry , Zebrafish Proteins/genetics , gamma-Aminobutyric Acid/metabolism
7.
Toxins (Basel) ; 12(4)2020 03 25.
Article in English | MEDLINE | ID: mdl-32218140

ABSTRACT

Research in toxinology has created a pharmacological paradox. With an estimated 220,000 venomous animals worldwide, the study of peptidyl toxins provides a vast number of effector molecules. However, due to the complexity of the protein-protein interactions, there are fewer than ten venom-derived molecules on the market. Structural characterization and identification of post-translational modifications are essential to develop biological lead structures into pharmaceuticals. Utilizing advancements in mass spectrometry, we have created a high definition approach that fuses conventional high-resolution MS-MS with ion mobility spectrometry (HDMSE) to elucidate these primary structure characteristics. We investigated venom from ten species of "tiger" spider (Genus: Poecilotheria) and discovered they contain isobaric conformers originating from non-enzymatic Asp isomerization. One conformer pair conserved in five of ten species examined, denominated PcaTX-1a and PcaTX-1b, was found to be a 36-residue peptide with a cysteine knot, an amidated C-terminus, and isoAsp33Asp substitution. Although the isomerization of Asp has been implicated in many pathologies, this is the first characterization of Asp isomerization in a toxin and demonstrates the isomerized product's diminished physiological effects. This study establishes the value of a HDMSE approach to toxin screening and characterization.


Subject(s)
Aspartic Acid/chemistry , Ion Mobility Spectrometry , Mass Spectrometry , NAV1.7 Voltage-Gated Sodium Channel/drug effects , Neurotoxins/pharmacology , Spider Venoms/pharmacology , Voltage-Gated Sodium Channel Agonists/pharmacology , Amino Acid Sequence , Animals , Binding Sites , Cell Line , Drug Discovery , Humans , Isomerism , Membrane Potentials , NAV1.7 Voltage-Gated Sodium Channel/metabolism , Neurotoxins/chemistry , Protein Binding , Protein Conformation , Protein Processing, Post-Translational , Spider Venoms/chemistry , Structure-Activity Relationship , Voltage-Gated Sodium Channel Agonists/chemistry
8.
Mol Pharmacol ; 97(6): 377-383, 2020 06.
Article in English | MEDLINE | ID: mdl-32193331

ABSTRACT

PF-05089771 is an aryl sulfonamide Nav1.7 channel blocker that binds to the inactivated state of Nav1.7 channels with high affinity but binds only weakly to channels in the resting state. Such aryl sulfonamide Nav1.7 channel blockers bind to the extracellular surface of the S1-S4 voltage-sensor segment of homologous Domain 4, whose movement is associated with inactivation. This binding site is different from that of classic sodium channel inhibitors like lidocaine, which also bind with higher affinity to the inactivated state than the resting state but bind at a site within the pore of the channel. The common dependence on gating state with distinct binding sites raises the possibility that inhibition by aryl sulfonamides and by classic local anesthetics might show an interaction mediated by their mutual state dependence. We tested this possibility by examining the state-dependent inhibition by PF-05089771 and lidocaine of human Nav1.7 channels expressed in human embryonic kidney 293 cells. At -80 mV, where a small fraction of channels are in an inactivated state under drug-free conditions, inhibition by PF-05089771 was both enhanced and speeded in the presence of lidocaine. The results suggest that lidocaine binding to the channel enhances PF-05089771 inhibition by altering the equilibrium between resting states (with D4S4 in the inner position) and inactivated states (with D4S4 in the outer position). The gating state-mediated interaction between the compounds illustrates a principle applicable to many state-dependent agents. SIGNIFICANCE STATEMENT: The results show that lidocaine enhances the degree and rate of inhibition of Nav1.7 channels by the aryl sulfonamide compound PF-05089771, consistent with state-dependent binding by lidocaine increasing the fraction of channels presenting a high-affinity binding site for PF-05089771 and suggesting that combinations of agents targeted to the pore-region binding site of lidocaine and the external binding site of aryl sulfonamides may have synergistic actions.


Subject(s)
Lidocaine/pharmacology , NAV1.7 Voltage-Gated Sodium Channel/metabolism , Phenyl Ethers/pharmacology , Sulfonamides/pharmacology , Voltage-Gated Sodium Channel Agonists/pharmacology , Drug Synergism , HEK293 Cells , Humans
9.
Sci Rep ; 9(1): 16890, 2019 11 15.
Article in English | MEDLINE | ID: mdl-31729429

ABSTRACT

Multiple voltage-gated Na+ (Nav) channelopathies can be ascribed to subtle changes in the Nav macromolecular complex. Fibroblast growth factor 14 (FGF14) is a functionally relevant component of the Nav1.6 channel complex, a causative link to spinocerebellar ataxia 27 (SCA27) and an emerging risk factor for neuropsychiatric disorders. Yet, how this protein:channel complex is regulated in the cell is still poorly understood. To search for key cellular pathways upstream of the FGF14:Nav1.6 complex, we have developed, miniaturized and optimized an in-cell assay in 384-well plates by stably reconstituting the FGF14:Nav1.6 complex using the split-luciferase complementation assay. We then conducted a high-throughput screening (HTS) of 267 FDA-approved compounds targeting known mediators of cellular signaling. Of the 65 hits initially detected, 24 were excluded based on counter-screening and cellular toxicity. Based on target analysis, potency and dose-response relationships, 5 compounds were subsequently repurchased for validation and confirmed as hits. Among those, the tyrosine kinase inhibitor lestaurtinib was highest ranked, exhibiting submicromolar inhibition of FGF14:Nav1.6 assembly. While providing evidence for a robust in-cell HTS platform that can be adapted to search for any channelopathy-associated regulatory proteins, these results lay the potential groundwork for repurposing cancer drugs for neuropsychopharmacology.


Subject(s)
Antineoplastic Agents , High-Throughput Screening Assays/methods , Protein Interaction Maps/physiology , Voltage-Gated Sodium Channel Agonists/isolation & purification , Voltage-Gated Sodium Channel Blockers/isolation & purification , Voltage-Gated Sodium Channels/drug effects , Antineoplastic Agents/isolation & purification , Antineoplastic Agents/pharmacology , Drug Evaluation, Preclinical/methods , Fibroblast Growth Factors/agonists , Fibroblast Growth Factors/antagonists & inhibitors , Fibroblast Growth Factors/chemistry , HEK293 Cells , Humans , Multiprotein Complexes/agonists , Multiprotein Complexes/antagonists & inhibitors , Multiprotein Complexes/chemistry , NAV1.6 Voltage-Gated Sodium Channel/drug effects , NAV1.6 Voltage-Gated Sodium Channel/metabolism , Protein Binding , Voltage-Gated Sodium Channel Agonists/pharmacology , Voltage-Gated Sodium Channel Blockers/pharmacology , Voltage-Gated Sodium Channels/metabolism
10.
Neuron ; 104(2): 370-384.e5, 2019 10 23.
Article in English | MEDLINE | ID: mdl-31537465

ABSTRACT

Sleep pressure increases during wake and dissipates during sleep, but the molecules and neurons that measure homeostatic sleep pressure remain poorly understood. We present a pharmacological assay in larval zebrafish that generates short-term increases in wakefulness followed by sustained rebound sleep after washout. The intensity of global neuronal activity during drug-induced wakefulness predicted the amount of subsequent rebound sleep. Whole-brain mapping with the neuronal activity marker phosphorylated extracellular signal-regulated kinase (pERK) identified preoptic Galanin (Galn)-expressing neurons as selectively active during rebound sleep, and the relative induction of galn transcripts was predictive of total rebound sleep time. Galn is required for sleep homeostasis, as galn mutants almost completely lacked rebound sleep following both pharmacologically induced neuronal activity and physical sleep deprivation. These results suggest that Galn plays a key role in responding to sleep pressure signals derived from neuronal activity and functions as an output arm of the vertebrate sleep homeostat.


Subject(s)
GABA Antagonists/pharmacology , Galanin/drug effects , Neurons/drug effects , Pentylenetetrazole/pharmacology , Sleep Deprivation/metabolism , Sleep/drug effects , Wakefulness/drug effects , 4-Aminopyridine/pharmacology , Aconitine/pharmacology , Animals , Caffeine/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Galanin/genetics , Galanin/metabolism , Homeostasis , Mutation , Neurons/metabolism , Phosphorylation , Potassium Channel Blockers/pharmacology , Preoptic Area , Purinergic P1 Receptor Antagonists/pharmacology , Sleep/genetics , Voltage-Gated Sodium Channel Agonists/pharmacology , Wakefulness/genetics , Zebrafish
11.
Mol Pharm ; 16(9): 3948-3956, 2019 09 03.
Article in English | MEDLINE | ID: mdl-31361498

ABSTRACT

Many oral mucosal conditions cause considerable and prolonged pain that to date has been difficult to alleviate via topical delivery, and the use of injection causes many patients dental anxiety and needle-prick pain. Therefore, developing a noninjectable drug delivery system as an alternative administration procedure may vastly improve the health and wellbeing of these patients. Recent advances in the development of mucoadhesive electrospun patches for the direct delivery of therapeutics to the oral mucosa offer a potential solution, but as yet, the release of local anesthetics from this system and their uptake by oral tissue have not been demonstrated. Here, we demonstrate the fabrication of lidocaine-loaded electrospun fiber patches, drug release, and subsequent uptake and permeation through the porcine buccal mucosa. Lidocaine HCl and lidocaine base were incorporated into the electrospun patches to evaluate the difference in drug permeation for the two drug compositions. Lidocaine released from the lidocaine HCl-containing electrospun patches was significantly quicker than from the lidocaine base patches, with double the amount of drug released from the lidocaine HCl patches in the first 15 min (0.16 ± 0.04 mg) compared to that from the lidocaine base patches (0.07 ± 0.01 mg). The permeation of lidocaine from the lidocaine HCl electrospun patches through ex vivo porcine buccal mucosa was also detected in 15 min, whereas permeation of lidocaine from the lidocaine base patch was not detected. Matrix-assisted laser desorption ionization-mass spectrometry imaging was used to investigate localization of lidocaine within the oral tissue. Lidocaine in the solution as well as from the mucoadhesive patch penetrated into the buccal mucosal tissue in a time-dependent manner and was detectable in the lamina propria after only 15 min. Moreover, the lidocaine released from lidocaine HCl electrospun patches retained biological activity, inhibiting veratridine-mediated opening of voltage-gated sodium channels in SH-SY5Y neuroblastoma cells. These data suggest that a mucoadhesive electrospun patch may be used as a vehicle for rapid uptake and sustained anesthetic drug delivery to treat or prevent oral pain.


Subject(s)
Anesthetics/pharmacokinetics , Drug Delivery Systems/methods , Lidocaine/pharmacokinetics , Mouth Mucosa/drug effects , Oral Mucosal Absorption/physiology , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Voltage-Gated Sodium Channel Blockers/pharmacokinetics , Administration, Buccal , Anesthetics/administration & dosage , Animals , Cell Line, Tumor , Drug Liberation , Facial Pain/drug therapy , Humans , Lidocaine/administration & dosage , Mouth Mucosa/metabolism , Neuroblastoma/metabolism , Neuroblastoma/pathology , Swine , Tissue Distribution , Veratridine/pharmacology , Voltage-Gated Sodium Channel Agonists/pharmacology , Voltage-Gated Sodium Channel Blockers/administration & dosage
12.
Res Vet Sci ; 124: 317-320, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31030119

ABSTRACT

Aconitine, a major aconitum alkaloid, is well known for its high toxicity that induces severe arrhythmias and neurological symptoms. One mechanism of aconitine-induced toxic responses is the induction of apoptosis. Apoptosis and autophagy are interconnected processes and the two pathways share critical components. In this study, we investigated the role of autophagy in aconitine-induced toxicity using mouse model. 120 mice were randomly divided into 4 experimental groups (normal saline), low dose group (0.14 µmol/L), medium dose group (0.28 µmol/L) and high dose group (0.56 µmol/ L). 30 mice in each group were administered with aconitine (lavage) for 30 days. The livers were collected for analysis of autophagy-related proteins by Western blotting. The expression of LC3II/LC3I ratio and Beclin 1 were found to increase and then decrease with the highest expression at 10 days and the p62 showed a time-dependent decreases. Autophagy is regulated by the mTOR pathway, we further analyzed the effects of aconitine on this pathway and found aconitine inhibited, phosphorylation of p-PI3K, p-Akt and p-mTOR. The p-p70s6k and p-4EBP1 which are downstream of mTOR were concomitantly decreased. These results suggest that aconitine induce autophagy in mouse liver. The PI3K/Akt/mTOR signaling pathway is involved in the regulation of aconitine-induced autophagy in the liver of mice.


Subject(s)
Aconitine/toxicity , Autophagy/genetics , Liver/drug effects , Signal Transduction/genetics , Animals , Dose-Response Relationship, Drug , Female , Liver/physiology , Male , Mice , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Voltage-Gated Sodium Channel Agonists/toxicity
13.
PLoS One ; 14(3): e0213751, 2019.
Article in English | MEDLINE | ID: mdl-30856233

ABSTRACT

Voltage-gated sodium channels (NaVs) are key therapeutic targets for pain, epilepsy and cardiac arrhythmias. Here we describe the development of a no-wash fluorescent sodium influx assay suitable for high-throughput screening and characterization of novel drug leads. Addition of red-violet food dyes (peak absorbance range 495-575 nm) to assays in HEK293 cells heterologously expressing hNaV1.1-1.8 effectively quenched background fluorescence of the sodium indicator dye Asante NaTRIUM Green-2 (ANG-2; peak emission 540 nm), negating the need for a wash step. Ponceau 4R (1 mM) was identified as a suitable quencher, which had no direct effect on NaV channels as assessed by patch-clamp experiments, and did not alter the pharmacology of the NaV1.1-1.7 activator veratridine (EC50 10-29 µM) or the NaV1.1-1.8 inhibitor tetracaine (IC50's 6-66 µM). In addition, we also identified that the food dyes Ponceau 4R, Brilliant Black BN, Allura Red and Amaranth are effective at quenching the background fluorescence of the calcium indicator dyes fluo-4, fura-2 and fura-5F, identifying them as potential inexpensive alternatives to no-wash calcium ion indicator kits. In summary, we have developed a no-wash fluorescent sodium influx assay suitable for high-throughput screening based on the sodium indicator dye ANG-2 and the quencher Ponceau 4R.


Subject(s)
High-Throughput Screening Assays/methods , Sodium/metabolism , Fluorescent Dyes/chemistry , Fluorescent Dyes/metabolism , HEK293 Cells , Humans , Patch-Clamp Techniques , Sodium/analysis , Spectrometry, Fluorescence , Tetracaine/chemistry , Tetracaine/metabolism , Veratridine/chemistry , Veratridine/metabolism , Voltage-Gated Sodium Channel Agonists/chemistry , Voltage-Gated Sodium Channel Agonists/metabolism , Voltage-Gated Sodium Channel Blockers/chemistry , Voltage-Gated Sodium Channel Blockers/metabolism , Voltage-Gated Sodium Channels/chemistry , Voltage-Gated Sodium Channels/metabolism
14.
Bioorg Med Chem Lett ; 29(6): 815-820, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30704812

ABSTRACT

The voltage-gated sodium channel, Nav1.1, is predominantly expressed in parvalbumin-positive fast spiking interneurons and has been genetically linked to Dravet syndrome. Starting from a high throughput screening hit isoxazole derivative 5, modifications of 5 via combinations of IonWorks and Q-patch assays successfully identified the nicotinamide derivative 4. Its increasing decay time constant (tau) of Nav1.1 currents at 0.03 µM along with significant selectivity against Nav1.2, Nav1.5, and Nav1.6 and acceptable brain exposure in mice was observed. Compound 4 is a promising Nav1.1 activator that can be used to analyze pathophysiological functions of the Nav1.1 channel towards treating various central nervous system diseases.


Subject(s)
Drug Discovery , Niacinamide/analogs & derivatives , Niacinamide/pharmacology , Pyrrolidines/pharmacology , Voltage-Gated Sodium Channel Agonists/pharmacology , Animals , Blood-Brain Barrier/metabolism , CHO Cells , Cricetulus , Mice , Molecular Structure , NAV1.1 Voltage-Gated Sodium Channel/metabolism , Niacinamide/chemical synthesis , Pyrrolidines/chemical synthesis , Pyrrolidines/chemistry , Structure-Activity Relationship , Voltage-Gated Sodium Channel Agonists/chemical synthesis , Voltage-Gated Sodium Channel Agonists/chemistry
15.
Br J Pharmacol ; 175(14): 2926-2939, 2018 07.
Article in English | MEDLINE | ID: mdl-29791744

ABSTRACT

BACKGROUND AND PURPOSE: Pharmacological agents that either inhibit or enhance flux of ions through voltage-gated sodium (Nav ) channels may provide opportunities for treatment of human health disorders. During studies to characterize agents that modulate Nav 1.3 function, we identified a compound that appears to exhibit both enhancement and inhibition of sodium ion conduction that appeared to be dependent on the gating state that the channel was in. The objective of the current study was to determine if these different modulatory effects are mediated by the same or distinct interactions with the channel. EXPERIMENTAL APPROACH: Electrophysiology and site-directed mutation were used to investigate the effects of PF-06526290 on Nav channel function. KEY RESULTS: PF-06526290 greatly slows inactivation of Nav channels in a subtype-independent manner. However, upon prolonged depolarization to induce inactivation, PF-06526290 becomes a Nav subtype-selective inhibitor. Mutation of the domain 4 voltage sensor modulates inhibition of Nav 1.3 or Nav 1.7 channels by PF-06526290 but has no effect on PF-06526290 mediated slowing of inactivation. CONCLUSIONS AND IMPLICATIONS: These findings suggest that distinct interactions may underlie the two modes of Nav channel modulation by PF-06526290 and that a single compound can affect sodium channel function in several ways.


Subject(s)
Sulfonamides/pharmacology , Thiazoles/pharmacology , Voltage-Gated Sodium Channel Agonists/pharmacology , Voltage-Gated Sodium Channel Blockers/pharmacology , Voltage-Gated Sodium Channels/physiology , Animals , CHO Cells , Cricetulus , Ganglia, Spinal , HEK293 Cells , Humans , Mice , Neurons/drug effects , Neurons/physiology
16.
Sci Rep ; 7(1): 16329, 2017 11 27.
Article in English | MEDLINE | ID: mdl-29180755

ABSTRACT

Voltage-gated sodium (Nav) channels play a key role in generating action potentials which leads to physiological signaling in excitable cells. The availability of probes for functional studies of mammalian Nav is limited. Here, by introducing two amino acid substitutions into the beta scorpion toxin Ts1, we have chemically synthesized a novel binder [S14R, W50Pra]Ts1 for Nav with high affinity, low dissociation rate and reduced toxicity while retaining the capability of conjugating Ts1 with molecules of interests for different applications. Using the fluorescent-dye conjugate, [S14R, W50Pra(Bodipy)]Ts1, we confirmed its binding to Nav1.4 through Lanthanide-based Resonance Energy Transfer. Moreover, using the gold nanoparticle conjugate, [S14R, W50Pra(AuNP)]Ts1, we were able to optically stimulate dorsal root ganglia neurons and generate action potentials with visible light via the optocapacitive effect as previously reported. [S14R, W50Pra]Ts1 is a novel probe with great potential for wider applications in Nav-related neuroscience research.


Subject(s)
Scorpion Venoms/chemistry , Scorpion Venoms/metabolism , Voltage-Gated Sodium Channels/chemistry , Voltage-Gated Sodium Channels/metabolism , Action Potentials/drug effects , Amino Acid Sequence , Animals , Binding Sites , Female , Gold , Ligands , Metal Nanoparticles , Models, Molecular , Neurons/drug effects , Neurons/physiology , Oocytes/drug effects , Oocytes/physiology , Patch-Clamp Techniques , Protein Binding , Protein Conformation , Rats , Scorpion Venoms/chemical synthesis , Scorpion Venoms/genetics , Structure-Activity Relationship , Voltage-Gated Sodium Channel Agonists
17.
Int J Biol Macromol ; 104(Pt A): 70-77, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28591591

ABSTRACT

Neuronal excitability controls the expression of a variety of genes and proteins and therefore regulates neurite outgrowth and synapse formation, fundamental physiological processes controlling learning and memory. Scorpion venom contains many neurotoxins which alter ion channel activities that influence neuronal excitability. In this study, a novel scorpion peptide termed BmK NT2 was purified from venom of Chinese scorpion Buthus martensii Karsch by combining mass spectrum mapping and intracellular Ca2+ concentration measurement in primary cultured neocortical neurons. Electrophysiological experiments demonstrated that BmK NT2 concentration-dependently delayed inactivation of voltage-gated sodium channels (VGSCs) with an EC50 value of 0.91µM, and shifted the steady-state activation and inactivation of VGSCs to hyperpolarized direction. The effects of BmK NT2 on electrophysiological characteristics of VGSCs were similar to that of α-scorpion toxins. BmK NT2 altered Ca2+ dynamics and increased phosphorylation of extracellular-regulated protein kinases (ERK) 1/2 and cAMP-response element binding (CREB) proteins, which were eliminated by the VGSC blocker, tetrodotoxin. These data demonstrate that BmK NT2 is a novel VGSC α-scorpion toxin which is sufficient to increase the phosphorylation of ERK1/2 and CREB proteins, suggesting that modulation of VGSC function by α-scorpion toxin exerts neurotrophic effect in primary cultured neocortical neurons.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neocortex/cytology , Neurons/metabolism , Scorpion Venoms/pharmacology , Voltage-Gated Sodium Channel Agonists/metabolism , Amino Acid Sequence , Calcium/metabolism , Phosphorylation/drug effects , Scorpion Venoms/chemistry
20.
Neuropharmacology ; 127: 87-108, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28416444

ABSTRACT

Toxins and venom components that target voltage-gated sodium (NaV) channels have evolved numerous times due to the importance of this class of ion channels in the normal physiological function of peripheral and central neurons as well as cardiac and skeletal muscle. NaV channel activators in particular have been isolated from the venom of spiders, wasps, snakes, scorpions, cone snails and sea anemone and are also produced by plants, bacteria and algae. These compounds have provided key insight into the molecular structure, function and pathophysiological roles of NaV channels and are important tools due to their at times exquisite subtype-selectivity. We review the pharmacology of NaV channel activators with particular emphasis on mammalian isoforms and discuss putative applications for these compounds. This article is part of the Special Issue entitled 'Venom-derived Peptides as Pharmacological Tools.'


Subject(s)
Peptides/pharmacology , Venoms/chemistry , Voltage-Gated Sodium Channel Agonists/pharmacology , Voltage-Gated Sodium Channels/drug effects , Animals , Ion Channel Gating/drug effects , Ion Channel Gating/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...