Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.206
Filter
1.
J Insect Physiol ; 139: 104385, 2022.
Article in English | MEDLINE | ID: mdl-35315336

ABSTRACT

Dorsal unpaired median (DUM) neurons, are a class of insect neurosecretory cells, which are involved in the control of several functions, such as excretion and reproduction, or the release of neurohormones. Previous studies demonstrated that they express different nicotinic acetylcholine receptor subtypes, in particular α-bungarotoxin-insensitive receptors, with nAChR1 and nAChR2 subtypes. Here, we demonstrated that pulse application of 1 mM nicotine (300 ms pulse duration) induced inward currents which were reduced under bath application of 15 µM calmidazolium, a calmodulin inhibitor. Bath application of 0.5 µM α-bungarotoxin had no effect on calmidazolium action, suggesting that it could have an indirect effect through α-bungarotoxin-insensitive receptors. Indeed, nicotine-evoked currents were reduced by 10 µM d-tubocurarine, and completely blocked by 5 µM mecamylamine, which affected nAChR1 and nAChR2 subtypes, respectively. Our results demonstrated that nAChR2 subtypes are involved in the indirect effect of calmidazolium. Moreover, we found that this calmidazolium effect was associated to a strong reduction in intracellular calcium levels after pulse application of 1 mM nicotine. Thus, compared to previous studies on mammalian cells, calmidazolium did not cause an increase in intracellular calcium levels in DUM neurons, suggesting that different calcium mechanisms are involved in the calmidazolium effect.


Subject(s)
Cockroaches , Nicotine , Animals , Bungarotoxins/pharmacology , Calcium , Imidazoles , Insecta , Mammals , Nicotine/pharmacology
2.
Toxins (Basel) ; 14(2)2022 01 21.
Article in English | MEDLINE | ID: mdl-35202107

ABSTRACT

α-bungarotoxin is a large, 74 amino acid toxin containing five disulphide bridges, initially identified in the venom of Bungarus multicinctus snake. Like most large toxins, chemical synthesis of α-bungarotoxin is challenging, explaining why all previous reports use purified or recombinant α-bungarotoxin. However, only chemical synthesis allows easy insertion of non-natural amino acids or new chemical functionalities. Herein, we describe a procedure for the chemical synthesis of a fluorescent-tagged α-bungarotoxin. The full-length peptide was designed to include an alkyne function at the amino-terminus through the addition of a pentynoic acid linker. Chemical synthesis of α-bungarotoxin requires hydrazide-based coupling of three peptide fragments in successive steps. After completion of the oxidative folding, an azide-modified Cy5 fluorophore was coupled by click chemistry onto the toxin. Next, we determined the efficacy of the fluorescent-tagged α-bungarotoxin to block acetylcholine (ACh)-mediated currents in response to muscle nicotinic receptor activation in TE671 cells. Using automated patch-clamp recordings, we demonstrate that fluorescent synthetic α-bungarotoxin has the expected nanomolar affinity for the nicotinic receptor. The blocking effect of fluorescent α-bungarotoxin could be displaced by incubation with a 20-mer peptide mimicking the α-bungarotoxin binding site. In addition, TE671 cells could be labelled with fluorescent toxin, as witnessed by confocal microscopy, and this labelling was partially displaced by the 20-mer competitive peptide. We thus demonstrate that synthetic fluorescent-tagged α-bungarotoxin preserves excellent properties for binding onto muscle nicotinic receptors.


Subject(s)
Bungarotoxins/chemical synthesis , Bungarotoxins/pharmacology , Cholinesterase Inhibitors/chemical synthesis , Cholinesterase Inhibitors/pharmacology , Fluorescent Dyes/chemistry , Acetylcholine , Cell Line , Click Chemistry , High-Throughput Screening Assays , Humans , Models, Molecular , Protein Conformation
3.
Proc Natl Acad Sci U S A ; 118(14)2021 04 06.
Article in English | MEDLINE | ID: mdl-33795516

ABSTRACT

The ability to monitor the release of neurotransmitters during synaptic transmission would significantly impact the diagnosis and treatment of neurological diseases. Here, we present a DNA-based enzymatic nanosensor for quantitative detection of acetylcholine (ACh) in the peripheral nervous system of living mice. ACh nanosensors consist of DNA as a scaffold, acetylcholinesterase as a recognition component, pH-sensitive fluorophores as signal generators, and α-bungarotoxin as a targeting moiety. We demonstrate the utility of the nanosensors in the submandibular ganglia of living mice to sensitively detect ACh ranging from 0.228 to 358 µM. In addition, the sensor response upon electrical stimulation of the efferent nerve is dose dependent, reversible, and we observe a reduction of ∼76% in sensor signal upon pharmacological inhibition of ACh release. Equipped with an advanced imaging processing tool, we further spatially resolve ACh signal propagation on the tissue level. Our platform enables sensitive measurement and mapping of ACh transmission in the peripheral nervous system.


Subject(s)
Acetylcholine/metabolism , Biosensing Techniques/methods , Ganglia, Parasympathetic/metabolism , Nanotechnology/methods , Acetylcholine/analysis , Acetylcholinesterase/chemistry , Acetylcholinesterase/metabolism , Animals , Bungarotoxins/pharmacology , Carbocyanines/chemistry , Cholinergic Antagonists/pharmacology , DNA/chemistry , Female , Fluorescent Dyes/chemistry , Ganglia, Parasympathetic/drug effects , Hydrogen-Ion Concentration , Male , Mice , Mice, Inbred C57BL , Receptors, Cholinergic/metabolism
4.
J Neurosci ; 41(1): 47-60, 2021 01 06.
Article in English | MEDLINE | ID: mdl-33203744

ABSTRACT

The lateral line (LL) is a sensory system that allows fish and amphibians to detect water currents. LL responsiveness is modulated by efferent neurons that aid in distinguishing between external and self-generated stimuli, maintaining sensitivity to relevant cues. One component of the efferent system is cholinergic, the activation of which inhibits afferent activity. LL hair cells (HCs) share structural, functional, and molecular similarities with those of the cochlea, making them a popular model for studying human hearing and balance disorders. Because of these commonalities, one could propose that the receptor at the LL efferent synapse is a α9α10 nicotinic acetylcholine receptor (nAChR). However, the identities of the molecular players underlying ACh-mediated inhibition in the LL remain unknown. Surprisingly, through the analysis of single-cell expression studies and in situ hybridization, we describe that α9, but not the α10, subunits are enriched in zebrafish HCs. Moreover, the heterologous expression of zebrafish α9 subunits indicates that homomeric receptors are functional and exhibit robust ACh-gated currents blocked by α-bungarotoxin and strychnine. In addition, in vivo Ca2+ imaging on mechanically stimulated zebrafish LL HCs show that ACh elicits a decrease in evoked Ca2+ signals, regardless of HC polarity. This effect is blocked by both α-bungarotoxin and apamin, indicating coupling of ACh-mediated effects to small-conductance Ca2+-activated potassium (SKs) channels. Our results indicate that an α9-containing (α9*) nAChR operates at the zebrafish LL efferent synapse. Moreover, the activation of α9* nAChRs most likely leads to LL HC hyperpolarization served by SK channels.SIGNIFICANCE STATEMENT The fish lateral line (LL) mechanosensory system shares structural, functional, and molecular similarities with those of the mammalian cochlea. Thus, it has become an accessible model for studying human hearing and balance disorders. However, the molecular players serving efferent control of LL hair cell (HC) activity have not been identified. Here we demonstrate that, different from the hearing organ of vertebrate species, a nicotinic acetylcholine receptor composed only of α9 subunits operates at the LL efferent synapse. Activation of α9-containing receptors leads to LL HC hyperpolarization because of the opening of small-conductance Ca2+-activated potassium channels. These results will further aid in the interpretation of data obtained from LL HCs as a model for cochlear HCs.


Subject(s)
Efferent Pathways/physiology , Lateral Line System/physiology , Parasympathetic Nervous System/physiology , Synapses/physiology , Animals , Bungarotoxins/pharmacology , Calcium Signaling/drug effects , Gene Expression Regulation , Hair Cells, Auditory/physiology , Nicotinic Antagonists/pharmacology , Oocytes , Physical Stimulation , Receptors, Nicotinic/drug effects , Small-Conductance Calcium-Activated Potassium Channels/drug effects , Strychnine/pharmacology , Xenopus , Zebrafish
5.
Eur J Histochem ; 64(s2)2020 Oct 30.
Article in English | MEDLINE | ID: mdl-33334089

ABSTRACT

Adipose-derived stem cells (ASCs) are an attractive source for regenerative medicine as they can be easily isolated, rapidly expandable in culture and show excellent in vitro differentiation potential. Acetylcholine (ACh), one of the main neurotransmitters in central and peripheral nervous systems, plays key roles in the control of several physiological processes also in non-neural tissues. As demonstrated in our previous studies, ACh can contribute to the rat ASCs physiology, negatively modulating ASCs proliferation and migration via M2 muscarinic receptor (mAChR) activation. In the present work we show that rat ASCs also express α7 nicotinic receptors (nAChRs). In particular, we have investigated the effects mediated by the selective activation of α7 nAChRs, which causes a reduction of ASC proliferation without affecting cell survival and morphology, and significantly promotes cell migration via upregulation of the CXCR4 expression. Interestingly, the activation of the α7 nAChR also upregulates the expression of M2 mAChR protein, indicating a cooperation between muscarinic and nicotinic receptors in the inhibition of ASC proliferation.


Subject(s)
Cell Movement/physiology , Cell Proliferation/physiology , Mesenchymal Stem Cells/metabolism , alpha7 Nicotinic Acetylcholine Receptor/metabolism , Adipose Tissue/cytology , Animals , Bungarotoxins/pharmacology , Fumarates/pharmacology , Male , Rats, Sprague-Dawley , Spiro Compounds/pharmacology , alpha7 Nicotinic Acetylcholine Receptor/agonists , alpha7 Nicotinic Acetylcholine Receptor/antagonists & inhibitors
6.
Dokl Biochem Biophys ; 493(1): 211-214, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32894468

ABSTRACT

The alpha7 nicotinic acetylcholine receptor (α7-nAChR) is considered a promising pharmacological target for the carcinoma therapy. We have previously shown that the recombinant analogue of the human protein SLURP-1 (rSLURP-1) effectively inhibits the growth of carcinomas of various origins via the interaction with α7-nAChR and down-regulation of expression of this receptor. Expression of α7-nAChR is increased in gliomas compared to healthy human brain tissues; however, the role of this receptor in the gliomas development is poorly understood. It was shown for the first time that rSLURP-1 significantly inhibits the growth of glioma model cells U251 MG and A172 up to ∼70%, which is comparable with the effect of α-bungarotoxin, a selective α7-nAChR inhibitor. The half-maximum effective concentrations of rSLURP-1 for U251 MG and A172 cells were 2.82 ± 0.2 and 8.9 ± 0.3 nM, respectively. Coincubation of U251 MG cells with rSLURP-1 and the nAChR inhibitor mecamylamine attenuates the antiproliferative activity of rSLURP-1, indicating nAChR as a molecular target for the rSLURP-1 action in gliomas.


Subject(s)
Antigens, Ly/pharmacology , Bungarotoxins/pharmacology , Glioma/drug therapy , Urokinase-Type Plasminogen Activator/pharmacology , alpha7 Nicotinic Acetylcholine Receptor/metabolism , Cell Line, Tumor , Cell Proliferation , Glioma/genetics , Glioma/metabolism , Humans , Recombinant Proteins/pharmacology
7.
Neuron ; 106(6): 952-962.e5, 2020 06 17.
Article in English | MEDLINE | ID: mdl-32275860

ABSTRACT

The nicotinic acetylcholine receptor, a pentameric ligand-gated ion channel, converts the free energy of binding of the neurotransmitter acetylcholine into opening of its central pore. Here we present the first high-resolution structure of the receptor type found in muscle-endplate membrane and in the muscle-derived electric tissues of fish. The native receptor was purified from Torpedo electric tissue and functionally reconstituted in lipids optimal for cryo-electron microscopy. The receptor was stabilized in a closed state by the binding of α-bungarotoxin. The structure reveals the binding of a toxin molecule at each of two subunit interfaces in a manner that would block the binding of acetylcholine. It also reveals a closed gate in the ion-conducting pore, formed by hydrophobic amino acid side chains, located ∼60 Å from the toxin binding sites. The structure provides a framework for understanding gating in ligand-gated channels and how mutations in the acetylcholine receptor cause congenital myasthenic syndromes.


Subject(s)
Bungarotoxins/metabolism , Electric Organ/metabolism , Receptors, Nicotinic/metabolism , Receptors, Nicotinic/ultrastructure , Animals , Binding Sites , Bungarotoxins/pharmacology , Carbachol/pharmacology , Cryoelectron Microscopy , Molecular Conformation , Nicotinic Agonists/pharmacology , Nicotinic Antagonists/pharmacology , Patch-Clamp Techniques , Protein Conformation , Receptors, Nicotinic/drug effects , Torpedo
8.
J Neurol Neurosurg Psychiatry ; 91(5): 526-532, 2020 05.
Article in English | MEDLINE | ID: mdl-32165373

ABSTRACT

OBJECTIVE: Direct inhibition of acetylcholine receptor (AChR) function by autoantibodies (Abs) is considered a rare pathogenic mechanism in myasthenia gravis (MG), but is usually studied on AChRs expressed in cell lines, rather than tightly clustered by the intracellular scaffolding protein, rapsyn, as at the intact neuromuscular junction. We hypothesised that clustered AChRs would provide a better target for investigating the functional effects of AChR-Abs. METHODS: Acetylcholine-induced currents were measured using whole-cell patch clamping and a fast perfusion system to assess fast (<2 min) functional effects of the serum samples. The sensitivity, specificity and rapidity of the system were first demonstrated by applying maternal AChR-Ab positive plasmas known to inhibit fetal AChR function in TE671 cells. Eleven previously untested AChR-Ab positive MG sera, 10 AChR-Ab negative MG sera and 5 healthy control sera were then applied to unclustered and rapsyn-clustered human adult AChRs in CN21 cells. RESULTS: The maternal AChR-Ab positive plasmas reduced fetal AChR currents, but not adult AChR currents, by >80% within 100 s. Only 2/11 AChR-Ab positive sera inhibited AChR currents in unclustered AChRs, but 6/11 AChR-Ab positive sera compared with none of the 10 AChR-Ab negative sera (p=0.0020) inhibited rapsyn-clustered AChR currents, and current inhibition by the AChR-Ab positive sera was greater when the AChRs were clustered (p=0.0385). None of the sera had detectable effects on desensitisation or recovery from desensitisation. CONCLUSION: These results show that antibodies can inhibit AChR function rapidly and demonstrate the importance of clustering in exploring pathogenic disease mechanisms of MG Abs.


Subject(s)
Autoantibodies/immunology , Muscle Proteins/immunology , Myasthenia Gravis/immunology , Receptors, Cholinergic/immunology , Adolescent , Adult , Aged , Bungarotoxins/pharmacology , Cell Line , Electrophysiological Phenomena , Female , Fluoxetine/pharmacology , Humans , Male , Microscopy, Fluorescence , Middle Aged , Myasthenia Gravis/etiology , Patch-Clamp Techniques , Receptors, Cholinergic/drug effects , Young Adult
9.
Biomolecules ; 10(3)2020 03 19.
Article in English | MEDLINE | ID: mdl-32204458

ABSTRACT

Alpha7 nicotinic acetylcholine receptors (α7nAChRs) are interesting not only because of their physiological effects, but because this receptor requires chaperones to traffic to cell surfaces (measured by alpha-bungarotoxin [αBGT] binding). While knockout (KO) animals and antibodies that react across species exist for tmem35a encoding the protein chaperone NACHO, commercially available antibodies against the chaperone RIC3 that allow Western blots across species have not been generally available. Further, no effects of deleting RIC3 function (ric3 KO) on α7nAChR expression are reported. Finally, antibodies against α7nAChRs have shown various deficiencies. We find mouse macrophages bind αBGT but lack NACHO. We also report on a new α7nAChR antibody and testing commercially available anti-RIC3 antibodies that react across species allowing Western blot analysis of in vitro cultures. These antibodies also react to specific RIC3 splice variants and single-nucleotide polymorphisms. Preliminary autoradiographic analysis reveals that ric3 KOs show subtle αBGT binding changes across different mouse brain regions, while tmem35a KOs show a complete loss of αBGT binding. These findings are inconsistent with effects observed in vitro, as RIC3 promotes αBGT binding to α7nAChRs expressed in HEK cells, even in the absence of NACHO. Collectively, additional regulatory factors are likely involved in the in vivo expression of α7nAChRs.


Subject(s)
Brain/metabolism , Gene Expression Regulation , Membrane Proteins/metabolism , alpha7 Nicotinic Acetylcholine Receptor/biosynthesis , Animals , Brain/pathology , Bungarotoxins/pharmacology , Gene Knockout Techniques , HEK293 Cells , Humans , Membrane Proteins/genetics , Mice , Mice, Knockout , alpha7 Nicotinic Acetylcholine Receptor/genetics
10.
Int Immunopharmacol ; 81: 106276, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32044666

ABSTRACT

Expression of α7 nicotinic acetylcholine receptors (nAChRs) on antigen presenting cells (APCs), such as macrophages and dendritic cells, is now well established. We have shown that GTS-21, a selective α7 nAChR agonist, downregulates APC-dependent CD4+ T cell differentiation into regulatory T cells (Tregs) and effector Th1, Th2 and Th17 cells by inhibiting antigen processing, thereby interfering with antigen presentation. α7 nAChRs on Jurkat human leukemic T cells require functional T cell receptors (TCRs)/CD3 and leukocyte-specific tyrosine kinase to mediate nicotine-induced Ca2+-signaling via Ca2+ release from intracellular stores, and are insensitive to two conventional α7 nAChR antagonists, α-bungarotoxin (α-BTX) and methyllycaconitine (MLA). We investigated the effects of GTS-21, α-BTX and MLA on ovalbumin (OVA)-induced Th cytokine release from spleen cells isolated from OVA-specific TCR transgenic DO11.10 mice. We found that: (1) GTS-21 dose-dependently suppresses OVA-induced IFN-γ, IL-4 and IL-17 release, but neither α-BTX nor MLA alone affected the OVA-induced cytokine release. (2) Neither α-BTX nor MLA abolished the suppressive effects of GTS-21 on IFN-γ and IL-17 release from OVA-activated DO11.10 spleen cells. (3) GTS-21 significantly suppressed OVA-induced APC-dependent CD4+ T cell differentiation into Tregs. Neither MLA nor mecamylamine, a non-specific nAChR antagonist, abolished the suppressive effect of GTS-21 on Treg differentiation. These results suggest that α7 nAChRs on APCs involved in cytokine synthesis and T cell differentiation are insensitive to the conventional α7 nAChR antagonists, α-BTX and MLA, and that α7 nAChRs on APCs differ pharmacologically from those in neurons.


Subject(s)
Aconitine/analogs & derivatives , Antigen-Presenting Cells/immunology , Bungarotoxins/pharmacology , CD4-Positive T-Lymphocytes/immunology , T-Lymphocytes, Regulatory/immunology , alpha7 Nicotinic Acetylcholine Receptor/metabolism , Aconitine/pharmacology , Animals , Antigen Presentation/drug effects , Benzylidene Compounds/pharmacology , Calcium Signaling/drug effects , Cell Differentiation/drug effects , Cells, Cultured , Cytokines/metabolism , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mice, Transgenic , Pyridines/pharmacology , alpha7 Nicotinic Acetylcholine Receptor/genetics
11.
Cell Physiol Biochem ; 53(4): 701-712, 2019.
Article in English | MEDLINE | ID: mdl-31592599

ABSTRACT

BACKGROUND/AIMS: Cholinergic signalling mediated by the activation of muscarinic and nicotinic receptors has been described in the literature as a classic and important signalling pathway in the regulation of the inflammatory response. Recent research has investigated the role of acetylcholine, the physiological agonist of these receptors, in the control of energy homeostasis at the central level. Studies have shown that mice that do not express acetylcholine in brain regions regulating energy homeostasis present with excessive weight gain and hyperphagia. However, it has not yet been well-described in the literature which cholinergic receptor subunits are involved in this response; moreover, the signalling pathways responsible for the observed effects are not fully delineated. The hypothalamus is the regulating centre of energy homeostasis, and the α7 subunit of the nicotinic acetylcholine receptor (α7nAChR) is highly expressed in this region. When active, α7nAChR recruits proteins such as JAK2/STAT3 to mediate its signalling; the same intracellular components are required by leptin, an anorexigenic hormone. The aim of the present study was to evaluate the role of the hypothalamic α7nAChR in the control of energy homeostasis. METHODS: The work was performed on Swiss male mice. Initially, using immunofluorescent staining on brain sections, the presence of α7nAChR in hypothalamic cells regulating energy homeostasis was evaluated. Animals were submitted to stereotaxis in the lateral ventricle and intracerebroventricular stimulation (ICV) was used for the administration of an agonist (PNU) or antagonist (α-bungarotoxin) of α7nAChR. Metabolic parameters were evaluated and the expression of neuropeptides was evaluated in the hypothalamus by real-time PCR and western blot. The expression of hypothalamic neuropeptides was evaluated in mice treated with siRNA or inhibitors of JAK2/STAT3 (AG490 and STATTIC) proteins. We also evaluated food intake in α7nAChR knockout animals (α7KO). Additionally, in mouse hypothalamic cell culture (the mypHoA-POMC/GFP lineage), we evaluated the expression of neuropeptides and pSTAT3 after stimulation with PNU. RESULTS: Our results indicate co-localisation of α7nAChR with α-MSH, AgRP and NPY in hypothalamic cells. Pharmacological activation of α7nAChR reduced food intake and increased hypothalamic POMC expression and decreased NPY and AgRP mRNA levels and the protein content of pAMPK. Inhibition of α7nAChR with an antagonist increased the mRNA content of NPY and AgRP. Inhibition of α7nAChR with siRNA led to the suppression of POMC expression and an increase in AgRP mRNA levels. α7KO mice showed no changes in food intake. Inhibition of proteins involved in the JAK2/STAT3 signalling pathway reversed the effects observed after PNU stimulation. POMC-GFP cells, when treated with PNU, showed increased POMC expression and nuclear translocation of pSTAT3. CONCLUSION: Thus, selective activation of α7nAChR is able to modulate important markers of the response to food intake, suggesting that α7nAChR activation can suppress the expression of orexigenic markers and favour the expression of anorexics using the intracellular JAK2/STAT3 machinery.


Subject(s)
Agouti-Related Protein/metabolism , Janus Kinase 2/metabolism , Pro-Opiomelanocortin/metabolism , STAT3 Transcription Factor/metabolism , alpha7 Nicotinic Acetylcholine Receptor/metabolism , Agouti-Related Protein/genetics , Animals , Benzamides/pharmacology , Bridged Bicyclo Compounds/pharmacology , Bungarotoxins/pharmacology , Cell Line , Eating/drug effects , Energy Metabolism/drug effects , Janus Kinase 2/antagonists & inhibitors , Janus Kinase 2/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/metabolism , Pro-Opiomelanocortin/genetics , RNA Interference , RNA, Small Interfering/metabolism , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/genetics , Signal Transduction/drug effects , alpha7 Nicotinic Acetylcholine Receptor/agonists , alpha7 Nicotinic Acetylcholine Receptor/antagonists & inhibitors , alpha7 Nicotinic Acetylcholine Receptor/genetics
12.
Blood Coagul Fibrinolysis ; 30(8): 379-384, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31415248

ABSTRACT

BACKGROUND: A phenomena of interest is the in vitro anticoagulant effects of neurotoxins found in elapid venoms that kill by paralysis. These enzymes include phospholipase A2 (PLA2), and it has recently been demonstrated that carbon monoxide inhibits the PLA2-dependent neurotoxin contained in Mojave rattlesnake type A venom. The purpose of this investigation was to assess if the anticoagulant activity of elapid venoms containing PLA2 and/or three finger toxins could be inhibited by carbon monoxide. METHODS: Venoms collected from Bungarus multicinctus, Micrurus fulvius, and five Naja species were exposed to carbon monoxide via carbon monoxide releasing molecule-2 prior to placement into human plasma. Coagulation kinetics were assessed via thrombelastography. RESULTS: Compared with plasma without venom addition, all venoms had significant anticoagulant effects, with a 160-fold range of concentrations having similar anticoagulant effects in a species-specific manner. Carbon monoxide significantly inhibited the anticoagulant effect of all venoms tested, but inhibition was not complete in all cases. CONCLUSION: Given that individual neurotoxin activity often depends on intact activity that includes anticoagulant action, it may be possible that carbon monoxide inhibits neurotoxicity. Future investigation is justified to assess such carbon monoxide mediated inhibition with purified neurotoxins in vitro and in vivo.


Subject(s)
Anticoagulants , Carbon Monoxide/pharmacology , Snake Venoms/pharmacology , Animals , Blood Coagulation/drug effects , Blood Specimen Collection , Bungarotoxins/antagonists & inhibitors , Bungarotoxins/chemistry , Bungarotoxins/pharmacology , Bungarus , Coral Snakes , Elapid Venoms/antagonists & inhibitors , Elapid Venoms/chemistry , Elapid Venoms/pharmacology , Elapidae , Humans , Neurotoxins/antagonists & inhibitors , Proteome/analysis , Snake Venoms/antagonists & inhibitors , Snake Venoms/chemistry , Thrombelastography
13.
Mol Cell Neurosci ; 98: 121-130, 2019 07.
Article in English | MEDLINE | ID: mdl-31212013

ABSTRACT

Kv4.2 voltage-gated K+ channel subunits, the primary source of the somatodendritic A-type K+ current in CA1 pyramidal neurons of the hippocampus, play important roles in regulating dendritic excitability and plasticity. To better study the trafficking and subcellular distribution of Kv4.2, we created and characterized a novel Kv4.2 construct encoding a bungarotoxin binding site in the extracellular S3-S4 linker region of the α-subunit. When expressed, this construct can be visualized in living cells after staining with rhodamine-conjugated bungarotoxin. We validated the utility of this construct by visualizing the spontaneous internalization and insertion of Kv4.2 in HEK 293T cells. We further report that Kv4.2 colocalized with several endosome markers in HEK 293T cells. In addition, Kv4.2 internalization is significantly impaired by mitogen-activated protein kinase (MAPK) inhibitors in transfected primary hippocampal neurons. Therefore, this newly developed BBS-Kv4.2 construct provides a novel and powerful tool for studying surface Kv4.2 channel localization and trafficking.


Subject(s)
Bungarotoxins/pharmacology , Shal Potassium Channels/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Binding Sites , Cells, Cultured , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/metabolism , HEK293 Cells , Hippocampus/cytology , Humans , Kv Channel-Interacting Proteins/metabolism , Mitogen-Activated Protein Kinase Kinases/metabolism , Neurons/drug effects , Neurons/metabolism , Protein Binding , Protein Kinase Inhibitors/pharmacology , Protein Transport , Rats , Shal Potassium Channels/chemistry , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
14.
Neurotoxicology ; 74: 132-138, 2019 09.
Article in English | MEDLINE | ID: mdl-31212017

ABSTRACT

Cockroach neurosecretory cells, dorsal unpaired median (DUM) neurons, express two distinct α-bungarotoxin-insensitive nicotinic acetylcholine receptor subtypes, nAChR1 and nAChR2 which are differently sensitive to the neonicotinoid insecticides and intracellular calcium pathways. The aim of this study is to determine whether sulfoxaflor acts as an agonist of nAChR1 and nAChR2 subtypes. We demonstrated that 1 mM sulfoxaflor induced high current amplitudes, compared to acetylcholine, suggesting that it was a full agonist of DUM neuron nAChR subtypes. Sulfoxaflor evoked currents were not inhibited by the nicotinic acetylcholine receptor antagonist d-tubocurarine (dTC) which reduced nAChR1. But, sulfoxaflor evoked currents were reduced in the presence of 5 µM mecamylamine which is known to reduce nAChR2 subtype. Interestingly, when 1 µM imidacloprid was added in the extracellular solution, sulfoxaflor-induced currents were significantly suppressed. Moreover, when extracellular calcium concentration was increased, bath application of 1 µM imidacloprid partially reduced sulfoxaflor activated currents when nAChR1 was inhibited with 20 µM dTC and completely suppressed sulfoxaflor currents when nAChR2 was inhibited with 5 µM mecamylamine. Our data demonstrated therefore that sulfoxaflor activates both nAChR1 and nAChR2 subtypes.


Subject(s)
Bungarotoxins/pharmacology , Cholinergic Agents/pharmacology , Cockroaches , Neonicotinoids/pharmacology , Nicotinic Agonists/pharmacology , Nitro Compounds/pharmacology , Pyridines/pharmacology , Receptors, Nicotinic/drug effects , Sulfur Compounds/pharmacology , Acetylcholine/pharmacology , Animals , Calcium/pharmacology , Mecamylamine/pharmacology , Nicotinic Antagonists/pharmacology , Patch-Clamp Techniques , Pyridines/antagonists & inhibitors , Sulfur Compounds/antagonists & inhibitors , Tubocurarine/toxicity
15.
Neuropharmacology ; 157: 107670, 2019 10.
Article in English | MEDLINE | ID: mdl-31202608

ABSTRACT

Processing of the amyloid precursor protein by alternative secretases results in ectodomain shedding of either secreted amyloid precursor protein-α (sAPPα) or its counterpart secreted amyloid precursor protein-ß (sAPPß). Although sAPPα contains only 16 additional amino acids at its C-terminus compared to sAPPß, it displays significantly greater potency in neuroprotection, neurotrophism and enhancement of long-term potentiation (LTP). In the current study, this 16 amino acid peptide sequence (CTα16) was characterised for its ability to replicate the synaptic plasticity-enhancing properties of sAPPα. An N-acetylated version of CTα16 produced concentration-dependent increases in the induction and persistence of LTP at Schaffer collateral/commissural synapses in area CA1 of young adult rat hippocampal slices. A scrambled peptide had no effect. CTα16 significantly enhanced de novo protein synthesis, and correspondingly its enhancement of LTP was blocked by the protein synthesis inhibitor cycloheximide, as well as by the α7-nicotinic receptor blocker α-bungarotoxin. The impaired LTP of 14-16 month old APPswe/PS1dE9 transgenic mice, a mouse model of Alzheimer's disease, was completely restored to the wild-type level by CTα16. These results indicate that the CTα16 peptide fragment of sAPPα mimics the larger protein's functionality with respect to LTP, stimulation of protein synthesis and activation of α7-nAChRs, and thus like sAPPα may have potential as a therapeutic agent against the plasticity and cognitive deficits observed in AD and other neurological disorders.


Subject(s)
Alzheimer Disease/physiopathology , Long-Term Potentiation/drug effects , Alzheimer Disease/genetics , Animals , Bungarotoxins/pharmacology , CA1 Region, Hippocampal/physiology , Cycloheximide/pharmacology , Dose-Response Relationship, Drug , Gene Expression/drug effects , Male , Mice , Mice, Transgenic , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/pharmacology , Rats
16.
Br J Pharmacol ; 176(18): 3475-3488, 2019 09.
Article in English | MEDLINE | ID: mdl-30981214

ABSTRACT

BACKGROUND AND PURPOSE: Progressive dysfunction of cholinergic transmission is a well-known characteristic of Alzheimer's disease (AD). Amyloid ß (Aß) peptide oligomers are known to play a central role in AD and are suggested to impair the function of the cholinergic nicotinic ACh receptor α7 (α7nAChR). However, the mechanism underlying the effect of Aß on α7nAChR function is not fully understood, limiting the therapeutic exploration of this observation in AD. Here, we aimed to detect and characterize Aß binding to α7nAChR, including the possibility of interfering with this interaction for therapeutic purposes. EXPERIMENTAL APPROACH: We developed a specific and quantitative time-resolved FRET (TR-FRET)-based binding assay for Aß to α7nAChR and pharmacologically characterized this interaction. KEY RESULTS: We demonstrated specific and high-affinity (low nanomolar) binding of Aß to the orthosteric binding site of α7nAChR. Aß binding was prevented and reversed by the well-characterized orthosteric ligands of α7nAChR (epibatidine, α-bungarotoxin, methylylcaconitine, PNU-282987, S24795, and EVP6124) and by the type II positive allosteric modulator (PAM) PNU-120596 but not by the type I PAM NS1738. CONCLUSIONS AND IMPLICATIONS: Our TR-FRET Aß binding assay demonstrates for the first time the specific binding of Aß to α7nAChR, which will be a crucial tool for the development, testing, and selection of a novel generation of AD drug candidates targeting Aß/α7nAChR complexes with high specificity and fewer side effects compared to currently approved α7nAChR drugs. LINKED ARTICLES: This article is part of a themed section on Therapeutics for Dementia and Alzheimer's Disease: New Directions for Precision Medicine. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.18/issuetoc.


Subject(s)
Amyloid beta-Peptides/metabolism , alpha7 Nicotinic Acetylcholine Receptor/metabolism , Aconitine/analogs & derivatives , Aconitine/pharmacology , Benzamides/pharmacology , Bridged Bicyclo Compounds/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Bungarotoxins/pharmacology , HEK293 Cells , Humans , Isoxazoles/pharmacology , Ligands , Phenylurea Compounds/pharmacology , Pyridines/pharmacology , Pyridinium Compounds/pharmacology , Quinuclidines/pharmacology , Thiophenes/pharmacology
17.
J Physiol ; 597(7): 1993-2006, 2019 04.
Article in English | MEDLINE | ID: mdl-30673133

ABSTRACT

KEY POINTS: Acetylcholine receptors are aggregated in the central regions of intrafusal muscle fibres. Single unit muscle spindle afferent responses from isolated mouse extensor digitorum longus muscle were recorded in the absence of fusimotor input to ramp and hold stretches as well as to sinusoidal vibrations in the presence and absence of the acetylcholine receptor blockers d-tubocurarine and α-bungarotoxin. Proprioceptive afferent responses to both types of stretch were enhanced in the presence of either blocker. Blocking acetylcholine uptake and vesicular acetylcholine release by hemicholinium-3 also enhanced stretch-evoked responses. These results represent the first evidence that acetylcholine receptors negatively modulate muscle spindle responses to stretch. The data support the hypothesis that the sensory nerve terminal is able to release vesicles to fine-tune proprioceptive afferent sensitivity. ABSTRACT: Muscle spindles are complex stretch-sensitive mechanoreceptors. They consist of specialized skeletal muscle fibres, called intrafusal fibres, which are innervated in the central (equatorial) region by afferent sensory axons and in both polar regions by efferent γ-motoneurons. Previously it was shown that acetylcholine receptors (AChR) are concentrated in the equatorial region at the contact site between the sensory neuron and the intrafusal muscle fibre. To address the function of these AChRs, single unit sensory afferents were recorded from an isolated mouse extensor digitorum longus muscle in the absence of γ-motoneuron activity. Specifically, we investigated the responses of individual sensory neurons to ramp-and-hold stretches and sinusoidal vibrations before and after the addition of the competitive and non-competitive AChR blockers d-tubocurarine and α-bungarotoxin, respectively. The presence of either drug did not affect the resting action potential discharge frequency. However, the action potential frequencies in response to stretch were increased. In particular, frequencies of the dynamic peak and dynamic index to ramp-and-hold stretches were significantly higher in the presence of either drug. Treatment of muscle spindle afferents with the high-affinity choline transporter antagonist hemicholinium-3 similarly increased muscle spindle afferent firing frequencies during stretch. Moreover, the firing rate during sinusoidal vibration stimuli at low amplitudes was higher in the presence of α-bungarotoxin compared to control spindles also indicating an increased sensitivity to stretch. Collectively these data suggest a modulation of the muscle spindle afferent response to stretch by AChRs in the central region of intrafusal fibres possibly fine-tuning muscle spindle sensitivity.


Subject(s)
Muscle Fibers, Skeletal/physiology , Muscle Spindles/physiology , Receptors, Cholinergic/metabolism , Action Potentials/drug effects , Animals , Bungarotoxins/pharmacology , Hemicholinium 3/pharmacology , Male , Mechanotransduction, Cellular , Mice , Mice, Inbred C57BL , Protein Transport , Sensory Receptor Cells , Tubocurarine/pharmacology
18.
Auton Neurosci ; 216: 25-32, 2019 01.
Article in English | MEDLINE | ID: mdl-30206032

ABSTRACT

Sympathetic neurons of SCG are dependent on availability of nerve growth factor (NGF) for their survival. SCG neurons express nicotinic receptors (nAChR) whose expression levels are modulated by nicotine. Nicotine exerts multiple effects on neurons, including neuroprotection, through nAChR binding. Although sympathetic neurons express robust levels of nAChR, a possible neuroprotective role for nicotine in these neurons is not well-understood. Therefore we determined the effect of nicotine exposure on survival of SCG neurons during NGF withdrawal in a well-established cell culture system. NGF was withdrawn in rat neonatal SCG neuron cultures which were then treated with either 10 µM nicotine alone or with nAChR antagonists 0.1 µM α-bungarotoxin (antagonist for α7 subunit bearing nAChR) and 10 µM mecamylamine (non-specific antagonist for ganglionic nAChR) for 48 h. Apoptotic death was determined by TUNEL staining. Cell survival was also determined by MTS assay. Western blot analysis of ERK1/2 was also performed. Our results showed that exposure to 10 µM nicotine significantly reduced apoptotic cell death in SCG neurons resulting from NGF withdrawal as shown by fewer TUNEL positive cells. The MTS assay results also revealed that 10 µM nicotine concentration significantly increased cell survival thus indicating neuroprotective effect of nicotine against cell death resulting from NGF withdrawal. Nicotinic receptor antagonists (bungarotoxin & mecamylamine) attenuated the effect of nicotine's action of neuroprotection. Western blot analysis showed an increased expression of ERK1/2 in nicotine treated cultures suggesting nicotine provided neuroprotection in SCG neurons by increasing the expression of ERK1/2 through nicotinic receptor dependent mechanisms.


Subject(s)
Neurons/drug effects , Neuroprotection/drug effects , Nicotine/pharmacology , Superior Cervical Ganglion/cytology , Animals , Animals, Newborn , Apoptosis/drug effects , Bungarotoxins/pharmacology , Cell Survival/drug effects , Cells, Cultured , Mecamylamine/pharmacology , Mitogen-Activated Protein Kinase 1/biosynthesis , Mitogen-Activated Protein Kinase 3/biosynthesis , Nerve Growth Factor/deficiency , Neurons/metabolism , Neuroprotective Agents/pharmacology , Rats , Superior Cervical Ganglion/drug effects
19.
Anal Chem ; 90(17): 10318-10325, 2018 09 04.
Article in English | MEDLINE | ID: mdl-30073836

ABSTRACT

The fruit fly, Drosophila melanogaster, is a popular model organism for studying neurological processes and diseases due to the availability of sophisticated genetic tools. While endogenous neurotransmitter release has been characterized in Drosophila larvae, here, we measured endogenous dopamine release in isolated adult Drosophila brains for the first time. Dopamine was measured with fast-scan cyclic voltammetry (FSCV), and acetylcholine or nicotine were used as the stimulus, as both interact with nicotinic acetylcholine receptors (nAChRs) to evoke endogenous dopamine release. Stimulations with 10 pmol of acetylcholine elicited 0.26 ± 0.05 µM dopamine, while 70 fmol nicotine stimulations evoked 0.29 ± 0.03 µM in the central complex. Nicotine-stimulated dopamine release lasted much longer than acetylcholine-stimulated release. Dopamine release is reduced in the presence of nAChR antagonist α-bungarotoxin and the sodium channel blocker tetrodotoxin, indicating release is mediated by nAChRs and exocytosis. The identity of dopamine was confirmed by using 3-iodotyrosine, a dopamine synthesis inhibitor, and by confirming that release was not changed in octopamine synthesis mutant flies, Tdc2 RO54. Additionally, the half-decay time ( t50) in fumin (67 ± 15 s), dopamine transporter mutant flies, was larger than in wild-type flies (16 ± 3.7 s) further proving that acetylcholine stimulation evokes dopamine release. This study demonstrates that stimulation of nAChRs can be used to elicit endogenous dopamine release in adult fly brains, which will be a useful technique for future studies probing dopamine changes during aging or in neurodegenerative diseases.


Subject(s)
Acetylcholine/pharmacology , Brain/drug effects , Dopamine/metabolism , Drosophila melanogaster/metabolism , Electrochemical Techniques/methods , Animals , Brain/metabolism , Bungarotoxins/pharmacology , Dopamine/biosynthesis , Dopamine Antagonists/pharmacology , Exocytosis/drug effects , Monoiodotyrosine/pharmacology , Nicotine/pharmacology , Nicotinic Antagonists/pharmacology , Octopamine/biosynthesis , Receptors, Nicotinic/metabolism , Reproducibility of Results , Sodium Channel Blockers/pharmacology , Tetrodotoxin/pharmacology
20.
Toxicon ; 152: 9-15, 2018 Sep 15.
Article in English | MEDLINE | ID: mdl-30012473

ABSTRACT

The Kv1.3 channel plays potential roles in immune, inflammation and coagulation system. Many studies showed that Kv1.3 channel inhibitors have immunosuppressive and anti-inflammatory activities, but no Kv1.3 channel inhibitors have been found to have anticoagulation activities. Here, based on our previous work about Kv1.3 channel toxin peptide inhibitors, we first attempt to test anticoagulation activities of four known venom-derived Kv1.3 channel inhibitors with different structural folds: BmKTX with CSα/ß structural fold, OmTx3 with CSα/α structural fold, BF9 with Kuntz-type structural fold, and SjAPI-2 with Ascaris-type structural fold. Our results showed that BmKTX and OmTx3 have no activities towards both intrinsic and extrinsic coagulation pathway, SjAPI-2 just has weak activity towards intrinsic coagulation pathway, and BF9 has potent activity towards intrinsic coagulation pathway with no apparent effect on extrinsic coagulation pathway. Enzyme and inhibitor reaction kinetics experiments further showed that BF9 inhibited intrinsic coagulation pathway-associated coagulation factor XIa, but have no apparent effects on common coagulation pathway coagulation factor IIa. Structure-activity relationship showed that Gly14, Asn17, Ala18 and Ile20 of BF9 are main residues involved in the inhibiting effect on factor XIa. To the best of our knowledge, BF9 is the first anticoagulant with Kv1.3 channel inhibitory activity. Together, our present studies found the first dual functional peptides with Kv1.3 channel and coagulation factor XIa inhibitory activities, and provided a new molecular template for the lead drug discovery towards immune and thrombosis-associated human diseases.


Subject(s)
Anticoagulants/pharmacology , Bungarotoxins/pharmacology , Factor XIa/antagonists & inhibitors , Kv1.3 Potassium Channel/antagonists & inhibitors , Blood Coagulation/drug effects , Humans , Kinetics , Scorpion Venoms/pharmacology , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...