Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
Infect Disord Drug Targets ; 24(1): e230623218222, 2024.
Article in English | MEDLINE | ID: mdl-37357523

ABSTRACT

Bee venoms are well-known for their important biological activities. More specifically, the venom of Apis mellifera syriaca was shown to exhibit various biological effects, including antimicrobial effects. It is suggested that the anti-microbial effect of venom could be accompanied by an immunomodulatory response in the host favoring anti-inflammatory responses. Thus, in this work, we investigated, for the first time, the immunomodulatory effects of A. mellifera syriaca venom in mice. Firstly, it was found that this venom exhibited mild toxicity in BALB/c mice after intraperitoneal injection with an LD50 of 3.8 mg/kg. We then investigated its immunomodulatory effects by evaluating the splenic levels of both pro- and anti-inflammatory cytokines in mice by ELISA. Interestingly, at 1 mg/kg, A. mellifera syriaca venom induced a decrease in IFN-γ, TNF-α, IL-4, and IL-10 at 24h postinjection. At a higher dose (3 mg/kg), an increase in IFN-γ and IL-4 levels was observed, while the levels of TNF-α and IL-10 remained low compared to the control. Altogether, these preliminary data suggest that A. mellifera syriaca venom exhibits anti-inflammatory effects at a sublethal dose (1 mg/kg), while at a higher dose (3 mg/kg), it induces inflammatory effects.


Subject(s)
Cytokines , Interleukin-10 , Mice , Bees , Animals , Tumor Necrosis Factor-alpha , Mice, Inbred BALB C , Interleukin-4 , Anti-Inflammatory Agents/pharmacology
2.
Membranes (Basel) ; 13(10)2023 Oct 15.
Article in English | MEDLINE | ID: mdl-37888003

ABSTRACT

The Xenopus is a special study model in experimental research [...].

3.
Mar Drugs ; 21(3)2023 Mar 22.
Article in English | MEDLINE | ID: mdl-36976245

ABSTRACT

Tetrodotoxin (TTX) poisoning through the consumption of contaminated fish leads to lethal symptoms, including severe hypotension. This TTX-induced hypotension is likely due to the downfall of peripheral arterial resistance through direct or indirect effects on adrenergic signaling. TTX is a high-affinity blocker of voltage-gated Na+ (NaV) channels. In arteries, NaV channels are expressed in sympathetic nerve endings, both in the intima and media. In this present work, we aimed to decipher the role of NaV channels in vascular tone using TTX. We first characterized the expression of NaV channels in the aorta, a model of conduction arteries, and in mesenteric arteries (MA), a model of resistance arteries, in C57Bl/6J mice, by Western blot, immunochemistry, and absolute RT-qPCR. Our data showed that these channels are expressed in both endothelium and media of aorta and MA, in which scn2a and scn1b were the most abundant transcripts, suggesting that murine vascular NaV channels consist of NaV1.2 channel subtype with NaVß1 auxiliary subunit. Using myography, we showed that TTX (1 µM) induced complete vasorelaxation in MA in the presence of veratridine and cocktails of antagonists (prazosin and atropine with or without suramin) that suppressed the effects of neurotransmitter release. In addition, TTX (1 µM) strongly potentiated the flow-mediated dilation response of isolated MA. Altogether, our data showed that TTX blocks NaV channels in resistance arteries and consecutively decreases vascular tone. This could explain the drop in total peripheral resistance observed during mammal tetrodotoxications.


Subject(s)
Aorta , Mesenteric Arteries , Mice , Animals , Tetrodotoxin/pharmacology , Mammals , Voltage-Gated Sodium Channel beta-1 Subunit
4.
Cell Mol Life Sci ; 79(10): 525, 2022 Sep 20.
Article in English | MEDLINE | ID: mdl-36125552

ABSTRACT

Understanding temperature production and regulation in endotherm organisms becomes a crucial challenge facing the increased frequency and intensity of heat strokes related to global warming. Mitochondria, located at the crossroad of metabolism, respiration, Ca2+ homeostasis, and apoptosis, were recently proposed to further act as cellular radiators, with an estimated inner temperature reaching 50 °C in common cell lines. This inner thermogenesis might be further exacerbated in organs devoted to produce consistent efforts as muscles, or heat as brown adipose tissue, in response to acute solicitations. Consequently, pathways promoting respiratory chain uncoupling and mitochondrial activity, such as Ca2+ fluxes, uncoupling proteins, futile cycling, and substrate supplies, provide the main processes controlling heat production and cell temperature. The mitochondrial thermogenesis might be further amplified by cytoplasmic mechanisms promoting the over-consumption of ATP pools. Considering these new thermic paradigms, we discuss here all conventional wisdoms linking mitochondrial functions to cellular thermogenesis in different physiological conditions.


Subject(s)
Mitochondria , Thermogenesis , Adenosine Triphosphate/metabolism , Adipose Tissue, Brown/metabolism , Mitochondria/metabolism , Mitochondrial Uncoupling Proteins/metabolism , Thermogenesis/physiology
5.
Molecules ; 27(13)2022 Jun 28.
Article in English | MEDLINE | ID: mdl-35807390

ABSTRACT

Voltage-gated Na+ (NaV) channels are significant therapeutic targets for the treatment of cardiac and neurological disorders, thus promoting the search for novel NaV channel ligands. With the objective of discovering new blockers of NaV channel ligands, we screened an In-House vegetal alkaloid library using fluorescence cell-based assays. We screened 62 isoquinoline alkaloids (IA) for their ability to decrease the FRET signal of voltage sensor probes (VSP), which were induced by the activation of NaV channels with batrachotoxin (BTX) in GH3b6 cells. This led to the selection of five IA: liriodenine, oxostephanine, thalmiculine, protopine, and bebeerine, inhibiting the BTX-induced VSP signal with micromolar IC50. These five alkaloids were then assayed using the Na+ fluorescent probe ANG-2 and the patch-clamp technique. Only oxostephanine and liriodenine were able to inhibit the BTX-induced ANG-2 signal in HEK293-hNaV1.3 cells. Indeed, liriodenine and oxostephanine decreased the effects of BTX on Na+ currents elicited by the hNaV1.3 channel, suggesting that conformation change induced by BTX binding could induce a bias in fluorescent assays. However, among the five IA selected in the VSP assay, only bebeerine exhibited strong inhibitory effects against Na+ currents elicited by the hNav1.2 and hNav1.6 channels, with IC50 values below 10 µM. So far, bebeerine is the first BBIQ to have been reported to block NaV channels, with promising therapeutical applications.


Subject(s)
Alkaloids , Fluorescent Dyes , Alkaloids/pharmacology , Batrachotoxins/metabolism , Batrachotoxins/pharmacology , Bias , HEK293 Cells , Humans , Isoquinolines/pharmacology , Ligands , Sodium/metabolism
6.
Biology (Basel) ; 11(6)2022 Jun 09.
Article in English | MEDLINE | ID: mdl-35741410

ABSTRACT

The complications following snake bite envenoming are due to the venom's biological activities, which can act on different systems of the prey. These activities arise from the fact that snake venoms are rich in bioactive molecules, which are also of interest for designing drugs. The venom of Montivipera bornmuelleri, known as the Lebanon viper, has been shown to exert antibacterial, anticancer, and immunomodulatory effects. However, the venom's activity on the nervous system has not yet been studied, and its effect on the cardiovascular system needs further investigation. Because zebrafish is a convenient model to study tissue alterations induced by toxic agents, we challenged it with the venom of Montivipera bornmuelleri. We show that this venom leads to developmental toxicity but not teratogenicity in zebrafish embryos. The venom also induces neurotoxic effects and disrupts the zebrafish cardiovascular system, leading to heartbeat rate reduction and hemorrhage. Our findings demonstrate the potential neurotoxicity and cardiotoxicity of M. bornmuelleri's venom, suggesting a multitarget strategy during envenomation.

8.
Int J Mol Sci ; 23(2)2022 Jan 13.
Article in English | MEDLINE | ID: mdl-35055012

ABSTRACT

Thanks to the crosstalk between Na+ and Ca2+ channels, Na+ and Ca2+ homeostasis interplay in so-called excitable cells enables the generation of action potential in response to electrical stimulation. Here, we investigated the impact of persistent activation of voltage-gated Na+ (NaV) channels by neurotoxins, such as veratridine (VTD), on intracellular Ca2+ concentration ([Ca2+]i) in a model of excitable cells, the rat pituitary GH3b6 cells, in order to identify the molecular actors involved in Na+-Ca2+ homeostasis crosstalk. By combining RT-qPCR, immunoblotting, immunocytochemistry, and patch-clamp techniques, we showed that GH3b6 cells predominantly express the NaV1.3 channel subtype, which likely endorses their voltage-activated Na+ currents. Notably, these Na+ currents were blocked by ICA-121431 and activated by the ß-scorpion toxin Tf2, two selective NaV1.3 channel ligands. Using Fura-2, we showed that VTD induced a [Ca2+]i increase. This effect was suppressed by the selective NaV channel blocker tetrodotoxin, as well by the selective L-type CaV channel (LTCC) blocker nifedipine. We also evidenced that crobenetine, a NaV channel blocker, abolished VTD-induced [Ca2+]i elevation, while it had no effects on LTCC. Altogether, our findings highlight a crosstalk between NaV and LTCC in GH3b6 cells, providing a new insight into the mode of action of neurotoxins.


Subject(s)
Large-Conductance Calcium-Activated Potassium Channels/metabolism , Signal Transduction/drug effects , Voltage-Gated Sodium Channels/metabolism , Animals , Calcium/metabolism , Cell Line , Electrophysiological Phenomena , Fluorescent Antibody Technique , Gene Expression , High-Throughput Screening Assays , Ion Channel Gating/drug effects , Large-Conductance Calcium-Activated Potassium Channels/genetics , Neurotoxins/pharmacology , Patch-Clamp Techniques , Protein Binding , Protein Isoforms , Rats , Voltage-Gated Sodium Channels/genetics
9.
Front Neurosci ; 15: 768466, 2021.
Article in English | MEDLINE | ID: mdl-34912189

ABSTRACT

Fipronil (FPN) is a worldwide-used neurotoxic insecticide, targeting, and blocking GABAA receptors (GABAARs). Beyond its efficiency on insect GABAARs, FPN causes neurotoxic effects in humans and mammals. Here, we investigated the mode of action of FPN on mammalian α6-containing GABAARs to understand its inhibitory effects on GABA-induced currents, as a function of the synaptic or extrasynaptic localization of GABAARs. We characterized the effects of FPN by electrophysiology using Xenopus oocytes which were microtransplanted with cerebellum membranes or injected with α6ß3, α6ß3γ2S (synaptic), and α6ß3δ (extrasynaptic) cDNAs. At micromolar concentrations, FPN dose-dependently inhibited cerebellar GABA currents. FPN acts as a non-competitive antagonist on ternary receptors. Surprisingly, the inhibition of GABA-induced currents was partial for extra-synaptic (α6ß3δ) and binary (α6ß3) receptors, while synaptic α6ß3γ2S receptors were fully blocked, indicating that the complementary γ or δ subunit participates in FPN-GABAAR interaction. FPN unexpectedly behaved as a positive modulator on ß3 homopentamers. These data show that FPN action is driven by the subunit composition of GABAARs-highlighting the role of the complementary subunit-and thus their localization within a physiological synapse. We built a docking model of FPN on GABAARs, which reveals two putative binding sites. This is consistent with a double binding mode of FPN on GABAARs, possibly one being of high affinity and the other of low affinity. Physiologically, the γ/δ subunit incorporation drives its inhibitory level and has important significance for its toxicity on the mammalian nervous system, especially in acute exposure.

10.
Mar Drugs ; 19(10)2021 Oct 05.
Article in English | MEDLINE | ID: mdl-34677461

ABSTRACT

Voltage-gated sodium channels (VGSCs) are considered to be one of the most important ion channels given their remarkable physiological role. VGSCs constitute a family of large transmembrane proteins that allow transmission, generation, and propagation of action potentials. This occurs by conducting Na+ ions through the membrane, supporting cell excitability and communication signals in various systems. As a result, a wide range of coordination and physiological functions, from locomotion to cognition, can be accomplished. Drugs that target and alter the molecular mechanism of VGSCs' function have highly contributed to the discovery and perception of the function and the structure of this channel. Among those drugs are various marine toxins produced by harmful microorganisms or venomous animals. These toxins have played a key role in understanding the mode of action of VGSCs and in mapping their various allosteric binding sites. Furthermore, marine toxins appear to be an emerging source of therapeutic tools that can relieve pain or treat VGSC-related human channelopathies. Several studies documented the effect of marine toxins on VGSCs as well as their pharmaceutical applications, but none of them underlined the principal marine toxins and their effect on VGSCs. Therefore, this review aims to highlight the neurotoxins produced by marine animals such as pufferfish, shellfish, sea anemone, and cone snail that are active on VGSCs and discuss their pharmaceutical values.


Subject(s)
Biological Products , Marine Toxins/pharmacology , Voltage-Gated Sodium Channels/drug effects , Analgesics/therapeutic use , Animals , Humans , Marine Toxins/therapeutic use , Pain/drug therapy , Sea Anemones , Shellfish , Snails , Tetraodontiformes
11.
Mar Drugs ; 19(9)2021 Sep 15.
Article in English | MEDLINE | ID: mdl-34564182

ABSTRACT

Brevetoxins (BTXs) are marine biotoxins responsible for neurotoxic shellfish poisoning (NSP) after ingestion of contaminated shellfish. NSP is characterized by neurological, gastrointestinal and/or cardiovascular symptoms. The main known producer of BTXs is the dinoflagellate Karenia brevis, but other microalgae are also suspected to synthesize BTX-like compounds. BTXs are currently not regulated in France and in Europe. In November 2018, they have been detected for the first time in France in mussels from a lagoon in the Corsica Island (Mediterranean Sea), as part of the network for monitoring the emergence of marine biotoxins in shellfish. To prevent health risks associated with the consumption of shellfish contaminated with BTXs in France, a working group was set up by the French Agency for Food, Environmental and Occupational Health & Safety (Anses). One of the aims of this working group was to propose a guidance level for the presence of BTXs in shellfish. Toxicological data were too limited to derive an acute oral reference dose (ARfD). Based on human case reports, we identified two lowest-observed-adverse-effect levels (LOAELs). A guidance level of 180 µg BTX-3 eq./kg shellfish meat is proposed, considering a protective default portion size of 400 g shellfish meat.


Subject(s)
Dinoflagellida , Marine Toxins/analysis , Oxocins/analysis , Shellfish Poisoning/prevention & control , Shellfish , Animals , Environmental Monitoring , France , Humans , Mediterranean Sea
12.
Int J Mol Sci ; 22(10)2021 May 12.
Article in English | MEDLINE | ID: mdl-34065933

ABSTRACT

Neonicotinoid insecticides are nicotine-derived molecules which exert acute neurotoxic effects over the insect central nervous system by activating nicotinic acetylcholine receptors (nAChRs). However, these receptors are also present in the mammalian central and peripheral nervous system, where the effects of neonicotinoids are faintly known. In mammals, cholinergic synapses are crucial for the control of vascular tone, blood pressure and skeletal muscle contraction. We therefore hypothesized that neonicotinoids could affect cholinergic networks in mammals and sought to highlight functional consequences of acute intoxication in rats with sub-lethal concentrations of the highly used acetamiprid (ACE) and clothianidin (CLO). In this view, we characterized their electrophysiological effects on rat α3ß4 nAChRs, knowing that it is predominantly expressed in ganglia of the vegetative nervous system and the adrenal medulla, which initiates catecholamine secretion. Both molecules exhibited a weak agonist effect on α3ß4 receptors. Accordingly, their influence on epinephrine secretion from rat adrenal glands was also weak at 100 µM, but it was stronger at 500 µM. Challenging ACE or CLO together with nicotine (NIC) ended up with paradoxical effects on secretion. In addition, we measured the rat arterial blood pressure (ABP) in vivo by arterial catheterization. As expected, NIC induced a significant increase in ABP. ACE and CLO did not affect the ABP in the same conditions. However, simultaneous exposure of rats to both NIC and ACE/CLO promoted an increase of ABP and induced a biphasic response. Modeling the interaction of ACE or CLO on α3ß4 nAChR is consistent with a binding site located in the agonist pocket of the receptor. We present a transversal experimental approach of mammal intoxication with neonicotinoids at different scales, including in vitro, ex vivo, in vivo and in silico. It paves the way of the acute and chronic toxicity for this class of insecticides on mammalian organisms.


Subject(s)
Epinephrine/metabolism , Insecticides/toxicity , Neonicotinoids/toxicity , Nicotine/toxicity , Receptors, Nicotinic/metabolism , Adrenal Medulla/drug effects , Adrenal Medulla/metabolism , Animals , Arterial Pressure/drug effects , Disease Models, Animal , Drug Partial Agonism , Ganglia/drug effects , Ganglia/metabolism , Gene Expression Regulation/drug effects , Guanidines/toxicity , Male , Rats , Thiazoles/toxicity , Toxicity Tests, Subacute
13.
Molecules ; 26(8)2021 Apr 12.
Article in English | MEDLINE | ID: mdl-33921462

ABSTRACT

Cardiovascular diseases (CVDs) are considered as a major cause of death worldwide. Therefore, identifying and developing therapeutic strategies to treat and reduce the prevalence of CVDs is a major medical challenge. Several drugs used for the treatment of CVDs, such as captopril, emerged from natural products, namely snake venoms. These venoms are complex mixtures of bioactive molecules, which, among other physiological networks, target the cardiovascular system, leading to them being considered in the development and design of new drugs. In this review, we describe some snake venom molecules targeting the cardiovascular system such as phospholipase A2 (PLA2), natriuretic peptides (NPs), bradykinin-potentiating peptides (BPPs), cysteine-rich secretory proteins (CRISPs), disintegrins, fibrinolytic enzymes, and three-finger toxins (3FTXs). In addition, their molecular targets, and mechanisms of action-vasorelaxation, inhibition of platelet aggregation, cardioprotective activities-are discussed. The dissection of their biological effects at the molecular scale give insights for the development of future snake venom-derived drugs.


Subject(s)
Cardiovascular Diseases/drug therapy , Snake Venoms/chemistry , Snake Venoms/therapeutic use , Animals , Cardiovascular Agents/pharmacology , Cardiovascular Agents/therapeutic use , Cardiovascular System/drug effects , Cardiovascular System/pathology , Humans , Models, Biological , Snake Venoms/pharmacology
14.
Environ Res ; 193: 110590, 2021 02.
Article in English | MEDLINE | ID: mdl-33307089

ABSTRACT

Anatoxin-a (ATX-a) is a neurotoxic alkaloid, produced by several freshwater planktonic and benthic cyanobacteria (CB). Such CB have posed human and animal health issues for several years, as this toxin is able to cause neurologic symptoms in humans following food poisoning and death in wild and domestic animals. Different episodes of animal intoxication have incriminated ATX-a worldwide, as confirmed by the presence of ATX-a-producing CB in the consumed water or biofilm, or the observation of neurotoxic symptoms, which match experimental toxicity in vivo. Regarding toxicity parameters, toxicokinetics knowledge is currently incomplete and needs to be improved. The toxin can passively cross biological membranes and act rapidly on nicotinic receptors, its main molecular target. In vivo and in vitro acute effects of ATX-a have been studied and make possible to draw its mode of action, highlighting its deleterious effects on the nervous systems and its effectors, namely muscles, heart and vessels, and the respiratory apparatus. However, very little is known about its putative chronic toxicity. This review updates available data on ATX-a, from the ecodynamic of the toxin to its physiological and molecular targets.


Subject(s)
Bacterial Toxins , Cyanobacteria , Animals , Bacterial Toxins/toxicity , Cyanobacteria Toxins , Humans , Neurotoxins/toxicity , Tropanes/toxicity
15.
Toxicon ; 180: 1-10, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32246951

ABSTRACT

Pinnatoxins (PnTXs) are a group of emerging marine biotoxins produced by the benthic dinoflagellate Vulcanodinium rugosum, currently not regulated in Europe or in any other country in the world. In France, PnTXs were detected for the first time in 2011, in mussels from the Ingril lagoon (South of France, Mediterranean coast). Since then, analyses carried out in mussels from this lagoon have shown high concentrations of PnTXs for several months each year. PnTXs have also been detected, to a lesser extent, in mussels from other Mediterranean lagoons and on the Atlantic and Corsican coasts. In the French data, the main analog is PnTX G (low levels of PnTX A are also present in some samples). No cases of PnTXs poisoning in humans have been reported so far in France or anywhere else in the world. In mice, PnTXs induce acute neurotoxic effects, within a few minutes after oral administration. Clinical signs of toxicity include decreased mobility, paralysis of the hind legs, tremors, jumps and breathing difficulties leading to death by respiratory arrest at high doses. The French agency for food safety (ANSES) recently conducted a review of the state of knowledge related to PnTXs and V. rugosum. Based on (i) the clinical signs of toxicity in mice, (ii) the mode of action of PnTXs as nicotinic acetylcholine receptor competitive antagonists and (iii) knowledge on drugs and natural toxins with PnTX-related pharmacology, potential human symptoms have been extrapolated and proposed. In this work, a provisional acute benchmark value for PnTX G of 0.13 µg/kg bw per day has been derived from an oral acute toxicity study in mice. Based on this value and a large shellfish meat portion size of 400g, a concentration lower than 23 µg PnTX G/kg shellfish meat is not expected to result in adverse effects in humans. ANSES recommends taking into account PnTXs in the French official monitoring program for shellfish production and identified data gaps to refine health risk assessment.


Subject(s)
Dietary Exposure/statistics & numerical data , Environmental Monitoring , Food Safety , Marine Toxins/analysis , Shellfish/statistics & numerical data , Animals , Bivalvia , Dinoflagellida , France , Humans , Marine Toxins/metabolism , Mice , Risk Assessment , Seafood/statistics & numerical data , Shellfish Poisoning
16.
Mar Drugs ; 17(7)2019 Jul 20.
Article in English | MEDLINE | ID: mdl-31330850

ABSTRACT

Pinnatoxins (PnTXs) are emerging neurotoxins that were discovered about 30 years ago. They are solely produced by the marine dinoflagellate Vulcanodinium rugosum, and may be transferred into the food chain, as they have been found in various marine invertebrates, including bivalves. No human intoxication has been reported to date although acute toxicity was induced by PnTxs in rodents. LD50 values have been estimated for the different PnTXs through the oral route. At sublethal doses, all symptoms are reversible, and no neurological sequelae are visible. These symptoms are consistent with impairment of central and peripheral cholinergic network functions. In fact, PnTXs are high-affinity competitive antagonists of nicotinic acetylcholine receptors (nAChRs). Moreover, their lethal effects are consistent with the inhibition of muscle nAChRs, inducing respiratory distress and paralysis. Human intoxication by ingestion of PnTXs could result in various symptoms observed in episodes of poisoning with natural nAChR antagonists. This review updates the available data on PnTX toxicity with a focus on their mode of action on cholinergic networks and suggests the effects that could be extrapolated on human physiology.


Subject(s)
Dinoflagellida/chemistry , Marine Toxins/toxicity , Nicotinic Antagonists/toxicity , Paralysis/chemically induced , Poisoning/etiology , Acetylcholine/metabolism , Alkaloids/chemistry , Alkaloids/toxicity , Animals , Disease Models, Animal , Humans , Lethal Dose 50 , Marine Toxins/chemistry , Muscles/drug effects , Muscles/innervation , Muscles/metabolism , Nicotinic Antagonists/chemistry , Receptors, Nicotinic/metabolism , Spiro Compounds/chemistry , Spiro Compounds/toxicity , Synaptic Transmission/drug effects , Toxicity Tests, Acute
17.
Pharmacol Res ; 145: 104250, 2019 07.
Article in English | MEDLINE | ID: mdl-31059790

ABSTRACT

Etifoxine (EFX) is a non-benzodiazepine psychoactive drug which exhibits anxiolytic effects through a dual mechanism, by directly binding to GABAA receptors (GABAARs) and to the mitochondrial 18-kDa translocator protein, resulting in the potentiation of the GABAergic function. The ß subunit subtype plays a key role in the EFX-GABAAR interaction, however this does not explain the anxiolytic effects of this drug. Here, we combined behavioral and electrophysiological experiments to challenge the role of the GABAAR α subunit in the EFX mode of action. After single administrations of anxiolytic doses (25-50 mg/kg, intraperitoneal), EFX did not induce any neurological nor locomotor impairments, unlike the benzodiazepine bromazepam (0.5-1 mg/kg, intraperitoneal). We established the EFX pharmacological profile on heteropentameric GABAARs constructed with α1 to α6 subunit expressed in Xenopus oocyte. Unlike what is known for benzodiazepines, neither the γ nor δ subunits influenced EFX-mediated potentiation of GABA-evoked currents. EFX acted first as a partial agonist on α2ß3γ2S, α3ß3γ2S, α6ß3γ2S and α6ß3δ GABAARs, but not on α1ß3γ2S, α4ß3γ2S, α4ß3δ nor α5ß3γ2S GABAARs. Moreover, EFX exhibited much higher positive allosteric modulation towards α2ß3γ2S, α3ß3γ2S and α6ß3γ2S than for α1ß3γ2S, α4ß3γ2S and α5ß3γ2S GABAARs. At 20 µM, corresponding to brain concentration at anxiolytic doses, EFX increased GABA potency to the highest extent for α3ß3γ2S GABAARs. We built a docking model of EFX on α3ß3γ2S GABAARs, which is consistent with a binding site located between α and ß subunits in the extracellular domain. In conclusion, EFX preferentially potentiates α2ß3γ2S and α3ß3γ2S GABAARs, which might support its advantageous anxiolytic/sedative balance.


Subject(s)
Anti-Anxiety Agents/pharmacology , Oxazines/pharmacology , Protein Subunits/metabolism , Receptors, GABA-A/metabolism , Animals , Anti-Anxiety Agents/therapeutic use , Anxiety/drug therapy , Anxiety/metabolism , Anxiety/physiopathology , Female , Locomotion/drug effects , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Models, Molecular , Oocytes/physiology , Oxazines/therapeutic use , Protein Subunits/genetics , Psychomotor Performance/drug effects , Receptors, GABA-A/genetics , Xenopus laevis
18.
Toxins (Basel) ; 11(4)2019 03 30.
Article in English | MEDLINE | ID: mdl-30935025

ABSTRACT

Bee venom is a mixture of several components with proven therapeutic benefits, among which are anti-inflammatory, analgesic, and various cardiovascular conditions. In this work, we analyzed for the first time the proteomic content and biological properties of the crude venom from Apis mellifera syriaca, a honeybee from the Middle East region. Using high-performance liquid chromatography-tandem mass spectrometry, we evidence the venom contains phospholipase A2, hyaluronidase, mast cell-degranulating peptide, adolapin, apamin, and melittin. The latter was purified by solid phase extraction method (SPE) and tested in parallel with crude venom for biological activities. Precisely, crude venom-but not melittin-exhibited antibacterial activity against Staphylococcus aureus and Pseudomonas aeruginosa strains. Alongside, hemolytic activity was observed in human blood subjected to the venom at high doses. A. mellifera syriaca venom displayed antioxidant activities, and not surprisingly, PLA2 catalytic activity. Eventually, the venom proved to exert antiproliferative effects against MCF-7 and 3T3 cancer cells lines. This first report of a new bee venom opens new avenues for therapeutic uses of bee venoms.


Subject(s)
Bee Venoms , 3T3 Cells , Animals , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antioxidants/chemistry , Antioxidants/pharmacology , Bacteria/drug effects , Bee Venoms/chemistry , Bee Venoms/pharmacology , Bees , Biphenyl Compounds/chemistry , Cell Survival/drug effects , Erythrocytes/drug effects , Hemolysis/drug effects , Humans , MCF-7 Cells , Mice , Middle East , Phospholipases A2/chemistry , Picrates/chemistry
19.
Mar Drugs ; 16(2)2018 Feb 22.
Article in English | MEDLINE | ID: mdl-29470418

ABSTRACT

Tetrodotoxin (TTX), the mode of action of which has been known since the 1960s, is widely used in pharmacology as a specific inhibitor of voltage-gated sodium channels (Nav channels). This toxin has contributed to the characterization of the allosteric model of the Nav channel, and to discriminating TTX-sensitive and TTX-resistant subtypes. In addition to its role as a pharmacological tool, TTX is now considered a therapeutic molecule, and its development should lead to its use in certain pathologies involving Nav channels, particularly in the field of pain. Specifically, the blockade of Nav channels expressed in nociceptive fibres is one strategy for alleviating pain and its deleterious consequences on health. Recent work has identified, in addition to the Nav1.7, 1.8 and 1.9 channels, the Nav1.1 subtype on dorsal root ganglion (DRG) neurons as a crucial player in mechanical and non-thermal pain. The sensitivity of Nav1.1 to TTX could be exploited at the therapeutic level, especially in chronic pain conditions.


Subject(s)
NAV1.1 Voltage-Gated Sodium Channel/metabolism , Tetrodotoxin/pharmacology , Animals , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Humans , Nociceptors/drug effects , Nociceptors/metabolism , Pain/drug therapy , Pain/metabolism
20.
Toxins (Basel) ; 10(1)2017 12 22.
Article in English | MEDLINE | ID: mdl-29271898

ABSTRACT

The implication of the cyanotoxin ß-N-methylamino-l-alanine (BMAA) in long-lasting neurodegenerative disorders is still a matter of controversy. It has been alleged that chronic ingestion of BMAA through the food chain could be a causative agent of amyotrophic lateral sclerosis (ALS) and several related pathologies including Parkinson syndrome. Both in vitro and in vivo studies of the BMAA mode of action have focused on different molecular targets, demonstrating its toxicity to neuronal cells, especially motoneurons, and linking it to human neurodegenerative diseases. Historically, the hypothesis of BMAA-induced excitotoxicity following the stimulation of glutamate receptors has been established. However, in this paradigm, most studies have shown acute, rather than chronic effects of BMAA. More recently, the interaction of this toxin with neuromelanin, a pigment present in the nervous system, has opened a new research perspective. The issues raised by this toxin are related to its kinetics of action, and its possible incorporation into cellular proteins. It appears that BMAA neurotoxic activity involves different targets through several mechanisms known to favour the development of neurodegenerative processes.


Subject(s)
Amino Acids, Diamino/metabolism , Neurodegenerative Diseases/metabolism , Animals , Cyanobacteria Toxins , Humans , Melanins/metabolism , Receptors, Glutamate/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...