Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 565
Filter
1.
Mar Drugs ; 22(5)2024 May 13.
Article in English | MEDLINE | ID: mdl-38786608

ABSTRACT

We identified a new human voltage-gated potassium channel blocker, NnK-1, in the jellyfish Nemopilema nomurai based on its genomic information. The gene sequence encoding NnK-1 contains 5408 base pairs, with five introns and six exons. The coding sequence of the NnK-1 precursor is 894 nucleotides long and encodes 297 amino acids containing five presumptive ShK-like peptides. An electrophysiological assay demonstrated that the fifth peptide, NnK-1, which was chemically synthesized, is an effective blocker of hKv1.3, hKv1.4, and hKv1.5. Multiple-sequence alignment with cnidarian Shk-like peptides, which have Kv1.3-blocking activity, revealed that three residues (3Asp, 25Lys, and 34Thr) of NnK-1, together with six cysteine residues, were conserved. Therefore, we hypothesized that these three residues are crucial for the binding of the toxin to voltage-gated potassium channels. This notion was confirmed by an electrophysiological assay with a synthetic peptide (NnK-1 mu) where these three peptides were substituted with 3Glu, 25Arg, and 34Met. In conclusion, we successfully identified and characterized a new voltage-gated potassium channel blocker in jellyfish that interacts with three different voltage-gated potassium channels. A peptide that interacts with multiple voltage-gated potassium channels has many therapeutic applications in various physiological and pathophysiological contexts.


Subject(s)
Peptides , Potassium Channel Blockers , Potassium Channels, Voltage-Gated , Scyphozoa , Animals , Humans , Potassium Channel Blockers/pharmacology , Potassium Channel Blockers/chemistry , Potassium Channels, Voltage-Gated/antagonists & inhibitors , Peptides/pharmacology , Peptides/chemistry , Amino Acid Sequence , Cnidarian Venoms/pharmacology , Cnidarian Venoms/chemistry , Sequence Alignment
2.
Mar Drugs ; 22(5)2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38786596

ABSTRACT

The escalation of jellyfish stings has drawn attention to severe skin reactions, underscoring the necessity for novel treatments. This investigation assesses the potential of hydroxybenzoic acid derivatives, specifically protocatechuic acid (PCA) and gentisic acid (DHB), for alleviating Nemopilema nomurai Nematocyst Venom (NnNV)-induced injuries. By employing an in vivo mouse model, the study delves into the therapeutic efficacy of these compounds. Through a combination of ELISA and Western blot analyses, histological examinations, and molecular assays, the study scrutinizes the inflammatory response, assesses skin damage and repair mechanisms, and investigates the compounds' ability to counteract venom effects. Our findings indicate that PCA and DHB significantly mitigate inflammation by modulating critical cytokines and pathways, altering collagen ratios through topical application, and enhancing VEGF and bFGF levels. Furthermore, both compounds demonstrate potential in neutralizing NnNV toxicity by inhibiting metalloproteinases and phospholipase-A2, showcasing the viability of small-molecule compounds in managing toxin-induced injuries.


Subject(s)
Cnidarian Venoms , Hydroxybenzoates , Skin , Animals , Hydroxybenzoates/pharmacology , Mice , Cnidarian Venoms/pharmacology , Skin/drug effects , Skin/pathology , Skin/metabolism , Gentisates/pharmacology , Nematocyst/drug effects , Disease Models, Animal , Cytokines/metabolism
3.
Front Immunol ; 15: 1369849, 2024.
Article in English | MEDLINE | ID: mdl-38779681

ABSTRACT

Background: Stomolophus meleagris envenomation causes severe cutaneous symptoms known as jellyfish dermatitis. The potential molecule mechanisms and treatment efficiency of dermatitis remain elusive because of the complicated venom components. The biological activity and molecular regulation mechanism of Troxerutin (TRX) was firstly examined as a potential treatment for jellyfish dermatitis. Methods: We examined the inhibit effects of the TRX on tentacle extract (TE) obtained from S. meleagris in vivo and in vitro using the mice paw swelling models and corresponding assays for Enzyme-Linked Immunosorbent Assay (ELISA) Analysis, cell counting kit-8 assay, flow cytometry, respectively. The mechanism of TRX on HaCaT cells probed the altered activity of relevant signaling pathways by RNA sequencing and verified by RT-qPCR, Western blot to further confirm protective effects of TRX against the inflammation and oxidative damage caused by TE. Results: TE significantly induced the mice paw skin toxicity and accumulation of inflammatory cytokines and reactive oxygen species in vivo and vitro. Moreover, a robust increase in the phosphorylation of mitogen-activated protein kinase (MAPKs) and nuclear factor-kappa B (NF-κB) signaling pathways was observed. While, the acute cutaneous inflammation and oxidative stress induced by TE were significantly ameliorated by TRX treatment. Notablly, TRX suppressed the phosphorylation of MAPK and NF-κB by initiating the nuclear factor erythroid 2-related factor 2 signaling pathway, which result in decreasing inflammatory cytokine release. Conclusion: TRX inhibits the major signaling pathway responsible for inducing inflammatory and oxidative damage of jellyfish dermatitis, offering a novel therapy in clinical applications.


Subject(s)
Dermatitis , Hydroxyethylrutoside , NF-E2-Related Factor 2 , Oxidative Stress , Scyphozoa , Signal Transduction , Animals , Oxidative Stress/drug effects , Mice , NF-E2-Related Factor 2/metabolism , Signal Transduction/drug effects , Dermatitis/drug therapy , Dermatitis/metabolism , Dermatitis/etiology , Humans , Hydroxyethylrutoside/analogs & derivatives , Hydroxyethylrutoside/pharmacology , Hydroxyethylrutoside/therapeutic use , Cnidarian Venoms/pharmacology , Heme Oxygenase-1/metabolism , Disease Models, Animal , Inflammation/drug therapy , Inflammation/metabolism , Male , Cytokines/metabolism , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , HaCaT Cells , Reactive Oxygen Species/metabolism , Membrane Proteins
4.
Pharmacol Rep ; 76(3): 585-599, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38619735

ABSTRACT

BACKGROUND: Amiodarone (AMIO) is an antiarrhythmic drug with the pKa in the physiological range. Here, we explored how mild extracellular pH (pHe) changes shape the interaction of AMIO with atrial tissue and impact its pharmacological properties in the classical model of sea anemone sodium channel neurotoxin type 2 (ATX) induced late sodium current (INa-Late) and arrhythmias. METHOD: Isolated atrial cardiomyocytes from male Wistar rats and human embryonic kidney cells expressing SCN5A Na+ channels were used for patch-clamp experiments. Isolated right atria (RA) and left atria (LA) tissue were used for bath organ experiments. RESULTS: A more acidophilic pHe caused negative inotropic effects on isolated RA and LA atrial tissue, without modification of the pharmacological properties of AMIO. A pHe of 7.0 changed the sodium current (INa) related components of the action potential (AP), which was enhanced in the presence of AMIO. ATXinduced arrhythmias in isolated RA and LA. Also, ATX prolonged the AP duration and enhanced repolarization dispersion in isolated cardiomyocytes in both pHe 7.4 and pHe 7.0. Pre-incubation of the isolated RA and LA and isolated atrial cardiomyocytes with AMIO prevented arrhythmias induced by ATX only at a pHe of 7.0. Moreover, AMIO was able to block INa-Late induced by ATX only at a pHe of 7.0. CONCLUSION: The pharmacological properties of AMIO concerning healthy rat atrial tissue are not dependent on pHe. However, the prevention of arrhythmias induced by INa-Late is pHe-dependent. The development of drugs analogous to AMIO with charge stabilization may help to create more effective drugs to treat arrhythmias related to the INa-Late.


Subject(s)
Action Potentials , Amiodarone , Anti-Arrhythmia Agents , Arrhythmias, Cardiac , Heart Atria , Myocytes, Cardiac , Rats, Wistar , Animals , Amiodarone/pharmacology , Anti-Arrhythmia Agents/pharmacology , Male , Humans , Rats , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Action Potentials/drug effects , Heart Atria/drug effects , Heart Atria/metabolism , Hydrogen-Ion Concentration , Arrhythmias, Cardiac/drug therapy , Arrhythmias, Cardiac/metabolism , Arrhythmias, Cardiac/chemically induced , NAV1.5 Voltage-Gated Sodium Channel/metabolism , HEK293 Cells , Sodium/metabolism , Patch-Clamp Techniques , Cnidarian Venoms/pharmacology
5.
Microb Pathog ; 186: 106486, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38056601

ABSTRACT

In this study, we investigated the potential in vitro anti-HSV-1 activities of the Cassiopea andromeda jellyfish tentacle extract (TE) and its fractions, as well as computational work on the thymidine kinase (TK) inhibitory activity of the identified secondary metabolites. The LD50, secondary metabolite identification, preparative and analytical chromatography, and in silico TK assessment were performed using the Spearman-Karber, GC-MS, silica gel column chromatography, RP-HPLC, LC-MS, and docking methods, respectively. The antiviral activity of TE and the two purified compounds Ca2 and Ca7 against HSV-1 in Vero cells was evaluated by MTT and RT-PCR assays. The LD50 (IV, mouse) values of TE, Ca2, and Ca7 were 104.0 ± 4, 5120 ± 14, and 197.0 ± 7 (µg/kg), respectively. They exhibited extremely effective antiviral activity against HSV-1. The CC50 and MNTD of TE, Ca2, and Ca7 were (125, 62.5), (25, 12.5), and (50, 3.125) µg/ml, respectively. GC-MS analysis of the tentacle extract revealed seven structurally distinct chemical compositions. Four of the seven compounds had a steroid structure. According to the docking results, all compounds showed binding affinity to the active sites of both thymidine kinase chains. Among them, the steroid compound Pregn-5-ene-3,11-dione, 17,20:20,21 bis [methylenebis(oxy)]-, cyclic 3-(1,2-ethane diyl acetal) (Ca2) exhibited the highest affinity for both enzyme chains, surpassing that of standard acyclovir. In silico data confirmed the experimental results. We conclude that the oxosteroid Ca2 may act as a potent agent against HSV-1.


Subject(s)
Cnidarian Venoms , Herpesvirus 1, Human , Chlorocebus aethiops , Animals , Mice , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , Vero Cells , Thymidine Kinase/genetics , Thymidine Kinase/chemistry , Cnidarian Venoms/pharmacology , Steroids/pharmacology
6.
J Proteomics ; 292: 105048, 2024 02 10.
Article in English | MEDLINE | ID: mdl-37981009

ABSTRACT

Toxin metalloproteinases are the primary components responsible for various toxicities in jellyfish venom, and there is still no effective specific therapy for jellyfish stings. The comprehension of the pathogenic mechanisms underlying toxin metalloproteinases necessitates further refinement. In this study, we conducted a differential analysis of a dermatitis mouse model induced by jellyfish Nemopilema nomurai venom (NnNV) samples with varying levels of metalloproteinase activity. Through skin tissue proteomics and serum metabolomics, the predominant influence of toxin metalloproteinase activity on inflammatory response was revealed, and the signal pathway involved in its regulation was identified. In skin tissues, many membrane proteins were significantly down-regulated, which might cause tissue damage. The expression of pro-inflammatory factors was mainly regulated by PI3K-Akt signaling pathway. In serum, many fatty acid metabolites were significantly down-regulated, which might be the anti-inflammation feedback regulated by NF-κB p65 signaling pathway. These results reveal the dermatitis mechanism of toxin metalloproteinases and provide new therapeutic targets for further studies. SIGNIFICANCE: Omics is an important method to analyze the pathological mechanism and discover the key markers, which can reveal the pathological characteristics of jellyfish stings. Our research first analyzed the impact of toxin metalloproteinases on jellyfish sting dermatitis by skin proteomics and serum metabolomics. The present results suggest that inhibition of toxin metalloproteinases may be an effective treatment strategy, and provide new references for further jellyfish sting studies.


Subject(s)
Cnidarian Venoms , Dermatitis , Scyphozoa , Toxins, Biological , Animals , Mice , Phosphatidylinositol 3-Kinases , Cnidarian Venoms/pharmacology , Metalloproteases , Anti-Inflammatory Agents
7.
Toxicon ; 233: 107266, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37625553

ABSTRACT

Jellyfish venoms have long been recognized as a potentially rich source of natural bioactive compounds with pharmacological potential for the creation of innovative drugs. Our previous study demonstrated that Nemopilema nomurai jellyfish venom (NnV) has a chymotrypsin-like serine protease with fibrinolytic activity in vitro. Therefore, the present study aims to investigate the potential effect of NnV on cell migration, proliferation, and differentiation of vascular smooth muscle cells (VSMC; A7r5 cells) involved in the probable mechanism pathways. We also determined its anti-thrombotic effect through κ-carrageenan-induced Sprague-Dawley (SD) rat tail thrombus model. NnV inhibits on Platelet-derived growth factor (PDGF)-BB-stimulated A7r5 cells migration and proliferation by decreasing matrix metalloproteinase 2 (MMP-2) level and phosphorylation of ERK and Akt in a dose-dependent manner, but not p38. Furthermore, NnV regulates the phenotype transition of differentiation in PDGF-BB-stimulated A7r5 cells via ɑ-SMA and calponin in a dose-dependent manner. In an in vivo study, NnV treatment demonstrated clear anti-thrombotic activity in a dose-dependent manner, which was associated with decreased thrombus formation and length in κ-carrageenan-induced SD rat tail. These findings suggested that NnV has a novel fibrinolytic enzyme that can be used to prevent and/or treat thrombosis-related cardiovascular disorders.


Subject(s)
Cnidarian Venoms , Thrombosis , Rats , Animals , Rats, Sprague-Dawley , Becaplermin/pharmacology , Cnidarian Venoms/pharmacology , Carrageenan , Matrix Metalloproteinase 2 , Muscle, Smooth, Vascular , Tail , Phenotype
8.
J Chem Inf Model ; 63(10): 3043-3053, 2023 05 22.
Article in English | MEDLINE | ID: mdl-37143234

ABSTRACT

Peptide toxins that adopt the ShK fold can inhibit the voltage-gated potassium channel KV1.3 with IC50 values in the pM range and are therefore potential leads for drugs targeting autoimmune and neuroinflammatory diseases. Nuclear magnetic resonance (NMR) relaxation measurements and pressure-dependent NMR have shown that, despite being cross-linked by disulfide bonds, ShK itself is flexible in solution. This flexibility affects the local structure around the pharmacophore for the KV1.3 channel blockade and, in particular, the relative orientation of the key Lys and Tyr side chains (Lys22 and Tyr23 in ShK) and has implications for the design of KV1.3 inhibitors. In this study, we have performed molecular dynamics (MD) simulations on ShK and a close homologue, HmK, to probe the conformational space occupied by the Lys and Tyr residues, and docked the different conformations with a recently determined cryo-EM structure of the KV1.3 channel. Although ShK and HmK have 60% sequence identity, their dynamic behaviors are quite different, with ShK sampling a broad range of conformations over the course of a 5 µs MD simulation, while HmK is relatively rigid. We also investigated the importance of conformational dynamics, in particular the distance between the side chains of the key dyad Lys22 and Tyr23, for binding to KV1.3. Although these peptides have quite different dynamics, the dyad in both adopts a similar configuration upon binding, revealing a conformational selection upon binding to KV1.3 in the case of ShK. Both peptides bind to KV1.3 with Lys22 occupying the pore of the channel. Intriguingly, the more flexible peptide, ShK, binds with significantly higher affinity than HmK.


Subject(s)
Cnidarian Venoms , Sea Anemones , Animals , Kv1.3 Potassium Channel/chemistry , Kv1.3 Potassium Channel/metabolism , Cnidarian Venoms/chemistry , Cnidarian Venoms/metabolism , Cnidarian Venoms/pharmacology , Sea Anemones/chemistry , Sea Anemones/metabolism , Peptides/chemistry , Molecular Conformation , Potassium Channel Blockers/pharmacology , Potassium Channel Blockers/chemistry , Kv1.2 Potassium Channel/metabolism
9.
Mar Drugs ; 21(3)2023 Mar 07.
Article in English | MEDLINE | ID: mdl-36976217

ABSTRACT

Nowadays, major attention is being paid to curing different types of cancers and is focused on natural resources, including oceans and marine environments. Jellyfish are marine animals with the ability to utilize their venom in order to both feed and defend. Prior studies have displayed the anticancer capabilities of various jellyfish. Hence, we examined the anticancer features of the venom of Cassiopea andromeda and Catostylus mosaicus in an in vitro situation against the human pulmonary adenocarcinoma (A549) cancer cell line. The MTT assay demonstrated that both mentioned venoms have anti-tumoral ability in a dose-dependent manner. Western blot analysis proved that both venoms can increase some pro-apoptotic factors and reduce some anti-apoptotic molecules that lead to the inducing of apoptosis in A549 cells. GC/MS analysis demonstrated some compounds with biological effects, including anti-inflammatory, antioxidant and anti-cancer activities. Molecular docking and molecular dynamic showed the best position of each biologically active component on the different death receptors, which are involved in the process of apoptosis in A549 cells. Ultimately, this study has proven that both venoms of C. andromeda and C. mosaicus have the capability to suppress A549 cells in an in vitro condition and they might be utilized in order to design and develop brand new anticancer agents in the near future.


Subject(s)
Adenocarcinoma , Cnidaria , Cnidarian Venoms , Lung Neoplasms , Scyphozoa , Animals , Humans , Cnidarian Venoms/pharmacology , Cnidarian Venoms/chemistry , A549 Cells , Molecular Docking Simulation , Adenocarcinoma/drug therapy , Apoptosis , Lung Neoplasms/drug therapy
10.
Int J Mol Sci ; 24(5)2023 Feb 22.
Article in English | MEDLINE | ID: mdl-36901790

ABSTRACT

Infections caused by multi-drug-resistant (MDR) bacteria are a global threat to human health. As venoms are the source of biochemically diverse bioactive proteins and peptides, we investigated the antimicrobial activity and murine skin infection model-based wound healing efficacy of a 13 kDa protein. The active component PaTx-II was isolated from the venom of Pseudechis australis (Australian King Brown or Mulga Snake). PaTx-II inhibited the growth of Gram-positive bacteria in vitro, with moderate potency (MICs of 25 µM) observed against S. aureus, E. aerogenes, and P. vulgaris. The antibiotic activity of PaTx-II was associated with the disruption of membrane integrity, pore formation, and lysis of bacterial cells, as evidenced by scanning and transmission microscopy. However, these effects were not observed with mammalian cells, and PaTx-II exhibited minimal cytotoxicity (CC50 > 1000 µM) toward skin/lung cells. Antimicrobial efficacy was then determined using a murine model of S. aureus skin infection. Topical application of PaTx-II (0.5 mg/kg) cleared S. aureus with concomitant increased vascularization and re-epithelialization, promoting wound healing. As small proteins and peptides can possess immunomodulatory effects to enhance microbial clearance, cytokines and collagen from the wound tissue samples were analyzed by immunoblots and immunoassays. The amounts of type I collagen in PaTx-II-treated sites were elevated compared to the vehicle controls, suggesting a potential role for collagen in facilitating the maturation of the dermal matrix during wound healing. Levels of the proinflammatory cytokines interleukin-1ß (IL-1ß), interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α), cyclooxygenase-2 (COX-2) and interleukin-10 (IL-10), factors known to promote neovascularization, were substantially reduced by PaTx-II treatment. Further studies that characterize the contributions towards efficacy imparted by in vitro antimicrobial and immunomodulatory activity with PaTx-II are warranted.


Subject(s)
Anti-Infective Agents , Cnidarian Venoms , Colubridae , Humans , Animals , Mice , Staphylococcus aureus , Australia , Wound Healing , Anti-Infective Agents/pharmacology , Cnidarian Venoms/pharmacology , Collagen/pharmacology , Peptides/pharmacology , Cytokines/pharmacology , Mammals
11.
Mar Drugs ; 20(9)2022 Sep 08.
Article in English | MEDLINE | ID: mdl-36135760

ABSTRACT

Pelagia noctiluca stings are common in Mediterranean coastal areas and, although the venom is non-lethal, they are painful. Due to its high toxicity and abundance, P. noctiluca is considered a target species for the focus of research on active ingredients to reduce the symptoms of its sting. To determine the effect of 31 substances and formulations on nematocyst discharge, we performed three tests: (1) screening of per se discharge activator solutions, (2) inhibitory test with nematocyst chemical stimulation (5% acetic acid) and (3) inhibitory test quantifying the hemolytic area. Ammonia, barium chloride, bleach, scented ammonia, carbonated cola, lemon juice, sodium chloride and papain triggered nematocyst discharge. All of them were ruled out as potential inhibitors. Butylene glycol showed a reduction in nematocyst discharge, while the formulations of 10% lidocaine in ethanol, 1.5% hydroxyacetophenone in distilled water + butylene glycol, and 3% Symsitive® in butylene glycol inhibited nematocyst discharge. These last results were subsequently correlated with a significant decrease in hemolytic area in the venom assays versus seawater, a neutral solution. The presented data represent a first step in research to develop preventive products for jellyfish stings while at the same time attempting to clarify some uncertainties about the role of various topical solutions in P. noctiluca first-aid protocols.


Subject(s)
Bites and Stings , Cnidaria , Cnidarian Venoms , Scyphozoa , Ammonia/analysis , Ammonia/pharmacology , Animals , Bites and Stings/prevention & control , Butylene Glycols/analysis , Butylene Glycols/pharmacology , Cnidarian Venoms/analysis , Cnidarian Venoms/pharmacology , Ethanol/pharmacology , Hemolysis , Lidocaine/pharmacology , Nematocyst/chemistry , Papain/pharmacology , Scyphozoa/chemistry , Sodium Chloride/pharmacology , Water
12.
Chem Biol Interact ; 365: 110113, 2022 Sep 25.
Article in English | MEDLINE | ID: mdl-35987279

ABSTRACT

The major jellyfish stings that occur in China are caused by scyphozoan Nemopilema nomurai, whose venom exhibits significant metalloproteinase activity that contributes to the toxic effects of jellyfish envenomation. Researching effective inhibitors suppressing the metalloproteinase activity of jellyfish venom represents a new attempt to cure jellyfish envenomations. In the present study, secondary metabolites produced by the jellyfish-associated fungus Aspergillus versicolor SmT07 were isolated and evaluated for their anti-proteolytic activities. Two xanthones, sterigmatocystin (JC-01) and oxisterigmatocystin C (JC-06), and four alkaloids, cottoquinazoline A (JC-02), phenazine-1-carboxylic acid (JC-03), viridicatin (JC-04) and viridicatol (JC-05), were isolated and identified. Only phenazine-1-carboxylic acid (PCA) showed significant anti-proteolytic activity of jellyfish venom assayed on azocasein, and the IC50 value was 2.16 mM. PCA also significantly inhibited fibrinogenolytic activity, protecting the Bß chain of fibrinogen from degradation when preincubated with jellyfish venom at a ratio of >1:0.6 (PCA:venom, w/w). Molecular docking with several well-characterized snake venom metalloproteinases suggested the venom metalloproteinases inhibitory property of PCA by forming complex interactions with the active site via hydrogen bonds, π-π stacking and salt bridges, which was distinct from the binding mode of batimastat. The present study represents the first study identifying natural jellyfish venom metalloproteinase inhibitors from marine natural products, which may provide an alternative to develop therapeutic agents for treating jellyfish envenomations.


Subject(s)
Cnidarian Venoms , Scyphozoa , Animals , Aspergillus/metabolism , Cnidarian Venoms/chemistry , Cnidarian Venoms/pharmacology , Metalloproteases/metabolism , Molecular Docking Simulation , Scyphozoa/metabolism
13.
Sci Rep ; 12(1): 5352, 2022 03 30.
Article in English | MEDLINE | ID: mdl-35354863

ABSTRACT

Toxin production in nematocysts by Cnidaria phylum represents an important source of bioactive compounds. Using electrophysiology and, heterologous expression of mammalian ion channels in the Xenopus oocyte membrane, we identified two main effects produced by the sea anemone Bartholomea annulata venom. Nematocysts isolation and controlled discharge of their content, revealed that venom had potent effects on both voltage-dependent Na+ (Nav) channels and GABA type A channel receptors (GABAAR), two essential proteins in central nervous system signaling. Unlike many others sea anemone toxins, which slow the inactivation rate of Nav channels, B. annulata venom potently inhibited the neuronal action potential and the Na+ currents generated by distinct Nav channels opening, including human TTX-sensitive (hNav1.6) and TTX-insensitive Nav channels (hNav1.5). A second effect of B. annulata venom was an agonistic action on GABAAR that activated distinct receptors conformed by either α1ß2γ2, α3ß2γ1 or, ρ1 homomeric receptors. Since GABA was detected in venom samples by ELISA assay at low nanomolar range, it was excluded that GABA from nematocysts directly activated the GABAARs. This revealed that substances in B. annulata nematocysts generated at least two potent and novel effects on mammalian ion channels that are crucial for nervous system signaling.


Subject(s)
Cnidarian Venoms , Sea Anemones , Animals , Cnidarian Venoms/pharmacology , Mammals , Receptors, GABA-A , Sea Anemones/physiology , gamma-Aminobutyric Acid
14.
Molecules ; 26(17)2021 Aug 26.
Article in English | MEDLINE | ID: mdl-34500621

ABSTRACT

Jellyfish venom is a rich source of bioactive proteins and peptides with various biological activities including antioxidant, antimicrobial and antitumor effects. However, the anti-proliferative activity of the crude extract of Rhopilema nomadica jellyfish venom has not been examined yet. The present study aimed at the investigation of the in vitro effect of R. nomadica venom on liver cancer cells (HepG2), breast cancer cells (MDA-MB231), human normal fibroblast (HFB4), and human normal lung cells (WI-38) proliferation by using MTT assay. The apoptotic cell death in HepG2 cells was investigated using Annexin V-FITC/PI double staining-based flow cytometry analysis, western blot analysis, and DNA fragmentation assays. R. nomadica venom displayed significant dose-dependent cytotoxicity on HepG2 cells after 48 h of treatment with IC50 value of 50 µg/mL and higher toxicity (3:5-fold change) against MDA-MB231, HFB4, and WI-38 cells. R. nomadica venom showed a prominent increase of apoptosis as revealed by cell cycle arrest at G2/M phase, upregulation of p53, BAX, and caspase-3 proteins, and the down-regulation of anti-apoptotic Bcl-2 protein and DNA fragmentation. These findings suggest that R. nomadica venom induces apoptosis in hepatocellular carcinoma cells. To the best of the authors' knowledge, this is the first scientific evidence demonstrating the induction of apoptosis and cell cycle arrest of R. nomadica jellyfish venom.


Subject(s)
Apoptosis/drug effects , Carcinoma, Hepatocellular/drug therapy , Cell Cycle Checkpoints/drug effects , Cell Death/drug effects , Cnidarian Venoms/pharmacology , Liver Neoplasms/drug therapy , Scyphozoa/metabolism , Animals , Apoptosis Regulatory Proteins/metabolism , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Down-Regulation/drug effects , Hep G2 Cells , Humans , Liver Neoplasms/metabolism
15.
Insect Biochem Mol Biol ; 137: 103625, 2021 10.
Article in English | MEDLINE | ID: mdl-34358664

ABSTRACT

Scorpion α-toxins bind at the pharmacologically-defined site-3 on the sodium channel and inhibit channel inactivation by preventing the outward movement of the voltage sensor in domain IV (IVS4), whereas scorpion ß-toxins bind at site-4 on the sodium channel and enhance channel activation by trapping the voltage sensor of domain II (IIS4) in its outward position. However, limited information is available on the role of the voltage-sensing modules (VSM, comprising S1-S4) of domains I and III in toxin actions. We have previously shown that charge reversing substitutions of the innermost positively-charged residues in IIIS4 (R4E, R5E) increase the activity of an insect-selective site-4 scorpion toxin, Lqh-dprIT3-c, on BgNav1-1a, a cockroach sodium channel. Here we show that substitutions R4E and R5E in IIIS4 also increase the activity of two site-3 toxins, LqhαIT from Leiurusquinquestriatus hebraeus and insect-selective Av3 from Anemonia viridis. Furthermore, charge reversal of either of two conserved negatively-charged residues, D1K and E2K, in IIIS2 also increase the action of the site-3 and site-4 toxins. Homology modeling suggests that S2-D1 and S2-E2 interact with S4-R4 and S4-R5 in the VSM of domain III (III-VSM), respectively, in the activated state of the channel. However, charge swapping between S2-D1 and S4-R4 had no compensatory effects on gating or toxin actions, suggesting that charged residue interactions are complex. Collectively, our results highlight the involvement of III-VSM in the actions of both site 3 and site 4 toxins, suggesting that charge reversing substitutions in III-VSM allosterically facilitate IIS4 or IVS4 voltage sensor trapping by these toxins.


Subject(s)
Cnidarian Venoms/pharmacology , Drosophila melanogaster/genetics , Insect Proteins/genetics , Scorpion Venoms/pharmacology , Sodium Channels/genetics , Animals , Drosophila melanogaster/drug effects , Drosophila melanogaster/metabolism , Insect Proteins/metabolism , Oocytes/drug effects , Oocytes/metabolism , Sodium Channels/metabolism
16.
Sci Rep ; 11(1): 14794, 2021 07 20.
Article in English | MEDLINE | ID: mdl-34285326

ABSTRACT

Force enhancement is one kind of myogenic spontaneous fasciculation in lengthening preload striated muscles. In cardiac muscle, the role of this biomechanical event is not well established. The physiological passive property is an essential part for maintaining normal diastole in the heart. In excessive preload heart, force enhancement relative erratic passive properties may cause muscle decompensating, implicate in the development of diastolic dysfunction. In this study, the force enhancement occurrence in mouse cardiac papillary muscle was evaluated by a microstepping stretch method. The intracellular Ca2+ redistribution during occurrence of force enhancement was monitored in real-time by a Flou-3 (2 mM) indicator. The force enhancement amplitude, the enhancement of the prolongation time, and the tension-time integral were analyzed by myography. The results indicated that the force enhancement occurred immediately after active stretching and was rapidly enhanced during sustained static stretch. The presence of the force and the increase in the amplitude synchronized with the acquisition and immediate transfer of Ca2+ to adjacent fibres. In highly preloaded fibres, the enhancement exceeded the maximum passive tension (from 4.49 ± 0.43 N/mm2 to 6.20 ± 0.51 N/mm2). The occurrence of force enhancement were unstable in each static stretch. The increased enhancement amplitude combined with the reduced prolongation time to induce a reduction in the tension-time integral. We concluded that intracellular Ca2+-synchronized force enhancement is one kind of interruption event in excessive preload cardiac muscle. During the cardiac muscle in its passive relaxation period, the occurrence of this interruption affected the rhythmic stability of the cardiac relaxation cycle.


Subject(s)
Cnidarian Venoms/pharmacology , Fasciculation/pathology , Papillary Muscles/pathology , Animals , Biomechanical Phenomena , Calcium/metabolism , Fasciculation/metabolism , Fasciculation/physiopathology , Male , Mice , Myocardial Contraction , Papillary Muscles/drug effects , Papillary Muscles/metabolism , Papillary Muscles/physiopathology
17.
Toxins (Basel) ; 13(6)2021 06 10.
Article in English | MEDLINE | ID: mdl-34200597

ABSTRACT

Jellyfish are rich in resources and widely distributed along coastal areas. As a potential approach to respond to jellyfish blooms, the use of jellyfish-derived products is increasing. The citrus spider mite (Panonychus citri) is one of the key citrus pests, negatively impacting the quality and quantity of oranges. Due to the resistance and residue of chemical acaricides, it is important to seek natural substitutes that are environmentally friendly. The field efficacy of the venom from the jellyfish Nemopilema nomurai against P. citri was assayed in a citrus garden. The frozen N. nomurai tentacles were sonicated in different buffers to isolate the venom. The venom isolated by PBS buffer (10 mM, pH 6.0) had the strongest acaricidal activity of the four samples, and the corrected field efficacy 7 days after treatment was up to 95.21%. This study demonstrated that jellyfish has potential use in agriculture.


Subject(s)
Acaricides/pharmacology , Biological Control Agents/pharmacology , Citrus/parasitology , Cnidarian Venoms/pharmacology , Scyphozoa , Tetranychidae/drug effects , Agriculture/methods , Animals , Citrus/drug effects , Tetranychidae/physiology
18.
Sci Rep ; 11(1): 12014, 2021 06 08.
Article in English | MEDLINE | ID: mdl-34103608

ABSTRACT

Late sodium current (late INa) inhibition has been proposed to suppress the incidence of arrhythmias generated by pathological states or induced by drugs. However, the role of late INa in the human heart is still poorly understood. We therefore investigated the role of this conductance in arrhythmias using adult primary cardiomyocytes and tissues from donor hearts. Potentiation of late INa with ATX-II (anemonia sulcata toxin II) and E-4031 (selective blocker of the hERG channel) slowed the kinetics of action potential repolarization, impaired Ca2+ homeostasis, increased contractility, and increased the manifestation of arrhythmia markers. These effects could be reversed by late INa inhibitors, ranolazine and GS-967. We also report that atrial tissues from donor hearts affected by atrial fibrillation exhibit arrhythmia markers in the absence of drug treatment and inhibition of late INa with GS-967 leads to a significant reduction in arrhythmic behaviour. These findings reveal a critical role for the late INa in cardiac arrhythmias and suggest that inhibition of this conductance could provide an effective therapeutic strategy. Finally, this study highlights the utility of human ex-vivo heart models for advancing cardiac translational sciences.


Subject(s)
Atrial Fibrillation/metabolism , ERG1 Potassium Channel/metabolism , Membrane Potentials , Models, Cardiovascular , Myocytes, Cardiac/metabolism , Adult , Calcium/metabolism , Cnidarian Venoms/pharmacology , ERG1 Potassium Channel/antagonists & inhibitors , Heart Atria/metabolism , Humans , Myocytes, Cardiac/pathology , Piperidines/pharmacology , Pyridines/pharmacology , Ranolazine/pharmacology , Sodium , Triazoles/pharmacology
19.
Mar Drugs ; 19(1)2021 Jan 17.
Article in English | MEDLINE | ID: mdl-33477357

ABSTRACT

Arthritis is a widespread inflammatory disease associated with progressive articular surface degradation, ongoing pain, and hyperalgesia causing the development of functional limitations and disability. TRPV1 channel is one of the high-potential targets for the treatment of inflammatory diseases. Polypeptide APHC3 from sea anemone Heteractis crispa is a mode-selective TRPV1 antagonist that causes mild hypothermia and shows significant anti-inflammatory and analgesic activity in different models of pain. We evaluated the anti-inflammatory properties of APHC3 in models of monosodium iodoacetate (MIA)-induced osteoarthritis and complete Freund's adjuvant (CFA)-induced rheumatoid monoarthritis in comparison with commonly used non-steroidal anti-inflammatory drugs (NSAIDs) such as diclofenac, ibuprofen, and meloxicam. Subcutaneous administration of APHC3 (0.1 mg/kg) significantly reversed joint swelling, disability, grip strength impairment, and thermal and mechanical hypersensitivity. The effect of APHC3 was equal to or better than that of reference NSAIDs. Protracted treatment with APHC3 decreased IL-1b concentration in synovial fluid, reduced inflammatory changes in joints, and prevented the progression of cartilage degradation. Therefore, polypeptide APHC3 has the potential to be an analgesic and anti-inflammatory substance for the alleviation of arthritis symptoms.


Subject(s)
Analgesics/pharmacology , Anti-Inflammatory Agents/pharmacology , Arthritis, Experimental/drug therapy , Cnidarian Venoms/pharmacology , Intercellular Signaling Peptides and Proteins/pharmacology , Analgesics/isolation & purification , Animals , Anti-Inflammatory Agents/isolation & purification , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Arthritis, Experimental/physiopathology , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/physiopathology , Cnidarian Venoms/isolation & purification , Disease Models, Animal , Disease Progression , Intercellular Signaling Peptides and Proteins/isolation & purification , Male , Osteoarthritis/drug therapy , Osteoarthritis/physiopathology , Pain/drug therapy , Pain/physiopathology , Rats , Rats, Sprague-Dawley , TRPV Cation Channels/antagonists & inhibitors
20.
Basic Clin Pharmacol Toxicol ; 128(4): 615-620, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33124101

ABSTRACT

Long QT syndrome type 3 (LQT-3) is a disease related to abnormal cardiac sodium channel function (Nav 1.5), usually due to augmented late sodium current (INaL ), and may lead to ventricular fibrillation. Amiodarone is approved for ventricular fibrillation. Thus, we investigated whether pacing frequency impacts the ability of amiodarone to reverse the arrhythmic phenotype observed in LQT-3. Anemone neurotoxin 2 (ATX-II, here named only ATX) was used to enhance INaL in mice left ventricular myocytes (LVM). A video detector system monitored sarcomere shortening. At 1 Hz, amiodarone attenuated sarcomere shortening only at 10 µmol/L; at 3 and 5 Hz, 0.1 and 1 µmol/L amiodarone also reduced sarcomere shortening. However, no effect of amiodarone was observed on time to 50% of sarcomere contraction and relaxation. In LVM exposed to ATX (10 nmol/L), an arrhythmic phenotype was observed, and it was more severe when cells were paced at 1 Hz. Amiodarone failed to reverse the ATX induced phenotype at different pacing frequencies. Thus, our results suggest that amiodarone's ability to reverse arrhythmias induced by augmentation of INaL is limited. These findings suggest further experimentation will be required to clarify whether a clinical effect can be ascribed to an effect of amiodarone on other ion channels in LQT-3.


Subject(s)
Amiodarone/pharmacology , Cardiac Conduction System Disease/drug therapy , Long QT Syndrome/drug therapy , Myocytes, Cardiac/drug effects , Voltage-Gated Sodium Channel Blockers/pharmacology , Amiodarone/therapeutic use , Animals , Cardiac Conduction System Disease/chemically induced , Cardiac Conduction System Disease/physiopathology , Cells, Cultured , Cnidarian Venoms/pharmacology , Disease Models, Animal , Humans , Long QT Syndrome/chemically induced , Long QT Syndrome/physiopathology , Male , Mice , Myocytes, Cardiac/physiology , NAV1.5 Voltage-Gated Sodium Channel/metabolism , Primary Cell Culture , Voltage-Gated Sodium Channel Blockers/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...