Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 393
Filtrar
1.
Peptides ; 153: 170796, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35367253

RESUMO

The N-methyl-D-aspartate (NMDA) receptors are heteromeric cation channels involved in memory, learning, and synaptic plasticity. The dysfunction associated with NMDA receptors results in neurodegenerative conditions. The conantokins comprise a family of Conus venom peptides that induce sleep upon intracranial injection into young mice and are known to be NMDA receptor antagonists. This work comprehensibly documents the conantokins that have been characterized to date, focusing on the biochemistry, solution structures in the presence or absence of divalent cations, functions as selective NMDA receptor antagonists, and structure-activity relationships. Furthermore, the applications of conantokins as potential therapeutics for certain neurological conditions, including neuropathic pain, epilepsy, and ischaemia that are linked to NMDA receptor dysfunction are reviewed.


Assuntos
Conotoxinas , Receptores de N-Metil-D-Aspartato , Animais , Conotoxinas/química , Camundongos , Venenos de Moluscos/química , Venenos de Moluscos/farmacologia , Peptídeos/química , Receptores de N-Metil-D-Aspartato/metabolismo , Relação Estrutura-Atividade
2.
Nat Prod Rep ; 39(1): 33-57, 2022 01 26.
Artigo em Inglês | MEDLINE | ID: mdl-34190283

RESUMO

Up to the end of 2020Every year, the appearance of marine biotoxins causes enormous socio-economic damage worldwide. Among the major groups of biotoxins, paralytic shellfish toxins, comprising saxitoxin and its analogues (STXs), are the ones that cause the most severe effects on humans, including death. However, the knowledge that currently exists on their chemistry, properties and mode of toxicological action is disperse and partially outdated. This review intends to systematically compile the dispersed information, updating and complementing it. With this purpose, it addresses several aspects related to the molecular structure of these toxins. Special focus is given to the bioconversion reactions that may occur in the different organisms (dinoflagellates, bivalves, and humans) and the possible mediators involved. A critical review of the most recently discovered analogues, the M-series toxins, is presented. Finally, a deep discussion about the relationship between the molecular structure (e.g., effect of the substituting groups and the net charge of the molecules) and the toxic activity of these molecules is performed, proposing the concept of "toxicological traffic light" based on the toxicity equivalency factors (TEFs).


Assuntos
Crustáceos/química , Toxinas Marinhas/química , Moluscos/química , Animais , Humanos , Toxinas Marinhas/farmacologia , Venenos de Moluscos/química , Venenos de Moluscos/farmacologia , Relação Estrutura-Atividade
3.
Pharmacol Biochem Behav ; 205: 173182, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33774007

RESUMO

Cannabinoid (CB) receptor agonists show robust antinociceptive effects in various pain models. However, most of the clinically potent CB1 receptor-active drugs derived from cannabis are considered concerning due to psychotomimetic side effects. Selective CB receptor ligands that do not induce CNS side effects are of clinical interest. The venoms of marine snail Conus are a natural source of various potent analgesic peptides, some of which are already FDA approved. In this study we evaluated the ability of several Conus venom extracts to interact with CB1 receptor. HEK293 cells expressing CB1 receptors were treated with venom extracts and CB1 receptor internalization was analyzed by immunofluorescence. Results showed C. textile (C. Tex) and C. miles (C. Mil) samples as the most potent. These were serially subfractionated by HPLC for subsequent analysis by internalization assays and for analgesic potency evaluated in the formalin test and after peripheral nerve injury. Intrathecal injection of C. Tex and C. Mil subfractions reduced flinching/licking behavior during the second phase of formalin test and attenuated thermal and mechanical allodynia in nerve injury model. Treatment with proteolytic enzymes reduced CB1 internalization of subfractions, indicating the peptidergic nature of CB1 active component. Further HPLC purification revealed two potent antinociceptive subfractions within C. Tex with CB1 and possible CB2 activity, with mild to no side effects in the CB tetrad assessment. CB conopeptides can be isolated from these active Conus venom-derived samples and further developed as novel analgesic agents for the treatment of chronic pain using cell based or gene therapy approaches.


Assuntos
Agonistas de Receptores de Canabinoides/farmacologia , Dor Crônica/tratamento farmacológico , Venenos de Moluscos/farmacologia , Analgésicos/farmacologia , Animais , Comportamento Animal/efeitos dos fármacos , Agonistas de Receptores de Canabinoides/administração & dosagem , Canabinoides/farmacologia , Dor Crônica/metabolismo , Caramujo Conus/química , Terapia Genética/métodos , Células HEK293 , Humanos , Hiperalgesia/tratamento farmacológico , Hiperalgesia/metabolismo , Injeções Espinhais , Venenos de Moluscos/administração & dosagem , Medição da Dor/efeitos dos fármacos , Doenças do Sistema Nervoso Periférico/tratamento farmacológico , Doenças do Sistema Nervoso Periférico/metabolismo , Ratos , Receptor CB1 de Canabinoide/metabolismo , Receptor CB2 de Canabinoide/metabolismo
4.
J Nat Prod ; 84(4): 1232-1243, 2021 04 23.
Artigo em Inglês | MEDLINE | ID: mdl-33764053

RESUMO

Natural products such as conotoxins have tremendous potential as tools for biomedical research and for the treatment of different human diseases. Conotoxins are peptides present in the venoms of predatory cone snails that have a rich diversity of pharmacological functions. One of the major bottlenecks in natural products research is the rapid identification and evaluation of bioactive molecules. To overcome this limitation, we designed a set of light-induced behavioral assays in zebrafish larvae to screen for bioactive conotoxins. We used this screening approach to test several unique conotoxins derived from different cone snail clades and discovered that a conorfamide from Conus episcopatus, CNF-Ep1, had the most dramatic alterations in the locomotor behavior of zebrafish larvae. Interestingly, CNF-Ep1 is also bioactive in several mouse assay systems when tested in vitro and in vivo. Our novel screening platform can thus accelerate the identification of bioactive marine natural products, and the first compound discovered using this assay has intriguing properties that may uncover novel neuronal circuitry.


Assuntos
Larva/efeitos dos fármacos , Locomoção/efeitos dos fármacos , Venenos de Moluscos/farmacologia , Neuropeptídeos/farmacologia , Peixe-Zebra , Animais , Caramujo Conus/química , Feminino , Masculino , Camundongos
5.
Asian Pac J Cancer Prev ; 22(S1): 49-57, 2021 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-33576212

RESUMO

BACKGROUND: Research on animal toxins have shown toxicity potential on cancerous cell and tissues in the cultures. Conotoxins obtained from marine cone snails show the highest toxicity potential, so that several human deaths have been attributed to this species of snail. These toxins have proven to be valuable agents to inhibit enzymes, channels and proteins, in the nervous systems of humans. METHODS: We have studied the effects of Conus textile crude venom on U87MG human glioma cells and their mitochondria as main inducers of apoptosis and human embryonic kidney 293 cells (HEK293) as non-cancerous normal control cells. Cellular toxicity assessments including MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay and measurement of caspase-3 activation as well as mitochondrial toxicity assays including measurement of the activity of succinate dehydrogenase (SDH) enzyme, mitochondrial swelling, reactive oxygen species (ROS) production, collapse of mitochondrial membrane potential (MMP) and cytochrome c release were performed in U87MG human glioma cells and HEK293 cells (as non-cancerous normal cells). RESULTS: The results illustrated the significant cytotoxic effect of Conus textile crude venom on U87MG human glioma cells, that inhibits 50% (IC50=10µg/mL) of the cell growth after 12 h of exposure. Viability measurement showed which the Conus textile crude venom is selectively cytotoxic to U87MG human glioma cells, and induced activation of caspase-3 and induction of cell apoptosis via through mitochondrial signaling. Conus textile crude venom also selectively increased mitochondria swelling, ROS formation, cytochrome c release and MMP decrease in cancerous mitochondria but not normal mitochondria. Conclusion; Based on the obtained results from this investigation, it is concluded that the Conus textile crude venom contains promising natural compounds to fight U87MG human glioma cells through activation of apoptosis intrinsic pathways.


Assuntos
Antineoplásicos/farmacologia , Apoptose , Glioblastoma/tratamento farmacológico , Mitocôndrias/efeitos dos fármacos , Venenos de Moluscos/farmacologia , Animais , Proliferação de Células , Glioblastoma/metabolismo , Glioblastoma/patologia , Humanos , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Espécies Reativas de Oxigênio/metabolismo , Células Tumorais Cultivadas
6.
J Proteomics ; 234: 104083, 2021 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-33373718

RESUMO

Using high-throughput BioPlex assays, we determined that six fractions from the venom of Conus nux inhibit the adhesion of various recombinant PfEMP-1 protein domains (PF08_0106 CIDR1α3.1, PF11_0521 DBL2ß3, and PFL0030c DBL3X and DBL5e) to their corresponding receptors (CD36, ICAM-1, and CSA, respectively). The protein domain-receptor interactions permit P. falciparum-infected erythrocytes (IE) to evade elimination in the spleen by adhering to the microvasculature in various organs including the placenta. The sequences for the main components of the fractions, determined by tandem mass spectrometry, yielded four T-superfamily conotoxins, one (CC-Loop-CC) with I-IV, II-III connectivity and three (CC-Loop-CXaaC) with a I-III, II-IV connectivity. The 3D structure for one of the latter, NuxVA = GCCPAPLTCHCVIY, revealed a novel scaffold defined by double turns forming a hairpin-like structure stabilized by the two disulfide bonds. Two other main fraction components were a miniM conotoxin, and a O2-superfamily conotoxin with cysteine framework VI/VII. This study is the first one of its kind suggesting the use of conotoxins for developing pharmacological tools for anti-adhesion adjunct therapy against malaria. Similarly, mitigation of emerging diseases like AIDS and COVID-19, can also benefit from conotoxins as inhibitors of protein-protein interactions as treatment. BIOLOGICAL SIGNIFICANCE: Among the 850+ species of cone snail species there are hundreds of thousands of diverse venom exopeptides that have been selected throughout several million years of evolution to capture prey and deter predators. They do so by targeting several surface proteins present in target excitable cells. This immense biomolecular library of conopeptides can be explored for potential use as therapeutic leads against persistent and emerging diseases affecting non-excitable systems. We aim to expand the pharmacological reach of conotoxins/conopeptides by revealing their in vitro capacity to disrupt protein-protein and protein-polysaccharide interactions that directly contribute to pathology of Plasmodium falciparum malaria. This is significant for severe forms of malaria, which might be deadly even after treated with current parasite-killing drugs because of persistent cytoadhesion of P. falciparum infected erythrocytes even when parasites within red blood cells are dead. Anti-adhesion adjunct drugs would de-sequester or prevent additional sequestration of infected erythrocytes and may significantly improve survival of malaria patients. These results provide a lead for further investigations into conotoxins and other venom peptides as potential candidates for anti-adhesion or blockade-therapies. This study is the first of its kind and it suggests that conotoxins can be developed as pharmacological tools for anti-adhesion adjunct therapy against malaria. Similarly, mitigation of emerging diseases like AIDS and COVID-19, can also benefit from conotoxins as potential inhibitors of protein-protein interactions as treatment.


Assuntos
Antígenos CD36 , Enzimas Reparadoras do DNA , Eritrócitos , Molécula 1 de Adesão Intercelular , Venenos de Moluscos , Plasmodium falciparum , Fatores de Transcrição , Animais , Antígenos CD36/química , Antígenos CD36/metabolismo , COVID-19 , Caramujo Conus , Enzimas Reparadoras do DNA/química , Enzimas Reparadoras do DNA/metabolismo , Eritrócitos/química , Eritrócitos/metabolismo , Eritrócitos/parasitologia , Humanos , Molécula 1 de Adesão Intercelular/química , Molécula 1 de Adesão Intercelular/metabolismo , Venenos de Moluscos/química , Venenos de Moluscos/farmacologia , Plasmodium falciparum/química , Plasmodium falciparum/metabolismo , Domínios Proteicos , Proteínas de Protozoários , SARS-CoV-2 , Fatores de Transcrição/química , Fatores de Transcrição/metabolismo
7.
Mar Drugs ; 18(8)2020 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-32823677

RESUMO

Recently, Conorfamide-Sr3 (CNF-Sr3) was isolated from the venom of Conus spurius and was demonstrated to have an inhibitory concentration-dependent effect on the Shaker K+ channel. The voltage-gated potassium channels play critical functions on cellular signaling, from the regeneration of action potentials in neurons to the regulation of insulin secretion in pancreatic cells, among others. In mammals, there are at least 40 genes encoding voltage-gated K+ channels and the process of expression of some of them may include alternative splicing. Given the enormous variety of these channels and the proven use of conotoxins as tools to distinguish different ligand- and voltage-gated ion channels, in this work, we explored the possible effect of CNF-Sr3 on four human voltage-gated K+ channel subtypes homologous to the Shaker channel. CNF-Sr3 showed a 10 times higher affinity for the Kv1.6 subtype with respect to Kv1.3 (IC50 = 2.7 and 24 µM, respectively) and no significant effect on Kv1.4 and Kv1.5 at 10 µM. Thus, CNF-Sr3 might become a novel molecular probe to study diverse aspects of human Kv1.3 and Kv1.6 channels.


Assuntos
Venenos de Moluscos/farmacologia , Bloqueadores dos Canais de Potássio/farmacologia , Superfamília Shaker de Canais de Potássio/antagonistas & inibidores , Animais , Caramujo Conus , Ativação do Canal Iônico , Canal de Potássio Kv1.3/antagonistas & inibidores , Canal de Potássio Kv1.3/genética , Canal de Potássio Kv1.3/metabolismo , Canal de Potássio Kv1.4/antagonistas & inibidores , Canal de Potássio Kv1.4/genética , Canal de Potássio Kv1.4/metabolismo , Canal de Potássio Kv1.5/antagonistas & inibidores , Canal de Potássio Kv1.5/genética , Canal de Potássio Kv1.5/metabolismo , Canal de Potássio Kv1.6/antagonistas & inibidores , Canal de Potássio Kv1.6/genética , Canal de Potássio Kv1.6/metabolismo , Potenciais da Membrana , Oócitos , Superfamília Shaker de Canais de Potássio/genética , Superfamília Shaker de Canais de Potássio/metabolismo , Xenopus laevis
8.
Nat Struct Mol Biol ; 27(7): 615-624, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32483339

RESUMO

Human insulin and its current therapeutic analogs all show propensity, albeit varyingly, to self-associate into dimers and hexamers, which delays their onset of action and makes blood glucose management difficult for people with diabetes. Recently, we described a monomeric, insulin-like peptide in cone-snail venom with moderate human insulin-like bioactivity. Here, with insights from structural biology studies, we report the development of mini-Ins-a human des-octapeptide insulin analog-as a structurally minimal, full-potency insulin. Mini-Ins is monomeric and, despite the lack of the canonical B-chain C-terminal octapeptide, has similar receptor binding affinity to human insulin. Four mutations compensate for the lack of contacts normally made by the octapeptide. Mini-Ins also has similar in vitro insulin signaling and in vivo bioactivities to human insulin. The full bioactivity of mini-Ins demonstrates the dispensability of the PheB24-PheB25-TyrB26 aromatic triplet and opens a new direction for therapeutic insulin development.


Assuntos
Antígenos CD/química , Insulina/química , Venenos de Moluscos/química , Venenos de Moluscos/metabolismo , Receptor de Insulina/química , Substituição de Aminoácidos , Animais , Antígenos CD/metabolismo , Sítios de Ligação , Cristalografia por Raios X , Insulina/análogos & derivados , Insulina/metabolismo , Insulina/farmacologia , Camundongos Endogâmicos C57BL , Modelos Moleculares , Simulação de Dinâmica Molecular , Venenos de Moluscos/genética , Venenos de Moluscos/farmacologia , Peptídeos/química , Peptídeos/genética , Peptídeos/metabolismo , Conformação Proteica , Ratos Sprague-Dawley , Receptor de Insulina/metabolismo , Relação Estrutura-Atividade , Tirosina
9.
Front Immunol ; 11: 428, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32231667

RESUMO

Gastropod Molluscs rely exclusively on the innate immune system to protect from pathogens, defending their embryos through maternally transferred effectors. In this regard, Pomacea snail eggs, in addition to immune defenses, have evolved the perivitellin-2 or PV2 combining two immune proteins into a neurotoxin: a lectin and a pore-forming protein from the Membrane Attack Complex/Perforin (MACPF) family. This binary structure resembles AB-toxins, a group of toxins otherwise restricted to bacteria and plants. Many of these are enterotoxins, leading us to explore this activity in PV2. Enterotoxins found in bacteria and plants act mainly as pore-forming toxins and toxic lectins, respectively. In animals, although both pore-forming proteins and lectins are ubiquitous, no enterotoxins have been reported. Considering that Pomacea snail eggs ingestion induce morpho-physiological changes in the intestinal mucosa of rodents and is cytotoxic to intestinal cells in culture, we seek for the factor causing these effects and identified PmPV2 from Pomacea maculata eggs. We characterized the enterotoxic activity of PmPV2 through in vitro and in vivo assays. We determined that it withstands the gastrointestinal environment and resisted a wide pH range and enzymatic proteolysis. After binding to Caco-2 cells it promoted changes in surface morphology and an increase in membrane roughness. It was also cytotoxic to both epithelial and immune cells from the digestive system of mammals. It induced enterocyte death by a lytic mechanism and disrupted enterocyte monolayers in a dose-dependent manner. Further, after oral administration to mice PmPV2 attached to enterocytes and induced large dose-dependent morphological changes on their small intestine mucosa, reducing the absorptive surface. Additionally, PmPV2 was detected in the Peyer's patches where it activated lymphoid follicles and triggered apoptosis. We also provide evidence that the toxin can traverse the intestinal barrier and induce oral adaptive immunity with evidence of circulating antibody response. As a whole, these results indicate that PmPV2 is a true enterotoxin, a role that has never been reported to lectins or perforin in animals. This extends by convergent evolution the presence of plant- and bacteria-like enterotoxins to animals, thus expanding the diversity of functions of MACPF proteins in nature.


Assuntos
Enterotoxinas/farmacologia , Imunidade Inata/imunologia , Mucosa Intestinal/efeitos dos fármacos , Venenos de Moluscos/farmacologia , Caramujos/imunologia , Animais , Complexo de Ataque à Membrana do Sistema Complemento , Camundongos , Óvulo/imunologia , Óvulo/metabolismo , Perforina/metabolismo
10.
Bioconjug Chem ; 31(1): 64-73, 2020 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-31790574

RESUMO

Disulfide-rich animal venom peptides targeting either the voltage-sensing domain or the pore domain of voltage-gated sodium channel 1.7 (NaV1.7) have been widely studied as drug leads and pharmacological probes for the treatment of chronic pain. However, despite intensive research efforts, the full potential of NaV1.7 as a therapeutic target is yet to be realized. In this study, using evolved sortase A, we enzymatically ligated two known NaV1.7 inhibitors-PaurTx3, a spider-derived peptide toxin that modifies the gating mechanism of the channel through interaction with the voltage-sensing domain, and KIIIA, a small cone snail-derived peptide inhibitor of the pore domain-with the aim of creating a bivalent inhibitor which could interact simultaneously with two noncompeting binding sites. Using electrophysiology, we determined the activity at NaV1.7, and to maximize potency, we systematically evaluated the optimal linker length, which was nine amino acids. Our optimized synthetic bivalent peptide showed improved channel affinity and potency at NaV1.7 compared to either PaurTx3 or KIIIA individually. This work shows that novel and improved NaV1.7 inhibitors can be designed by combining a pore blocker toxin and a gating modifier toxin to confer desired pharmacological properties from both the voltage sensing domain and the pore domain.


Assuntos
Canal de Sódio Disparado por Voltagem NAV1.7/metabolismo , Peptídeos/química , Peptídeos/farmacologia , Bloqueadores do Canal de Sódio Disparado por Voltagem/química , Bloqueadores do Canal de Sódio Disparado por Voltagem/farmacologia , Sequência de Aminoácidos , Animais , Células HEK293 , Humanos , Modelos Moleculares , Venenos de Moluscos/química , Venenos de Moluscos/farmacologia , Caramujos/química , Venenos de Aranha/química , Venenos de Aranha/farmacologia , Aranhas/química
11.
Mar Drugs ; 17(10)2019 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-31627357

RESUMO

Increasingly cancer is being viewed as a channelopathy because the passage of ions via ion channels and transporters mediate the regulation of tumor cell survival, death, and motility. As a result, a potential targeted therapy for cancer is to use venom peptides that are selective for ion channels and transporters overexpressed in tumor cells. Here we describe the selectivity and mechanism of action of terebrid snail venom peptide, Tv1, for treating the most common type of liver cancer, hepatocellular carcinoma (HCC). Tv1 inhibited the proliferation of murine HCC cells and significantly reduced tumor size in Tv1-treated syngeneic tumor-bearing mice. Tv1's mechanism of action involves binding to overexpressed transient receptor potential (TRP) channels leading to calcium dependent apoptosis resulting from down-regulation of cyclooxygenase-2 (COX-2). Our findings demonstrate the importance of modulating ion channels and the unique potential of venom peptides as tumor specific ligands in the quest for targeted cancer therapies.


Assuntos
Neoplasias Hepáticas/tratamento farmacológico , Venenos de Moluscos/farmacologia , Peptídeos/farmacologia , Animais , Apoptose/efeitos dos fármacos , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Ciclo-Oxigenase 2/metabolismo , Regulação para Baixo/efeitos dos fármacos , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Células Hep G2 , Humanos , Neoplasias Hepáticas/metabolismo , Camundongos , Camundongos Endogâmicos BALB C
12.
Mol Biol Rep ; 46(5): 5479-5486, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31102148

RESUMO

Constant research into the pharmaceutical properties of marine natural products has led to the discovery of many potentially active agents considered worthy of medical applications. Genus Conus, which approximately comprises 700 species, is currently under every researcher's interest because of the conopeptides in their crude venom. Conopeptides have a wide range of pharmacological classes and properties. This research focused on the crude venom of Conus striatus to assess its analgesic activity, mutagenicity, nephrotoxicity, and hepatotoxicity in mice. The crude venom was extracted from the conus snails and the protein concentration was determined using Bradford's method. The analgesic activity of the venom was determined using the hot-plate method and standard IFCC method was used to determine the alanine aminotransferase (ALT) and aspartate aminotransferase (AST). Evaluation of mutagenicity was done using micronucleus assay and the nephrotoxicity of the venom was determined using Kidney Coefficient and serum creatinine concentration. The maximum tolerable dose (MTD) of the crude venom was found to be 75 ppm. The venom exhibited potent analgesic activity even higher than the positive control (Ibuprofen). Most of the analgesic drugs can usually impact damage in the liver and kidneys. However, AST and ALT results revealed that the venom has no adverse effects on the liver. Although the venom increased the incidence of micronucleated polychromatic erythrocytes, making it mutagenic, with MTD concentration's mutagenicity comparable to the positive control methyl methanesulfonate (MMS). The kidney coefficients, on the other hand, showed no significant difference between the treated groups and that of the untreated group. The serum creatinine also showed a concentration-dependent increase; with MTD treated mice got the highest creatinine concentration. However, MTD/2 and MTD/4 showed no significant difference in creatinine levels with respect to the untreated groups. Hence, the nephrotoxicity of the venom was only evident when used at higher concentration. The venom exhibited potent analgesic activity indicated that the C. striatus crude venom extract could have a potential therapeutic component as analgesic drugs that displayed no hepatic damage. This study also suggests that for this venom to be utilized for future medical applications, their usage must be regulated and properly monitored to avoid nephrotoxic effect.


Assuntos
Venenos de Moluscos/metabolismo , Venenos de Moluscos/farmacologia , Alanina Transaminase/sangue , Analgésicos/metabolismo , Analgésicos/farmacologia , Animais , Aspartato Aminotransferases/sangue , Caramujo Conus , Creatinina/sangue , Feminino , Rim/efeitos dos fármacos , Rim/metabolismo , Fígado/efeitos dos fármacos , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Peçonhas/metabolismo
13.
Biochem Pharmacol ; 164: 342-348, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31028742

RESUMO

Conorfamides are a poorly studied family of cone snail venom peptides with broad biological activities, including inhibition of glutamate receptors, acid-sensing ion channels, and voltage-gated potassium channels. The aim of this study was to characterize the pharmacological activity of two novel linear conorfamides (conorfamide_As1a and conorfamide_As2a) and their non-amidated counterparts (conopeptide_As1b and conopeptide_As2b) that were isolated from the venom of the Mexican cone snail Conus austini. Although As1a, As2a, As1b and As2b were identified by activity-guided fractionation using a high-throughput fluorescence imaging plate reader (FLIPR) assay assessing α7 nAChR activity, sequence determination revealed activity associated with four linear peptides of the conorfamide rather than the anticipated α-conotoxin family. Pharmacological testing revealed that the amidated peptide variants altered desensitization of acid-sensing ion channels (ASICs) 1a and 3, and the native lysine to arginine mutation differentiating As1a and As1b from As2a and As2b introduced ASIC1a peak current potentiation. Surprisingly, these conorfamides also inhibited α7 and muscle-type nicotinic acetylcholine receptors (nAChR) at nanomolar concentrations. This is the first report of conorfamides with dual activity, with the nAChR activity being the most potent molecular target of any conorfamide discovered to date.


Assuntos
Bloqueadores do Canal Iônico Sensível a Ácido/farmacologia , Canais Iônicos Sensíveis a Ácido/fisiologia , Venenos de Moluscos/farmacologia , Neuropeptídeos/farmacologia , Antagonistas Nicotínicos/farmacologia , Receptores Nicotínicos/fisiologia , Bloqueadores do Canal Iônico Sensível a Ácido/isolamento & purificação , Animais , Caramujo Conus , Relação Dose-Resposta a Droga , Feminino , Humanos , Venenos de Moluscos/isolamento & purificação , Neuropeptídeos/isolamento & purificação , Antagonistas Nicotínicos/isolamento & purificação , Xenopus laevis
14.
Int J Biol Macromol ; 124: 1145-1155, 2019 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-30521901

RESUMO

A low molecular weight posterior salivary gland (PSG) toxin was isolated and purified from the cuttlefish Sepia prashadi by Reverse Phase High Performance Liquid Chromatography (RP-HPLC). The protein and neutral sugar content of the PSG toxin was determined to be 1.033 mg/g and 282 µg/g. Fourier Transform Infrared (FT-IR) spectroscopy revealed the presence of υ-OH, υ-CO and δ-NH functional groups. Circular Dichroism (CD) spectroscopy and K2D2 analysis quantified the presence of 38.39% α-helix and 9.25% ß-sheet and 52.36% of ß-turn. Matrix Assisted Laser Desorption/Ionization-Time-of Flight/Mass Spectrometry (MALDI-TOF/MS) and MASCOT analysis revealed the amino acid sequence of MEMQSKQQNSKAPANRKIFPWMKTSAVATASKRVEMASLLNLQERQIKIWFQNRMKQKSQQPQTR (1.92 kDa) homologous to homeobox protein H4 of pufferfish, T. rubripes. The PSG toxin showed differential stability with pH and induced premature hatching in Zebrafish eggs and dose dependant developmental malformations in embryos with a Maximum tolerated dose of 1.85 µM. The PSG toxin exhibited significant antibacterial activity with pronounced zone of inhibition against S. typhimurium (12.94 mm) and inhibited avian RBC binding of Newcastle Disease virus (NDV) at a titre value of 1/4. The present study strongly advocates the biomedical potential of the PSG toxin from S. prashadi and illustrates its promise as a potential avian antimicrobial agent of the future.


Assuntos
Antibacterianos/farmacologia , Antivirais/farmacologia , Decapodiformes/química , Glicoproteínas/farmacologia , Venenos de Moluscos/farmacologia , Glândulas Salivares/química , Sequência de Aminoácidos , Animais , Antibacterianos/química , Antibacterianos/isolamento & purificação , Antivirais/química , Antivirais/isolamento & purificação , Galinhas , Embrião não Mamífero/efeitos dos fármacos , Eritrócitos/efeitos dos fármacos , Eritrócitos/virologia , Glicoproteínas/química , Glicoproteínas/isolamento & purificação , Concentração de Íons de Hidrogênio , Testes de Sensibilidade Microbiana , Peso Molecular , Venenos de Moluscos/química , Venenos de Moluscos/isolamento & purificação , Vírus da Doença de Newcastle/efeitos dos fármacos , Vírus da Doença de Newcastle/fisiologia , Conformação Proteica em alfa-Hélice , Conformação Proteica em Folha beta , Estabilidade Proteica , Salmonella typhimurium/efeitos dos fármacos , Salmonella typhimurium/crescimento & desenvolvimento , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Ligação Viral/efeitos dos fármacos , Peixe-Zebra , Zigoto/efeitos dos fármacos , Zigoto/crescimento & desenvolvimento
15.
Sci Rep ; 8(1): 15276, 2018 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-30323294

RESUMO

The number of newly discovered peptides from the transcriptomes and proteomes of animal venom arsenals is rapidly increasing, resulting in an abundance of uncharacterized peptides. There is a pressing need for a systematic, cost effective, and scalable approach to identify physiological effects of venom peptides. To address this discovery-to-function gap, we developed a sequence driven:activity-based hybrid approach for screening venom peptides that is amenable to large-venom peptide libraries with minimal amounts of peptide. Using this approach, we characterized the physiological and behavioral phenotypes of two peptides from the venom of predatory terebrid marine snails, teretoxins Tv1 from Terebra variegata and Tsu1.1 from Terebra subulata. Our results indicate that Tv1 and Tsu1.1 have distinct bioactivity. Tv1 (100 µM) had an antinociceptive effect in adult Drosophila using a thermal nociception assay to measure heat avoidance. Alternatively, Tsu1.1 (100 µM) increased food intake. These findings describe the first functional bioactivity of terebrid venom peptides in relation to pain and diet and indicate that Tv1 and Tsu1.1 may, respectively, act as antinociceptive and orexigenic agents. Tv1 and Tsu1.1 are distinct from previously identified venom peptides, expanding the toolkit of peptides that can potentially be used to investigate the physiological mechanisms of pain and diet.


Assuntos
Comportamento Animal/efeitos dos fármacos , Drosophila/efeitos dos fármacos , Venenos de Moluscos/química , Peptídeos/química , Animais , Comportamento Animal/fisiologia , Drosophila/fisiologia , Venenos de Moluscos/farmacologia , Venenos de Moluscos/toxicidade , Peptídeos/farmacologia , Peptídeos/toxicidade , Caramujos/química , Caramujos/genética , Transcriptoma/genética
16.
Toxicon ; 154: 28-34, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30243794

RESUMO

Members of Mas related G-protein coupled receptors (Mrgpr) are known to mediate itch. To date, several compounds have been shown to activate these receptors, including chloroquine, a common antimalarial drug, and peptides of the RF-amide family. However, specific ligands for these receptors are still lacking and there is a need for novel compounds that can be used to modulate the receptors in order to understand the cellular and molecular mechanism in which they mediate itch. Some cone snail venoms were previously shown to induce itch in mice. Here, we show that the venom of Conus textile induces itch through activation of itch-sensing sensory neurons, marked by their sensitivity to chloroquine. Two RF-amide peptides, CNF-Tx1 and CNF-Tx2, were identified in a C. textile venom gland transcriptome. These belong to the conorfamide family of peptides which includes previously described peptides from the venoms of Conus victoriae (CNF-Vc1) and Conus spurius (CNF-Sr1 and CNF-Sr2). We show that CNF-Vc1 and CNF-Sr1 activate MrgprC11 whereas CNF-Vc1 and CNF-Tx2 activate the human MrgprX1 (hMrgprX1). The peptides CNF-Tx1 and CNF-Sr2 do not activate MrgprC11 or hMrgprX1. Intradermal injection of CNF-Vc1 and CNF-Tx2 into the cheek of a transgenic mouse expressing hMrgprX1 instead of endogenous mouse Mrgprs resulted in itch-related scratching thus demonstrating the in vivo activity of these peptides. Using truncated analogues of CNF-Vc1, we identified amino acids at positions 7-14 as important for activity against hMrgprX1. The conopeptides reported here are tools that can be used to advance our understanding of the cellular and molecular mechanism of itch mediated by Mrgprs.


Assuntos
Conotoxinas/farmacologia , Prurido/induzido quimicamente , Prurido/fisiopatologia , Receptores Acoplados a Proteínas G/fisiologia , Sequência de Aminoácidos , Animais , Células Cultivadas , Masculino , Camundongos , Venenos de Moluscos/farmacologia , Neuropeptídeos/farmacologia , Peptídeos , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/biossíntese , Receptores Acoplados a Proteínas G/genética , Células Receptoras Sensoriais/efeitos dos fármacos , Células Receptoras Sensoriais/fisiologia , Transcriptoma
17.
Mol Pharmacol ; 94(4): 1114-1124, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30012583

RESUMO

Acid-sensing ion channel 3 (ASIC3) is a proton-gated Na+ channel with important roles in pain. ASIC3 quickly desensitizes in less than a second, limiting its capacity to sense sustained acidosis during pain. RFamide neuropeptides are modulators of ASIC3 that slow its desensitization and induce a variable sustained current. The molecular mechanism of slowed desensitization and the RFamide binding site on ASIC3 are unknown. RPRFamide, a RFamide from the venom of a cone snail, has a comparatively high affinity for ASIC3 and strongly slows its desensitization. Here we show that covalent binding of a UV-sensitive RPRFamide variant to ASIC3 prevents desensitization, suggesting that RPRFamide has to unbind from ASIC3 before it can desensitize. Moreover, we show by in silico docking to a homology model of ASIC3 that a cavity in the lower palm domain, which is also known as the nonproton ligand-sensing domain, is a potential binding site of RPRFamide. Finally, using extensive mutagenesis of residues lining the nonproton ligand-sensing domain, we confirm that this domain is essential for RPRFamide modulation of ASIC3. As comparative analysis of ASIC crystal structures in the open and in the desensitized conformation suggests that the lower palm domain contracts during desensitization, our results collectively suggest that RPRFamide, and probably also other RFamide neuropeptides, bind to the nonproton ligand-sensing domain to stabilize the open conformation of ASIC3.


Assuntos
Canais Iônicos Sensíveis a Ácido/metabolismo , Venenos de Moluscos/farmacologia , Neuropeptídeos/farmacologia , Animais , Feminino , Ligantes , Dor/tratamento farmacológico , Conformação Proteica , Domínios Proteicos , Prótons , Ratos , Xenopus laevis/metabolismo
18.
Toxicon ; 138: 53-58, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28774677

RESUMO

Conorfamides (CNFs) are toxins initially characterized from the venom duct of the venomous marine snail Conus spurius from the Gulf of Mexico; at their C-termini, these toxins are amidated and have high sequence similarity with the molluskan cardioexcitatory tetrapeptide Phe-Met-Arg-Phe-NH2 (FMRFamide or FMRFa) and other FMRFa-related peptides (FaRPs) found in the five molluskan classes, and in other invertebrate and vertebrate phyla. These peptides were the first FaRPs found to be present in any venom, and they are biologically active in mice, limpets, and/or freshwater snails. However, the molecular targets of the known CNFs (CNF-Sr1 and CNF-Sr2 from C. spurius, and CNF-Vc1 from C. victoriae) remain unidentified. Very recently, three FaRPs from C. textile have been found to potentiate the currents of acid-sensing ion channels. In this work, we characterized a novel conorfamide, CNF-Sr3 (ATSGPMGWLPVFYRF-NH2), comprised of 15 amino acid residues, and with a specific blocking activity for the Shaker subtype of the voltage-gated potassium channels, without significant effect on the Shab, Shaw, Shal and Eag channels. This peptide is the third type of disulfide-free conotoxins that has been discovered to target K+ channels.


Assuntos
Caramujo Conus/química , Venenos de Moluscos/química , Neuropeptídeos/farmacologia , Peptídeos/farmacologia , Superfamília Shaker de Canais de Potássio/antagonistas & inibidores , Sequência de Aminoácidos , Animais , Baculoviridae , Células HEK293 , Humanos , Venenos de Moluscos/síntese química , Venenos de Moluscos/farmacologia , Neuropeptídeos/síntese química , Neuropeptídeos/química , Peptídeos/química , Bloqueadores dos Canais de Potássio/química , Bloqueadores dos Canais de Potássio/farmacologia , Células Sf9/virologia
19.
Neuropharmacology ; 127: 196-223, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28623170

RESUMO

The nicotinic acetylcholine receptor (nAChR) represents the prototype of ligand-gated ion channels. It is vital for neuromuscular transmission and an important regulator of neurotransmission. A variety of toxic compounds derived from diverse species target this receptor and have been of elemental importance in basic and applied research. They enabled milestone discoveries in pharmacology and biochemistry ranging from the original formulation of the receptor concept, the first isolation and structural analysis of a receptor protein (the nAChR) to the identification, localization, and differentiation of its diverse subtypes and their validation as a target for therapeutic intervention. Among the venom-derived compounds, α-neurotoxins and α-conotoxins provide the largest families and still represent indispensable pharmacological tools. Application of modified α-neurotoxins provided substantial structural and functional details of the nAChR long before high resolution structures were available. α-bungarotoxin represents not only a standard pharmacological tool and label in nAChR research but also for unrelated proteins tagged with a minimal α-bungarotoxin binding motif. A major advantage of α-conotoxins is their smaller size, as well as superior selectivity for diverse nAChR subtypes that allows their development into ligands with optimized pharmacological and chemical properties and potentially novel drugs. In the following, these two groups of nAChR antagonists will be described focusing on their respective roles in the structural and functional characterization of nAChRs and their development into research tools. In addition, we provide a comparative overview of the diverse α-conotoxin selectivities that can serve as a practical guide for both structure activity studies and subtype classification. This article is part of the Special Issue entitled 'Venom-derived Peptides as Pharmacological Tools.'


Assuntos
Venenos de Moluscos/farmacologia , Antagonistas Nicotínicos/farmacologia , Receptores Nicotínicos/metabolismo , Venenos de Serpentes/farmacologia , Animais , Desenho Assistido por Computador , Venenos de Moluscos/química , Antagonistas Nicotínicos/química , Receptores Nicotínicos/efeitos dos fármacos , Caramujos , Venenos de Serpentes/química , Serpentes
20.
Expert Opin Drug Discov ; 12(6): 611-623, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28398099

RESUMO

INTRODUCTION: Peptide toxins are potent and often exquisitely selective probes of the structure and function of ion channels and receptors, and as such are of significant interest to the pharmaceutical and biotech industries as both therapeutic leads and pharmacological tools. Their progression as clinical candidates, however, faces many of the challenges that are common to peptide drugs generally. Areas covered: The attributes of peptide toxins as therapeutic leads are outlined, as well as some of the limiting factors that have hampered the clinical development of many promising candidates. Strategies to overcome or circumvent these limitations are described, and their applications to peptide toxins from cone snails, sea anemones and scorpions are exemplified. Expert opinion: Peptide toxins have exceeded their promise as valuable pharmacological tools but have yet to yield the anticipated bounty of therapeutic leads. As the number of new peptides identified in venom transcriptomes and proteomes expands rapidly, screening approaches that capture those with genuine therapeutic potential are required, along with methods for enhancing the stability, pharmacokinetics and pharmacodynamics of these peptides.


Assuntos
Desenho de Fármacos , Descoberta de Drogas/métodos , Peptídeos/farmacologia , Animais , Venenos de Cnidários/isolamento & purificação , Venenos de Cnidários/farmacologia , Caramujo Conus/metabolismo , Humanos , Venenos de Moluscos/isolamento & purificação , Venenos de Moluscos/farmacologia , Peptídeos/isolamento & purificação , Proteoma , Venenos de Escorpião/isolamento & purificação , Venenos de Escorpião/farmacologia , Escorpiões/metabolismo , Anêmonas-do-Mar/metabolismo , Transcriptoma
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...