Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 67
Filtrar
1.
Artigo em Inglês | MEDLINE | ID: mdl-38649084

RESUMO

Melittin is a powerful toxin present in honeybee venom that is active in a wide range of animals, from insects to humans. Melittin exerts numerous biological, toxicological, and pharmacological effects, the most important of which is destruction of the cell membrane. The phospholipase activity of melittin and its ability to activate phospholipases in the venom contribute to these actions. Using analytical methods, we discovered that the honeybee Apis mellifera produces melittin not only in the venom gland but also in its fat body cells, which remain resistant to this toxin's effects. We suggest that melittin acts as an anti-bacterial agent, since its gene expression is significantly upregulated when honeybees are infected with Escherichia coli and Listeria monocytogenes bacteria; additionally, melittin effectively kills these bacteria in the disc diffusion test. We hypothesize that the chemical and physicochemical properties of the melittin molecule (hydrophilicity, lipophilicity, and capacity to form tetramers) in combination with reactive conditions (melittin concentration, salt concentration, pH, and temperature) are responsible for the targeted destruction of bacterial cells and apparent tolerance towards own tissue cells. Considering that melittin is an important current and, importantly, potential broad-spectrum medication, a thorough understanding of the observed phenomena may significantly increase its use in clinical practice.


Assuntos
Antibacterianos , Venenos de Abelha , Escherichia coli , Corpo Adiposo , Meliteno , Animais , Antibacterianos/farmacologia , Antibacterianos/toxicidade , Venenos de Abelha/farmacologia , Venenos de Abelha/toxicidade , Abelhas , Escherichia coli/efeitos dos fármacos , Corpo Adiposo/metabolismo , Proteínas de Insetos/metabolismo , Listeria monocytogenes/efeitos dos fármacos , Meliteno/farmacologia , Meliteno/toxicidade
2.
Toxicon ; 229: 107136, 2023 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-37116588

RESUMO

Bee stings represent a public health subject, but the mechanisms involved in bee venom toxicity are not yet fully understood. To evaluate the reactions of adrenocortical cells, through which organisms respond to stress, two honeybee venom components: melittin (Mlt) and phospholipase A2 (PLA2) were tested as potential chemical stressors. Modifications were investigated with transmission electron microscopy and microanalysis. A single dose of Mlt (31 mg/kg) or PLA2 (9.3 mg/kg) was injected in rats of groups ML and PL; daily doses of Mlt (350 µg/kg) or PLA2 (105 µg/kg) were injected 30 days in rats of groups M30 and P30. Adrenocortical cells in ML group showed ultrastructural degenerative alterations of nuclei, endoplasmic reticulum, and mitochondria that exhibited lipid inclusions and mitochondrial cristae (MC) re-organized into mono- or multimembrane large vesicles, and whorls of membranes. Many MC were degenerated. In the M30 group, similar ultrastructural changes, but of lower amplitude were noted; lipid cytosolic droplets were heterogenous. MC diameters in Mlt groups (melittin treated groups) were significantly higher than in control (C) group. In PL group, mitochondria contained large lipid inclusions, vesicular MC of different sizes and multiple membranes, and debris, or whorl structures. In P30 group MC were tubular with increased diameters. In both PLA2 groups (PLA2 treated groups) MC were significantly larger than in C group. We concluded that Mlt and PLA2 were powerful stressors, toxic at the tested doses, cellular reactions concerning in all groups mainly mitochondria, but also other cellular compartments. Apart from degenerative regression of MC, the rearrangement of tubular MC occurred into one or multiple large multimembrane vesicular MC. Reactions to the high doses were more pronounced, with the highest amplitude in ML group, and the lowest in P30 group.


Assuntos
Venenos de Abelha , Mordeduras e Picadas de Insetos , Abelhas , Ratos , Animais , Venenos de Abelha/toxicidade , Venenos de Abelha/química , Meliteno/toxicidade , Fosfolipases A2 , Mitocôndrias , Lipídeos
3.
Toxins (Basel) ; 14(5)2022 05 16.
Artigo em Inglês | MEDLINE | ID: mdl-35622592

RESUMO

Melittin, the main toxic component in the venom of the European honeybee, interacts with natural and artificial membranes due to its amphiphilic properties. Rather than interacting with a specific receptor, melittin interacts with the lipid components, disrupting the lipid bilayer and inducing ion leakage and osmotic shock. This mechanism of action is shared with pneumolysin and other members of the cholesterol-dependent cytolysin family. In this manuscript, we investigated the inverse correlation for cholesterol dependency of these two toxins. While pneumolysin-induced damage is reduced by pretreatment with the cholesterol-depleting agent methyl-ß-cyclodextrin, the toxicity of melittin, after cholesterol depletion, increased. A similar response was also observed after a short incubation with lipophilic simvastatin, which alters membrane lipid organization and structure, clustering lipid rafts. Therefore, changes in toxin sensitivity can be achieved in cells by depleting cholesterol or changing the lipid bilayer organization.


Assuntos
Bicamadas Lipídicas , Meliteno , Animais , Proteínas de Bactérias , Abelhas , Colesterol , Meliteno/química , Meliteno/toxicidade , Estreptolisinas/toxicidade
4.
ACS Appl Mater Interfaces ; 13(36): 42533-42542, 2021 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-34472829

RESUMO

Despite potency against a variety of cancers in preclinical systems, melittin (MEL), a major peptide in bee venom, exhibits non-specific toxicity, severe hemolytic activity, and poor pharmacological properties. Therefore, its advancement in the clinical translation system has been limited to early-stage trials. Herein, we report a biohybrid involving a bottlebrush-architectured poly(ethylene glycol) (PEG) and MEL. Termed pacMEL, the conjugate consists of a high-density PEG arrangement, which provides MEL with steric inhibition against protein access, while the high molecular weight of pacMEL substantially enhances plasma pharmacokinetics with a ∼10-fold increase in the area under the curve (AUC∞) compared to free MEL. pacMEL also significantly reduces hepatic damage and unwanted innate immune response and all but eliminated hemolytic activities of MEL. Importantly, pacMEL passively accumulates at subcutaneously inoculated tumor sites and exhibits stronger tumor-suppressive activity than molecular MEL. Collectively, pacMEL makes MEL a safer and more appealing drug candidate.


Assuntos
Antineoplásicos/uso terapêutico , Meliteno/análogos & derivados , Meliteno/uso terapêutico , Neoplasias/tratamento farmacológico , Polietilenoglicóis/uso terapêutico , Animais , Antineoplásicos/síntese química , Antineoplásicos/farmacocinética , Antineoplásicos/toxicidade , Linhagem Celular Tumoral , Feminino , Humanos , Meliteno/farmacocinética , Meliteno/toxicidade , Camundongos Endogâmicos C57BL , Polietilenoglicóis/síntese química , Polietilenoglicóis/farmacocinética , Polietilenoglicóis/toxicidade , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Chem Biol Interact ; 347: 109622, 2021 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-34375656

RESUMO

Glioblastoma multiforme (GBM) is a frequent form of malignant glioma. Strategic therapeutic approaches to treat this type of brain tumor currently involves a combination of surgery, radiotherapy and chemotherapy. Nevertheless, survival of GBM patients remains in the 12-15 months range following diagnosis. Development of novel therapeutic approaches for this malignancy is therefore of utmost importance. Interestingly, bee venom and its components have shown promising anti-cancer activities in various types of cancer even though information pertaining to GBMs have been limited. The current work was thus undertaken to better characterize the anti-cancer properties of bee venom and its components in Hs683, T98G and U373 human glioma cells. MTT-based cell viability assays revealed IC50 values of 7.12, 15.35 and 7.60 µg/mL for cell lines Hs683, T98G and U373 treated with bee venom, respectively. Furthermore, melittin treatment of these cell lines resulted in IC50 values of 7.77, 31.53 and 12.34 µg/mL, respectively. Cell viability assessment by flow cytometry analysis confirmed signs of late apoptosis and necrosis after only 1 h of treatment with either bee venom or melittin in all three cell lines. Immunoblotting-based quantification of apoptotic markers demonstrated increased expression of Bak and Bax, while Caspsase-3 levels were significantly lower when compared to control cells. Quantification by qRT-PCR showed increased expression levels of long non-coding RNAs RP11-838N2.4 and XIST in glioma cells treated with either bee venom or melittin. Overall, this study provides preliminary insight on molecular mechanisms via which bee venom and its main components can impact viability of glioma cells and warrants further investigation of its anticancer potential in gliomas.


Assuntos
Antineoplásicos/uso terapêutico , Glioblastoma/tratamento farmacológico , Meliteno/uso terapêutico , Antineoplásicos/toxicidade , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sinergismo Farmacológico , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glioblastoma/metabolismo , Humanos , Linfócitos/efeitos dos fármacos , Meliteno/toxicidade , Monócitos/efeitos dos fármacos , Necrose/tratamento farmacológico , Fosfolipases A2/uso terapêutico , RNA Longo não Codificante/metabolismo , Temozolomida/uso terapêutico
6.
BMC Pharmacol Toxicol ; 22(1): 42, 2021 07 14.
Artigo em Inglês | MEDLINE | ID: mdl-34261542

RESUMO

BACKGROUND: Melittin is one of the most studied antimicrobial peptides, and several in vitro experiments have demonstrated its antibacterial efficacy. However, there is evidence showing melittin has non-promising effects such as cytotoxicity and hemolysis. Therefore, concerns about unwanted collateral toxicity of melittin lie ahead in the path toward its clinical development. With these considerations, the present study aimed to fill the gap between in vitro and in vivo studies. METHODS: In the first step, in vitro toxicity profile of melittin was assessed using cytotoxicity and hemolysis tests. Next, a maximum intraperitoneal (i.p.) sub-lethal dose was determined using BALB/c mice. Besides toxicity, antimicrobial efficacy of melittin against extensively drug-resistant (XDR) Acinetobacter baumannii, methicillin-resistant Staphylococcus aureus (MRSA), and KPC-producing Klebsiella pneumonia (KPC-KP) pathogens were tested using both in vitro and in vivo methods. RESULTS: Melittin showed extensive hemolysis (HD50 = 0.44 µg/mL), and cytotoxicity (IC50 = 6.45 µg/mL) activities with i.p. LD50 value of 4.98 mg/kg in BALB/c mice. In vitro antimicrobial evaluation showed melittin MIC range from 8 to 32 µg/mL for the studied pathogens. Treatment of infected mice with repeated sub-lethal doses of melittin (2.4 mg/kg) displayed no beneficial effect on their survival and peritoneal bacterial loads. CONCLUSIONS: These results indicate that melittin at its safe dose could not exhibit antimicrobial activity, which hinders its application in clinical practice.


Assuntos
Antibacterianos/toxicidade , Meliteno/toxicidade , Infecções por Acinetobacter/tratamento farmacológico , Acinetobacter baumannii/efeitos dos fármacos , Acinetobacter baumannii/crescimento & desenvolvimento , Animais , Antibacterianos/uso terapêutico , Linhagem Celular , Farmacorresistência Bacteriana , Hemólise/efeitos dos fármacos , Humanos , Infecções por Klebsiella/tratamento farmacológico , Klebsiella pneumoniae/efeitos dos fármacos , Klebsiella pneumoniae/crescimento & desenvolvimento , Masculino , Meliteno/uso terapêutico , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos , Staphylococcus aureus Resistente à Meticilina/crescimento & desenvolvimento , Camundongos Endogâmicos BALB C , Testes de Sensibilidade Microbiana , Peritonite/tratamento farmacológico , Sepse/tratamento farmacológico , Infecções Estafilocócicas/tratamento farmacológico
7.
Biochem Biophys Res Commun ; 555: 32-39, 2021 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-33812056

RESUMO

Protein-protein (e.g., antibody-antigen) interactions comprise multiple weak interactions. We have previously reported that lipid nanoparticles (LNPs) bind to and neutralize target toxic peptides after multifunctionalization of the LNP surface (MF-LNPs) with amino acid derivatives that induce weak interactions; however, the MF-LNPs aggregated after target capture and showed short blood circulation times. Here we optimized polyethylene glycol (PEG)-modified MF-LNPs (PEG-MF-LNPs) to inhibit the aggregation and increase the blood circulation time. Melittin was used as a target toxin, and MF-LNPs were prepared with negatively charged, hydrophobic, and neutral amino-acid-derivative-conjugated functional lipids. In this study, MF-LNPs modified with only PEG5k (PEG5k-MF-LNPs) and with both PEG5k and PEG2k (PEGmix-MF-LNPs) were prepared, where PEG5k and PEG2k represent PEG with a molecular weight of 5000 and 2000, respectively. PEGylation of the MF-LNPs did not decrease the melittin neutralization ability of nonPEGylated MF-LNPs, as tested by hemolysis assay. The PEGmix-MF-LNPs showed better blood circulation characteristics than the PEG5k-MF-LNPs. Although the nonPEGylated MF-LNPs immediately aggregated when mixed with melittin, the PEGmix-MF-LNPs did not aggregate. The PEGmix-MF-LNPs dramatically increased the survival rate of melittin-treated mice, whereas the nonPEGylated MF-LNPs increased slightly. These results provide a fundamental strategy to improve the in vivo toxin neutralization ability of MF-LNPs.


Assuntos
Antídotos/farmacologia , Meliteno/toxicidade , Nanopartículas Multifuncionais/química , Polietilenoglicóis/química , Animais , Antídotos/química , Antídotos/farmacocinética , Bovinos , Linhagem Celular , Hemólise/efeitos dos fármacos , Interações Hidrofóbicas e Hidrofílicas , Lipídeos/química , Masculino , Meliteno/sangue , Meliteno/metabolismo , Meliteno/farmacocinética , Camundongos Endogâmicos BALB C , Nanopartículas Multifuncionais/administração & dosagem , Nanopartículas Multifuncionais/metabolismo , Distribuição Tecidual
8.
Acta Pharmacol Sin ; 42(8): 1256-1266, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32939034

RESUMO

Acute liver failure (ALF) is a fatal clinical syndrome with no special drug. Recent evidence shows that modulation of macrophage to inhibit inflammation may be a promising strategy for ALF treatment. In this study we investigated the potential therapeutic effects of melittin, a major peptide component of bee venom both in mice model of ALF and in LPS-stimulated macrophages in vitro, and elucidated the underlying mechanisms. ALF was induced in mice by intraperitoneal injection of D-galactosamine/LPS. Then the mice were treated with melittin (2, 4, and 8 mg/kg, ip). We showed that melittin treatment markedly improved mortality, attenuated severe symptoms and signs, and alleviated hepatic inflammation in D-galactosamine/LPS-induced ALF mice with the optimal dose being 4 mg/kg. In addition, melittin within the effective doses did not cause significant in vivo toxicity. In LPS-stimulated RAW264.7 macrophages, melittin (0.7 µM) exerted anti-oxidation and anti-inflammation effects. We showed that LPS stimulation promoted aerobic glycolysis of macrophages through increasing glycolytic rate, upregulated the levels of Warburg effect-related enzymes and metabolites including lactate, LDHA, LDH, and GLUT-1, and activated Akt/mTOR/PKM2/HIF-1α signaling. Melittin treatment suppressed M2 isoform of pyruvate kinase (PKM2), thus disrupted the Warburg effect to alleviate inflammation. Molecular docking analysis confirmed that melittin targeted PKM2. In LPS-stimulated RAW264.7 macrophages, knockdown of PKM2 caused similar anti-inflammation effects as melittin did. In D-galactosamine/LPS-induced ALF mice, melittin treatment markedly decreased the expression levels of PKM2 and HIF-1α in liver. This work demonstrates that melittin inhibits macrophage activation-mediated inflammation via inhibition of aerobic glycolysis by targeting PKM2, which highlights a novel strategy of using melittin for ALF treatment.


Assuntos
Anti-Inflamatórios/uso terapêutico , Antioxidantes/uso terapêutico , Glicólise/efeitos dos fármacos , Falência Hepática Aguda/tratamento farmacológico , Meliteno/uso terapêutico , Piruvato Quinase/metabolismo , Animais , Anti-Inflamatórios/metabolismo , Anti-Inflamatórios/toxicidade , Antioxidantes/metabolismo , Antioxidantes/toxicidade , Galactosamina , Inflamação/tratamento farmacológico , Inflamação/etiologia , Lipopolissacarídeos , Falência Hepática Aguda/induzido quimicamente , Falência Hepática Aguda/complicações , Masculino , Meliteno/metabolismo , Meliteno/toxicidade , Camundongos , Camundongos Endogâmicos C57BL , Simulação de Acoplamento Molecular , Ligação Proteica , Células RAW 264.7
9.
Angew Chem Int Ed Engl ; 59(26): 10461-10465, 2020 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-32203634

RESUMO

Inhibition of phospholipase A2 (PLA2) has long been considered for treating various diseases associated with an elevated PLA2 activity. However, safe and effective PLA2 inhibitors remain unavailable. Herein, we report a biomimetic nanoparticle design that enables a "lure and kill" mechanism designed for PLA2 inhibition (denoted "L&K-NP"). The L&K-NPs are made of polymeric cores wrapped with modified red blood cell membrane with two inserted key components: melittin and oleyloxyethyl phosphorylcholine (OOPC). Melittin acts as a PLA2 attractant that works together with the membrane lipids to "lure" in-coming PLA2 for attack. Meanwhile, OOPC acts as inhibitor that "kills" PLA2 upon enzymatic attack. Both compounds are integrated into the L&K-NP structure, which voids toxicity associated with free molecules. In the study, L&K-NPs effectively inhibit PLA2-induced hemolysis. In mice administered with a lethal dose of venomous PLA2, L&K-NPs also inhibit hemolysis and confer a significant survival benefit. Furthermore, L&K-NPs show no obvious toxicity in mice. and the design provides a platform technology for a safe and effective anti-PLA2 approach.


Assuntos
Materiais Biomiméticos/farmacologia , Meliteno/farmacologia , Nanopartículas/química , Inibidores de Fosfolipase A2/farmacologia , Fosfolipases A2/metabolismo , Fosforilcolina/análogos & derivados , Animais , Materiais Biomiméticos/química , Materiais Biomiméticos/toxicidade , Membrana Eritrocítica/química , Hemólise/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana , Humanos , Masculino , Meliteno/química , Meliteno/toxicidade , Camundongos Endogâmicos ICR , Nanopartículas/toxicidade , Inibidores de Fosfolipase A2/química , Inibidores de Fosfolipase A2/toxicidade , Fosforilcolina/química , Fosforilcolina/farmacologia , Fosforilcolina/toxicidade
10.
Int J Pharm ; 577: 119071, 2020 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-31991184

RESUMO

Gating modifier toxins (GMTs) from animal venom have shown great potential in controlling blood glucose levels in type II diabetes (T2D), but their high acute toxicity and quick clearance in the body hamper their potential therapeutic use. Inspired by their highly positive charge, we have developed a nanocomplex system based on polyelectrolytes, in which strong interactions form between positively charged GMTs and negatively charged dextran sulfate (DS). Using melittin as a model GMT and adapting flash nanocomplexation (FNC) technology for complex preparation, uniform nanocomplexes (polydispersity index: ~0.1) with high melittin encapsulation efficiency (~100%), high payload capacity (~30%), and tunable release profiles were formulated. In contrast to the high acute liver toxicity and low survival rate (60% after 8 days) observed after a single intraperitoneal (i.p.) injection of 3 mg/kg free melittin, melittin-loaded nanocomplexes displayed improved safety (100% survival after 8 days) due to prolonged melittin release. In a mouse model of T2D, a single i.p. injection of nanocomplexes decreased the blood glucose level to 12 mmol/L within 12 h and maintained it within the therapeutic range (<15 mmol/L) for 48 h. In addition, body weight decreased following treatment. This GMT/DS binary system shows great promise due to its simple components, facile preparation method, and enhanced potential druggability, including a decreased dosing frequency, decreased acute toxicity, and improved pathological indicators.


Assuntos
Glicemia/efeitos dos fármacos , Diabetes Mellitus Experimental/tratamento farmacológico , Diabetes Mellitus Tipo 2/tratamento farmacológico , Meliteno/administração & dosagem , Animais , Preparações de Ação Retardada , Sulfato de Dextrana/química , Portadores de Fármacos/química , Liberação Controlada de Fármacos , Feminino , Masculino , Meliteno/farmacologia , Meliteno/toxicidade , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Nanopartículas , Polieletrólitos/química , Testes de Toxicidade Aguda
11.
Sci Rep ; 8(1): 14329, 2018 09 25.
Artigo em Inglês | MEDLINE | ID: mdl-30254334

RESUMO

In recent years, synthetic peptides have been considered promising targets for drug development that possess low side-effects, are cost-effective and are susceptible to rational design. Hecate was initially described as a potent bacterial inhibitor and subsequently as an anticancer drug with functions related to its lipid interaction property. Viruses, such as hepatitis C virus (HCV), have a lipid-dependent life cycle and could be affected by Hecate in many ways. Here, we assessed modifications on Hecate's N-terminus region and its effects on HCV and hepatotoxicity. Gallic acid-conjugated Hecate was the most efficient Hecate-derivative, presenting high potential as an antiviral and inhibiting between 50 to 99% of all major steps within the HCV infectious cycle. However, the most promising aspect was GA-Hecate's mechanism of action, which was associated with a balanced lipid interaction with the viral envelope and lipid droplets, as well as dsRNA intercalation, allowing for the possibility to affect other ssRNA viruses and those with a lipid-dependent cycle.


Assuntos
Antivirais/química , Antivirais/farmacologia , Ácido Gálico/química , Hepacivirus/efeitos dos fármacos , Meliteno/química , Meliteno/farmacologia , Sequência de Aminoácidos , Antivirais/toxicidade , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Hepacivirus/fisiologia , Hepatócitos/citologia , Hepatócitos/efeitos dos fármacos , Humanos , Meliteno/toxicidade , Replicação Viral/efeitos dos fármacos
12.
Micron ; 112: 42-54, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29908421

RESUMO

We tested the ability of bee venom (BV), melittin (Mlt), and phospholipase A2 (PLA) - used in 5 concentrations each (5, 10, 15, 20 and 40 µg/100 µl) - to promote ultrastructural changes and reorganization of cristae in vitro in mitochondria isolated from rat adrenal cortex after a protocol optimized by us. Thus, apart from two control grups (CI and CS), in which the mitochondria were suspended into saline buffer and isolation medium respectively, 15 more groups of mitochondria were constituted, corresponding to the five different doses of the three substance tested (BV5 to M40; M5 to M40 and P5 to P40). The ultrastructural effects were quantified on transmission electron micrographs using a morphometry software. Values of 84.49 nm and 95.45 nm were calculated for median diameters of mitochondrial cristae in two control groups. Large and very large vesicular cristae, many with 2 or 3 membranes, were generated depending on dose among normal cristae in all treated groups. In the BV and Mlt treated groups, after an initial increase (up to 127.27 nm in V15 group and 151.2 nm in M10 group) due to stimulation of cristae fusion, the cristae diameter diminished as the doses increased, mainly by the collapse of the cristae. In the PLA treated groups, the cristae diameter increased continuously from 83.84 nm to 136.01 nm, by stimulated fusion of cristae, only the two largest doses promoting the collapse of cristae in some mitochondria. The highest percentage of abnormal cristae was found in the Mlt treated groups and next in BV treated groups. All substances tested produced pronounced ultrastructural variability of mitochondrial cristae in vitro: they also changed (depending on dose) mitochondrial shapes, generated matrix debris and the highest concentrations of BV and Mlt were responsible for mitochondrial breakdown. These ultrastructural alterations of mitochondrial criste in the presence of the BV molecules suggest a reduced capacity of adrenocortical mitochondria to synthetize steroid hormones consequently to BV envenomations and partially explain the toxic effects of the BV.


Assuntos
Córtex Suprarrenal/efeitos dos fármacos , Venenos de Abelha/toxicidade , Meliteno/toxicidade , Mitocôndrias/efeitos dos fármacos , Membranas Mitocondriais/ultraestrutura , Fosfolipases A2/toxicidade , Córtex Suprarrenal/ultraestrutura , Animais , Masculino , Microscopia Eletrônica de Transmissão , Mitocôndrias/ultraestrutura , Ratos , Ratos Wistar
13.
Int J Nanomedicine ; 13: 3251-3261, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29910613

RESUMO

BACKGROUND: Melittin, the main active peptide ingredient of bee venom, can cause severe cell membrane lysis due to its robust interaction with negatively charged phospholipids. So far, no effective anti-melittin vaccine has been developed to protect people from undesired melittin intoxication. METHODS: Herein, we prepared a polydiacetylene (PDA) nanoparticle with cell membrane-mimic surface to complex melittin, forming an anti-melittin vaccine (PDA-melittin). RESULTS: PDA nanoparticles could effectively combine with melittin and neutralize its toxicity. PDA-melittin nanocomplex is demonstrated to enhance melittin uptake by DCs and stimulate strong melittin-specific immunity. Mice immunized with PDA-melittin nanocomplex showed higher survival rate after exposion to melittin than untreated mice. CONCLUSION: The PDA-melittin nanocomplex can efficiently and safely generate a specific immunity against melittin to protect body from melittin intoxication, providing a new method with potential clinical application for the treatment of melittin intoxication.


Assuntos
Venenos de Abelha/química , Meliteno/imunologia , Nanopartículas/química , Vacinas/química , Vacinas/imunologia , Células 3T3 , Animais , Venenos de Abelha/toxicidade , Biomimética , Células Dendríticas , Feminino , Meliteno/toxicidade , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Polímero Poliacetilênico , Polímeros/química , Poli-Inos/química , Toxoides/imunologia , Vacinas/farmacologia
14.
Toxicon ; 141: 94-103, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29229236

RESUMO

In this study, we aimed to investigate the testicular toxicity of two molecules derived from bee venom (BV): phospholipase A2 (PlA2) and melittin (Mlt). Ultrastructural effects of purified BV PlA2 and Mlt were assessed consecutive to repeated dose (30 days) and acute toxicity studies. For the subchronic treatment, PlA2 and Mlt were injected in daily doses equivalent to those released by a bee sting (105 µg PlA2/kg/day and 350 µg Mlt/kg/day), while in the acute treatment their doses corresponded to those released by 100 bee stings (9.3 mg PlA2/kg and 31 mg Mlt/kg). Both PlA2 and Mlt affected the Leydig cells and the cells in seminiferous tubules, the Sertoli cells first of all. PlA2 injection resulted in detachment of the Sertoli cells from the surrounding cells, and extracellular vacuolations, cytoplasmic vacuolations in their basal region and in branches as well, detachment of spermatids, residual bodies and sometimes even spermatocytes into the lumen, changes that had a higher magnitude after the acute treatment. Mlt injection induced similar ultrastructural alterations, but more severe, including degeneration of cellular organelles and cellular necrosis, resulting into rarefaction of the seminiferous epithelium; the ultrastructural changes had a higher magnitude after the 30 repeated dose treatment. We concluded that either of the two molecules tested here, PlA2 and Mlt, were Sertoli cells toxicants at the used doses, and they participated both in the BV testicular toxicity. We consider the observed changes as part of a preceding mechanism of the more severe alterations produced by the BV. It also remains possible that these early unspecific changes reported here could represent the response of the SCs not only to the components of bee venom, but to molecules of other venoms as well. The Sertoli cells were the primary target of PlA2 and Mlt in the spermatogenic epithelium, and their alteration led to further degenerative changes of the germ cells. Since the exposure to PlA2 and Mlt caused severe alteration, including cell death and detachment of immature germ cells into the lumen, we may also conclude that the bee venom molecules had a potential to interfere with normal progression of spermatogenesis. All the degenerative changes observed in the Sertoli cells were accompanied with changes of the Leydig cells.


Assuntos
Venenos de Abelha/toxicidade , Meliteno/toxicidade , Fosfolipases A2/toxicidade , Testículo/efeitos dos fármacos , Animais , Venenos de Abelha/química , Masculino , Necrose , Ratos Wistar , Túbulos Seminíferos/efeitos dos fármacos , Túbulos Seminíferos/ultraestrutura , Células de Sertoli/efeitos dos fármacos , Células de Sertoli/ultraestrutura , Testículo/ultraestrutura , Testes de Toxicidade Aguda , Testes de Toxicidade Subcrônica
15.
J Hematol Oncol ; 10(1): 71, 2017 03 20.
Artigo em Inglês | MEDLINE | ID: mdl-28320480

RESUMO

Melittin is the main effective component of bee venom and has extensive biological functions; however, serious side effects have restricted its clinical application. Preclinical and clinical studies showed that the main adverse events were allergic reaction and pain at the administration site. To decrease the toxicity, we prepared melittin nano-liposomes by encapsulating melittin with poloxamer 188 and explored the inhibitory activities on liver cancer together with biological safety. Here, we showed that melittin nano-liposomes significantly inhibited the survival of hepatocellular carcinoma (HCC) cells in vitro and prominently suppressed the growth of subcutaneous and orthotopic HCC transplantation tumors in vivo. It was important that it induced less inflammation and allergy in mice compared with melittin. Overall, melittin nano-liposomes would have a better application in HCC therapy due to its significant anti-tumor activity and better biological safety.


Assuntos
Carcinoma Hepatocelular/tratamento farmacológico , Neoplasias Hepáticas/tratamento farmacológico , Meliteno/administração & dosagem , Poloxâmero/uso terapêutico , Animais , Abelhas , Cápsulas/química , Carcinoma Hepatocelular/complicações , Xenoenxertos , Humanos , Hipersensibilidade/prevenção & controle , Inflamação/induzido quimicamente , Inflamação/prevenção & controle , Lipossomos , Neoplasias Hepáticas/complicações , Meliteno/efeitos adversos , Meliteno/toxicidade , Camundongos , Nanopartículas
16.
Neurosci Bull ; 32(3): 265-72, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26983715

RESUMO

Melittin is a basic 26-amino-acid polypeptide that constitutes 40-60% of dry honeybee (Apis mellifera) venom. Although much is known about its strong surface activity on lipid membranes, less is known about its pain-producing effects in the nervous system. In this review, we provide lines of accumulating evidence to support the hypothesis that melittin is the major pain-producing substance of bee venom. At the psychophysical and behavioral levels, subcutaneous injection of melittin causes tonic pain sensation and pain-related behaviors in both humans and animals. At the cellular level, melittin activates primary nociceptor cells through direct and indirect effects. On one hand, melittin can selectively open thermal nociceptor transient receptor potential vanilloid receptor channels via phospholipase A2-lipoxygenase/cyclooxygenase metabolites, leading to depolarization of primary nociceptor cells. On the other hand, algogens and inflammatory/pro-inflammatory mediators released from the tissue matrix by melittin's pore-forming effects can activate primary nociceptor cells through both ligand-gated receptor channels and the G-protein-coupled receptor-mediated opening of transient receptor potential canonical channels. Moreover, subcutaneous melittin up-regulates Nav1.8 and Nav1.9 subunits, resulting in the enhancement of tetrodotoxin-resistant Na(+) currents and the generation of long-term action potential firing. These nociceptive responses in the periphery finally activate and sensitize the spinal dorsal horn pain-signaling neurons, resulting in spontaneous nociceptive paw flinches and pain hypersensitivity to thermal and mechanical stimuli. Taken together, it is concluded that melittin is the major pain-producing substance of bee venom, by which peripheral persistent pain and hyperalgesia (or allodynia), primary nociceptive neuronal sensitization, and CNS synaptic plasticity (or metaplasticity) can be readily induced and the molecular and cellular mechanisms underlying naturally-occurring venomous biotoxins can be experimentally unraveled.


Assuntos
Venenos de Abelha/toxicidade , Abelhas/química , Meliteno/toxicidade , Dor/induzido quimicamente , Animais , Nociceptores/efeitos dos fármacos , Nociceptores/fisiologia
17.
Toxicon ; 112: 59-67, 2016 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-26829652

RESUMO

Africanized Apis mellifera bees, also known as killer bees, have an exceptional defensive instinct, characterized by mass attacks that may cause envenomation or death. From the years 2000-2013, 77,066 bee accidents occurred in Brazil. Bee venom comprises several substances, including melittin and phospholipase A2 (PLA2). Due to the lack of antivenom for bee envenomation, this study aimed to produce human monoclonal antibody fragments (single chain fragment variable; scFv), by using phage display technology. These fragments targeted melittin and PLA2, the two major components of bee venom, to minimize their toxic effects in cases of mass envenomation. Two phage antibody selections were performed using purified melittin. As the commercial melittin is contaminated with PLA2, phages specific to PLA2 were also obtained during one of the selections. Specific clones for melittin and PLA2 were selected for the production of soluble scFvs, named here Afribumabs: prefix: afrib- (from Africanized bee); stem/suffix: -umab (fully human antibody). Afribumabs 1 and 2 were tested in in vitro and in vivo assays to assess their ability to inhibit the toxic actions of purified melittin, PLA2, and crude bee venom. Afribumabs reduced hemolysis caused by purified melittin and PLA2 and by crude venom in vitro and reduced edema formation in the paws of mice and prolonged the survival of venom-injected animals in vivo. These results demonstrate that Afribumabs may contribute to the production of the first non-heterologous antivenom treatment against bee envenomation. Such a treatment may overcome some of the difficulties associated with conventional immunotherapy techniques.


Assuntos
Antivenenos/uso terapêutico , Venenos de Abelha/antagonistas & inibidores , Desenho de Fármacos , Mordeduras e Picadas de Insetos/tratamento farmacológico , Proteínas de Insetos/antagonistas & inibidores , Meliteno/antagonistas & inibidores , Anticorpos de Cadeia Única/uso terapêutico , Animais , Antivenenos/genética , Antivenenos/metabolismo , Antivenenos/farmacologia , Venenos de Abelha/química , Venenos de Abelha/enzimologia , Venenos de Abelha/toxicidade , Técnicas de Visualização da Superfície Celular , Células Clonais , Quimioterapia Combinada , Edema/etiologia , Edema/prevenção & controle , Hemólise/efeitos dos fármacos , Humanos , Mordeduras e Picadas de Insetos/fisiopatologia , Proteínas de Insetos/análise , Proteínas de Insetos/toxicidade , Masculino , Meliteno/análise , Meliteno/toxicidade , Camundongos , Inibidores de Fosfolipase A2/farmacologia , Inibidores de Fosfolipase A2/uso terapêutico , Fosfolipases A2 Secretórias/antagonistas & inibidores , Fosfolipases A2 Secretórias/toxicidade , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , Proteínas Recombinantes/uso terapêutico , Anticorpos de Cadeia Única/genética , Anticorpos de Cadeia Única/metabolismo , Anticorpos de Cadeia Única/farmacologia , Tela Subcutânea/efeitos dos fármacos , Análise de Sobrevida
18.
Toxicon ; 110: 56-67, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26704293

RESUMO

Melittin (MEL) is the main constituent and principal toxin of bee venom. It is a small basic peptide, consisting of a known amino acid sequence, with powerful haemolytic activity. Since MEL is a nonspecific cytolytic peptide that attacks lipid membranes thus leading to toxicity, the presumption is that it could have significant therapeutic benefits. The aim was to evaluate the cyto/genotoxic effects of MEL in human peripheral blood lymphocytes (HPBLs) and the molecular mechanisms involved using a multi-biomarker approach. We found that MEL was cytotoxic for HPBLs in a dose- and time-dependent manner. It also induced morphological changes in the cell membrane, granulation and lysis of exposed cells. After treating HPBLs with non-cytotoxic concentrations of MEL, we observed increased DNA damage including oxidative DNA damage as well as increased formation of micronuclei and nuclear buds, and decreased lymphocyte proliferation determined by comet and micronucleus assays. The observed genotoxicity coincided with increased formation of reactive oxygen species, reduction of glutathione level, increased lipid peroxidation and phospholipase C activity, showing the induction of oxidative stress. MEL also modulated the expression of selected genes involved in DNA damage response (TP53, CDKN1A, GADD45α, MDM), oxidative stress (CAT, SOD1, GPX1, GSR and GCLC) and apoptosis (BAX, BCL-2, CAS-3 and CAS-7). Results indicate that MEL is genotoxic to HPBLs and provide evidence that oxidative stress is involved in its DNA damaging effects. MEL toxicity towards normal cells has to be considered if used for potential therapeutic purposes.


Assuntos
Aberrações Cromossômicas/induzido quimicamente , Regulação da Expressão Gênica/efeitos dos fármacos , Linfócitos/efeitos dos fármacos , Meliteno/toxicidade , Mutagênicos/toxicidade , Oxidantes/toxicidade , Estresse Oxidativo , Adulto , Apoptose/efeitos dos fármacos , Biomarcadores/metabolismo , Células Cultivadas , Ensaio Cometa , Dano ao DNA , Glutationa/antagonistas & inibidores , Glutationa/metabolismo , Humanos , Peroxidação de Lipídeos/efeitos dos fármacos , Linfócitos/citologia , Linfócitos/metabolismo , Masculino , Testes para Micronúcleos , Oxirredução , Espécies Reativas de Oxigênio/agonistas , Espécies Reativas de Oxigênio/metabolismo
19.
Xenotransplantation ; 22(4): 295-301, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26031609

RESUMO

Endothelial cell activation and injury by the terminal pathway of complement is important in various pathobiological processes, including xenograft rejection. Protection against injury by human complement can be induced in porcine endothelial cells (ECs) with IL-4 and IL-13 through metabolic activation. However, despite this resistance, the complement-treated ECs were found to lose membrane permeability control assessed with the small molecule calcein. Therefore, to define the apparent discrepancy of permeability changes vis-à-vis the protection from killing, we now investigated whether IL-4 and IL-13 influence the release of the large cytoplasmic protein lactate dehydrogenase (LDH) in ECs incubated with complement or the pore-forming protein melittin. Primary cultures of ECs were pre-treated with IL-4 or IL-13 and then incubated with human serum as source of antibody and complement or melittin. Cell death was assessed using neutral red. Membrane permeability was quantitated measuring LDH release. We found that IL-4-/IL-13-induced protection of ECs from killing by complement or melittin despite loss of LDH in amounts similar to control ECs. However, the cytokine-treated ECs that were protected from killing rapidly regained effective control of membrane permeability. Moreover, the viability of the protected ECs was maintained for at least 2 days. We conclude that the protection induced by IL-4/IL-13 in ECs against lethal attack by complement or melittin is effective and durable despite severe initial impairment of membrane permeability. The metabolic changes responsible for protection allow the cells to repair the membrane injury caused by complement or melittin.


Assuntos
Proteínas do Sistema Complemento/toxicidade , Células Endoteliais/imunologia , Rejeição de Enxerto/imunologia , Rejeição de Enxerto/prevenção & controle , Interleucina-13/administração & dosagem , Interleucina-4/administração & dosagem , Meliteno/toxicidade , Animais , Permeabilidade da Membrana Celular/efeitos dos fármacos , Permeabilidade da Membrana Celular/imunologia , Citoplasma/metabolismo , Citotoxicidade Imunológica , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Humanos , L-Lactato Desidrogenase/metabolismo , Suínos , Transplante Heterólogo/efeitos adversos , Transplante Heterólogo/métodos
20.
Environ Pollut ; 203: 145-152, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25884346

RESUMO

Melittin is a major constituent of the bee venom of Apis mellifera with a broad spectrum of activities. Melittin therapeutical potential is subject to its toxicity and the assessment of ecotoxicity and genotoxicity is of particular interest for therapeutic use. Here we analyzed the biological effects of melittin on two aquatic species, which are representative of two different levels of the aquatic trophic chain: the invertebrate Daphnia magna and the unicellular microalgae Pseudokirchneriella subcapitata. The attention was focused on the determination of: i) ecotoxicity; ii) genotoxicity; iii) antigenotoxicity. Our main finding is that melittin is detrimental to D. magna reproduction and its sub-lethal concentrations create an accumulation dependent on exposition times and a negative effect on DNA. We also observed that melittin significantly delayed time to first eggs. Moreover, results showed that melittin exerted its toxic and genotoxic effects in both species, being a bit more aggressive towards P. subcapitata.


Assuntos
Clorófitas/efeitos dos fármacos , Daphnia/efeitos dos fármacos , Meliteno/toxicidade , Mutagênicos/toxicidade , Animais , Clorófitas/crescimento & desenvolvimento , Dano ao DNA , Daphnia/fisiologia , Feminino , Reprodução/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA