ABSTRACT
Insect venom can cause systemic allergic reactions, including anaphylaxis. Improvements in diagnosis and venom immunotherapy (VIT) are based on a better understanding of an immunological response triggered by venom allergens. Previously, we demonstrated that the recombinant phospholipase A1 (rPoly p 1) from Polybia paulista wasp venom induces specific IgE and IgG antibodies in sensitized mice, which recognized the native allergen. Here, we addressed the T cell immune response of rPoly p 1-sensitized BALB/c mice. Cultures of splenocytes were stimulated with Polybia paulista venom extract and the proliferation of CD8+ and CD4+ T cells and the frequency of T regulatory cells (Tregs) populations were assessed by flow cytometry. Cytokines were quantified in cell culture supernatants in ELISA assays. The in vitro stimulation of T cells from sensitized mice induces a significant proliferation of CD4+ T cells, but not of CD8+ T cells. The cytokine pattern showed a high concentration of IFN-γ and IL-6, and no significant differences to IL-4, IL-1ß and TGF-ß1 production. In addition, the rPoly p 1 group showed a pronounced expansion of CD4+CD25+FoxP3+ and CD4+CD25-FoxP3+ Tregs. rPoly p 1 sensitization induces a Th1/Treg profile in CD4+ T cell subset, suggesting its potential use in wasp venom immunotherapy.
Subject(s)
Allergens/pharmacology , CD4-Positive T-Lymphocytes/drug effects , Desensitization, Immunologic , Insect Proteins/pharmacology , Phospholipases A1/pharmacology , Wasp Venoms/pharmacology , Allergens/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Cytokines/metabolism , Female , Hypersensitivity/immunology , Hypersensitivity/metabolism , Hypersensitivity/therapy , Insect Bites and Stings/immunology , Insect Bites and Stings/metabolism , Insect Bites and Stings/therapy , Insect Proteins/immunology , Lymphocyte Activation/drug effects , Mice, Inbred BALB C , Phospholipases A1/immunology , Wasp Venoms/immunologyABSTRACT
BACKGROUND: The horn fly Haematobia irritans is a blood-sucking ectoparasite responsible for substantial economic loss of livestock. Like other hematophagous arthropods species, the successful blood-feeding of H. irritans is highly dependent on the modulation of the host's hemostasis and immune system. Here, we evaluated the biological activity of hematobin (HTB), a protein recently identified in the H. irritans saliva, on macrophage biology. The goal was to understand the putative interactions between the components of H. irritans saliva and the early host immune responses. RESULTS: Thioglycolate-elicited peritoneal macrophages from BALB/c mice were stimulated by lipopolysaccharide (LPS) plus interferon-γ (IFN-γ) in the presence or absence of recombinant HTB. The presence of the salivary protein in the cultures inhibited nitric oxide production and decreased the inducible nitric oxide synthase (iNOS) expression induced by LPS plus IFN-γ. The tumor necrosis factor-α (TNF-α) and interleukin-12p40 (IL-12p40) levels were also reduced in the macrophages pre-incubated with HTB; these findings correlated to the decreased NF-κB expression. The biological activities described here were not associated with changes in annexin V binding to macrophages suggesting that HTB does not induce cell death. In addition, the activity of HTB seems to be specific to macrophages because no changes were observed in lymphocyte proliferation or cytokine production. CONCLUSIONS: We describe here the first bioactive salivary protein of H. irritans. We characterized its ability to modulate macrophage inflammatory response, and the results can help explain how horn flies modulate the host immune system to feed on blood.
Subject(s)
Diptera/metabolism , Inflammation/metabolism , Insect Proteins/metabolism , Insect Proteins/pharmacology , Macrophages, Peritoneal/drug effects , Amino Acid Sequence , Animals , Cells, Cultured , Cytokines , Dinoprostone , Gene Expression Regulation/drug effects , Lymphocytes/drug effects , Mice , Mice, Inbred BALB C , Nitric Oxide , Nitric Oxide Synthase Type II , Spleen/cytologyABSTRACT
Among venomous animals, Hymenoptera have been suggested as a rich source of natural toxins. Due to their broad ecological diversity, venom from Hymenoptera insects (bees, wasps and ants) have evolved differentially thus widening the types and biological functions of their components. To date, insect toxinology analysis have scarcely uncovered the complex composition of bee, wasp and ant venoms which include low molecular weight compounds, highly abundant peptides and proteins, including several allergens. In Hymenoptera, these complex mixtures of toxins represent a potent arsenal of biological weapons that are used for self-defense, to repel intruders and to capture prey. Consequently, Hymenoptera venom components have a broad range of pharmacological targets and have been extensively studied, as promising sources of new drugs and biopesticides. In addition, the identification and molecular characterization of Hymenoptera venom allergens have allowed for the rational design of component-resolved diagnosis of allergy, finally improving the outcome of venom immunotherapy (VIT). Until recently, a limited number of Hymenoptera venoms had been unveiled due to the technical limitations of the approaches used to date. Nevertheless, the application of novel techniques with high dynamic range has significantly increased the number of identified peptidic and proteinaceous toxins. Considering this, the present review summarizes the current knowledge about the most representative Hymenoptera venom peptides and proteins which are under study for a better understanding of the insect-caused envenoming process and the development of new drugs and biopesticides.
Subject(s)
Arthropod Venoms/chemistry , Arthropod Venoms/toxicity , Hymenoptera/chemistry , Animals , Arthropod Venoms/pharmacology , Insect Proteins/chemistry , Insect Proteins/pharmacology , Insect Proteins/toxicity , Peptides/chemistry , Peptides/pharmacology , Peptides/toxicityABSTRACT
ABSTRACT This study aimed to describe a Bacillus subtilis expression system based on genetically modified B. subtilis. Abaecin, an antimicrobial peptide obtained from Apis mellifera, can enhance the effect of pore-forming peptides from other species on the inhibition of bacterial growth. For the exogenous expression, the abaecin gene was fused with a tobacco etch virus protease cleavage site, a promoter Pglv, and a mature beta-glucanase signal peptide. Also, a B. subtilis expression system was constructed. The recombinant abaecin gene was expressed and purified as a recombinant protein in the culture supernatant. The purified abaecin did not inhibit the growth of Escherichia coli strain K88. Cecropin A and hymenoptaecin exhibited potent bactericidal activities at concentrations of 1 and 1.5 µM. Combinatorial assays revealed that cecropin A and hymenoptaecin had sublethal concentrations of 0.3 and 0.5 µM. This potentiating functional interaction represents a promising therapeutic strategy. It provides an opportunity to address the rising threat of multidrug-resistant pathogens that are recalcitrant to conventional antibiotics.
Subject(s)
Antimicrobial Cationic Peptides/genetics , Antimicrobial Cationic Peptides/metabolism , Bacillus subtilis/genetics , Genetic Vectors/genetics , Insect Proteins/genetics , Insect Proteins/metabolism , Anti-Bacterial Agents/isolation & purification , Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/pharmacology , Antimicrobial Cationic Peptides/isolation & purification , Antimicrobial Cationic Peptides/pharmacology , Bacillus subtilis/metabolism , Escherichia coli/drug effects , Escherichia coli/growth & development , Gene Expression , Genetic Vectors/metabolism , Insect Proteins/isolation & purification , Insect Proteins/pharmacology , Protein Engineering , Protein Transport , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacologyABSTRACT
This study aimed to describe a Bacillus subtilis expression system based on genetically modified B. subtilis. Abaecin, an antimicrobial peptide obtained from Apis mellifera, can enhance the effect of pore-forming peptides from other species on the inhibition of bacterial growth. For the exogenous expression, the abaecin gene was fused with a tobacco etch virus protease cleavage site, a promoter Pglv, and a mature beta-glucanase signal peptide. Also, a B. subtilis expression system was constructed. The recombinant abaecin gene was expressed and purified as a recombinant protein in the culture supernatant. The purified abaecin did not inhibit the growth of Escherichia coli strain K88. Cecropin A and hymenoptaecin exhibited potent bactericidal activities at concentrations of 1 and 1.5µM. Combinatorial assays revealed that cecropin A and hymenoptaecin had sublethal concentrations of 0.3 and 0.5µM. This potentiating functional interaction represents a promising therapeutic strategy. It provides an opportunity to address the rising threat of multidrug-resistant pathogens that are recalcitrant to conventional antibiotics.
Subject(s)
Antimicrobial Cationic Peptides/genetics , Antimicrobial Cationic Peptides/metabolism , Bacillus subtilis/genetics , Genetic Vectors/genetics , Insect Proteins/genetics , Insect Proteins/metabolism , Anti-Bacterial Agents/isolation & purification , Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/pharmacology , Antimicrobial Cationic Peptides/isolation & purification , Antimicrobial Cationic Peptides/pharmacology , Bacillus subtilis/metabolism , Escherichia coli/drug effects , Escherichia coli/growth & development , Gene Expression , Genetic Vectors/metabolism , Insect Proteins/isolation & purification , Insect Proteins/pharmacology , Protein Engineering , Protein Transport , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacologyABSTRACT
BACKGROUND: Antimicrobial peptides (AMPs) are the first line of host immune defense against pathogens. Among AMPs from the honeybee Apis mellifera, abaecin is a major broad-spectrum antibacterial proline-enriched cationic peptide. RESULTS: For heterologous expression of abaecin in Pichia pastoris, we designed an ORF with HisTag, and the codon usage was optimized. The gene was chemically synthetized and cloned in the pUC57 vector. The new ORF was sub-cloned in the pPIC9 expression vector and transformed into P. pastoris. After selection of positive clones, the expression was induced by methanol. The supernatant was analyzed at different times to determine the optimal time for the recombinant peptide expression. As a proof-of-concept, Escherichia coli was co-incubated with the recombinant peptide to verify its antimicrobial potential. DISCUSSION: Briefly, the recombinant Abaecin (rAbaecin) has efficiently decreased E. coli growth (P < 0.05) through an in vitro assay, and may be considered as a novel therapeutic agent that may complement other conventional antibiotic therapies.
Subject(s)
Antimicrobial Cationic Peptides/biosynthesis , Antimicrobial Cationic Peptides/genetics , Gene Expression , Insect Proteins/biosynthesis , Insect Proteins/genetics , Pichia/genetics , Animals , Antimicrobial Cationic Peptides/metabolism , Antimicrobial Cationic Peptides/pharmacology , Bees , Cloning, Molecular , Escherichia coli/drug effects , Escherichia coli/growth & development , Escherichia coli/metabolism , Genetic Engineering/methods , Insect Proteins/metabolism , Insect Proteins/pharmacology , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/pharmacologyABSTRACT
Fibroblasts are the main cellular component of connective tissues and play important roles in health and disease through the production of collagen, fibronectin and growth factors. Under certain conditions, such as wound healing, fibroblasts intensify their metabolic demand, while the restriction of nutrients affect matrix composition, cell metabolism and behavior. In lepidopterans, wound healing is regulated by ecdysteroid hormones, which upregulate multifunctional proteins such as hemolin. However, the role of hemolin in cell proliferation and wound healing is not clear. rLosac is a recombinant hemolin from the caterpillar Lonomia obliqua whose proliferative and cytoprotective effects on endothelial cells have been described. Here, we show that rLosac induces a marked cell survival effect on fibroblast submitted to serum deprivation, which is observable as early as 24h, as demonstrated through the MTT assay, as well as an increase in migration of human dermal fibroblasts (HDF). No effects on cell proliferation or cell cycle distribution of fibroblasts in normal conditions were observed, suggesting that rLosac induces an effect in stressful conditions such serum deprivation but not when nutrient are sufficient. By flow cytometry, rLosac caused an apparent dose-dependent increase in cells in the S phase of the cell cycle and a significant reduction of cells with fragmented DNA. Furthermore, treatment with rLosac results in a significant decrease in the production of reactive oxygen species and in the loss of mitochondrial membrane potential, indicating that a reduction in oxidative stress is involved in rLosac-mediated cytoprotection. Our results also show an up-regulation of Bcl-2 and a down-regulation of Bax protein levels, inhibition of cytochrome c release and a reduction in caspase-3 levels, all considered critical factors for apoptosis. Moreover, rLosac treatment reduces the morphological changes induced by prolonged serum deprivation including the emergence of apoptotic bodies, nucleus fragmentation, cytoplasmic vacuolization and loss of extracellular matrix organization. The wound scratch test assay revealed that rLosac could enhance wound healing in vitro. Altogether, these findings suggest that rLosac strongly induces cellular protection in conditions of stress by serum deprivation preventing damage and loss of mitochondrial function by inhibiting apoptosis. This finding opens a new perspective to further understand the role of hemolin proteins during cellular processes such as wound healing and development.
Subject(s)
Apoptosis/drug effects , Fibroblasts/cytology , Immunoglobulins/pharmacology , Insect Proteins/pharmacology , Animals , Caspase 3/metabolism , Cell Movement/drug effects , Cell Shape/drug effects , Cell Survival/drug effects , Culture Media, Serum-Free/pharmacology , Dermis/cytology , Enzyme Activation/drug effects , Fibroblasts/drug effects , Fibroblasts/ultrastructure , Flow Cytometry , Humans , Membrane Potential, Mitochondrial/drug effects , Peptide Hydrolases/pharmacology , Reactive Oxygen Species/metabolism , bcl-2-Associated X Protein/metabolismABSTRACT
Mycobacterium abscessus subsp. massiliense, a rapidly growing mycobacteria (RGM) that is becoming increasingly important among human infectious diseases, is virulent and pathogenic and presents intrinsic resistance to several antimicrobial drugs that might hamper their elimination. Therefore, the identification of new drugs to improve the current treatment or lower the risk of inducing resistance is urgently needed. Wasp venom primarily comprises peptides that are responsible for most of the biological activities in this poison. Here, a novel peptide Polydim-I, from Polybia dimorpha Neotropical wasp, was explored as an antimycobacterial agent. Polydim-I provoked cell wall disruption and exhibited non-cytotoxicity towards mammalian cells. Polydim-I treatment of macrophages infected with different M. abscessus subsp. massiliense strains reduced 40 to 50% of the bacterial load. Additionally, the Polydim-I treatment of highly susceptible mice intravenously infected with M. abscessus subsp. massiliense induced 0.8 to 1 log reduction of the bacterial load in the lungs, spleen, and liver. In conclusion, this is the first study to show the therapeutic potential of a peptide derived from wasp venom in treating mycobacteria infections. Polydim-I acts on the M. abscessus subsp. massiliense cell wall and reduce 40-90% of the bacterial load both in vitro and in vivo. The presented results encourage further studies on the use of Polydim-I as one of the components for M. abscessus subsp. massiliense treatment.
Subject(s)
Anti-Bacterial Agents/pharmacology , Mycobacterium Infections/drug therapy , Mycobacterium/drug effects , Peptides/pharmacology , Wasps/metabolism , Amino Acid Sequence , Animals , Anti-Bacterial Agents/chemistry , Cell Line , Cells, Cultured , Chromatography, High Pressure Liquid/methods , Female , Host-Pathogen Interactions/drug effects , Insect Proteins/chemistry , Insect Proteins/pharmacology , Interferon-gamma/deficiency , Interferon-gamma/genetics , Macrophages/drug effects , Macrophages/microbiology , Mice, Inbred BALB C , Mice, Knockout , Microbial Sensitivity Tests , Microscopy, Electron, Scanning , Molecular Sequence Data , Mycobacterium/physiology , Mycobacterium/ultrastructure , Mycobacterium Infections/genetics , Mycobacterium Infections/microbiology , Peptides/chemistry , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Wasp Venoms/metabolismABSTRACT
Blood-feeding insects inject potent salivary components including complement inhibitors into their host's skin to acquire a blood meal. Sand fly saliva was shown to inhibit the classical pathway of complement; however, the molecular identity of the inhibitor remains unknown. Here, we identified SALO as the classical pathway complement inhibitor. SALO, an 11 kDa protein, has no homology to proteins of any other organism apart from New World sand flies. rSALO anti-complement activity has the same chromatographic properties as the Lu. longipalpis salivary gland homogenate (SGH)counterparts and anti-rSALO antibodies blocked the classical pathway complement activity of rSALO and SGH. Both rSALO and SGH inhibited C4b deposition and cleavage of C4. rSALO, however, did not inhibit the protease activity of C1s nor the enzymatic activity of factor Xa, uPA, thrombin, kallikrein, trypsin and plasmin. Importantly, rSALO did not inhibit the alternative or the lectin pathway of complement. In conclusion our data shows that SALO is a specific classical pathway complement inhibitor present in the saliva of Lu. longipalpis. Importantly, due to its small size and specificity, SALO may offer a therapeutic alternative for complement classical pathway-mediated pathogenic effects in human diseases.
Subject(s)
Complement Inactivating Agents/pharmacology , Complement Pathway, Classical/drug effects , Insect Proteins/pharmacology , Psychodidae/immunology , Psychodidae/metabolism , Saliva/metabolism , Animals , Chromatography, High Pressure Liquid , Complement Activation/drug effects , Complement C1/antagonists & inhibitors , Complement C1/immunology , Complement C1/metabolism , Complement C4/antagonists & inhibitors , Complement C4/immunology , Complement C4/metabolism , Humans , Recombinant Proteins/pharmacologyABSTRACT
We identified Tf2, the first ß-scorpion toxin from the venom of the Brazilian scorpion Tityus fasciolatus. Tf2 is identical to Tb2-II found in Tityus bahiensis. We found that Tf2 selectively activates human (h)Nav1.3, a neuronal voltage-gated sodium (Nav) subtype implicated in epilepsy and nociception. Tf2 shifts hNav1.3 activation voltage to more negative values, thereby opening the channel at resting membrane potentials. Seven other tested mammalian Nav channels (Nav1.1-1.2; Nav1.4-1.8) expressed in Xenopus oocytes are insensitive upon application of 1 µM Tf2. Therefore, the identification of Tf2 represents a unique addition to the repertoire of animal toxins that can be used to investigate Nav channel function.
Subject(s)
Insect Proteins/pharmacology , Ion Channel Gating/drug effects , NAV1.3 Voltage-Gated Sodium Channel/metabolism , Scorpion Venoms/pharmacology , Scorpions/metabolism , Sodium Channels/metabolism , Amino Acid Sequence , Animals , Humans , Insect Proteins/chemistry , Insect Proteins/isolation & purification , Models, Molecular , Molecular Sequence Data , NAV1.3 Voltage-Gated Sodium Channel/genetics , Oocytes/metabolism , Patch-Clamp Techniques , Protein Structure, Tertiary , Scorpion Venoms/chemistry , Scorpion Venoms/isolation & purification , Scorpion Venoms/metabolism , Sequence Alignment , Sodium Channels/genetics , Xenopus/growth & development , Xenopus/metabolismABSTRACT
BACKGROUND: The caterpillar of the moth Premolis semirufa, commonly named pararama, is found in the Brazilian Amazon region. Accidental contact with the caterpillar bristles causes an intense itching sensation, followed by symptoms of an acute inflammation, which last for three to seven days after the first incident. After multiple accidents a chronic inflammatory reaction, called "Pararamose", characterized by articular synovial membrane thickening with joint deformities common to chronic synovitis, frequently occurs. Although complement mediated inflammation may aid the host defense, inappropriate or excessive activation of the complement system and generation of anaphylatoxins can lead to inflammatory disorder and pathologies. The aim of the present study was to evaluate, in vitro, whether the Premolis semirufa's bristles extract could interfere with the human complement system. RESULTS: The bristles extract was able to inhibit the haemolytic activity of the alternative pathway, as well as the activation of the lectin pathway, but had no effect on the classical pathway, and this inhibition seemed to be caused by activation and consumption of complement components. The extract induced the production of significant amounts of all three anaphylatoxins, C3a, C4a and C5a, promoted direct cleavage of C3, C4 and C5 and induced a significant generation of terminal complement complexes in normal human serum. By using molecular exclusion chromatography, a serine protease of 82 kDa, which activates complement, was isolated from P. semirufa bristles extract. The protease, named here as Ps82, reduced the haemolytic activity of the alternative and classical pathways and inhibited the lectin pathway. In addition, Ps82 induced the cleavage of C3, C4 and C5 and the generation of C3a and C4a in normal human serum and it was capable to cleave human purified C5 and generate C5a. The use of Phenanthroline, metalloprotease inhibitor, in the reactions did not significantly interfere with the activity of the Ps82, whereas the presence of PMSF, serine protease inhibitor, totally blocked the activity. CONCLUSION: These data show that a serine protease present in the Premolis semirufa's bristles extract has the ability to activate the complement system, which may contribute to the inflammatory process presented in humans after envenomation.
Subject(s)
Complement Activation/drug effects , Insect Proteins/pharmacology , Moths/enzymology , Serine Proteases/pharmacology , Anaphylatoxins/chemistry , Anaphylatoxins/isolation & purification , Animals , Complement Membrane Attack Complex/chemistry , Erythrocytes/drug effects , Humans , Insect Proteins/isolation & purification , Proteolysis , Serine Proteases/isolation & purificationABSTRACT
Aegyptin is a mosquito salivary gland protein and potent inhibitor of platelet aggregation. Aegyptin binds to the von Willebrand factor-binding site on collagen and prevents its interaction with platelets. Because collagen also induces plasma clotting by activation of factor XII, we evaluated the effects of aegyptin on collagen-induced coagulation activation and how it interferes with thrombosis in three different in vivo models. Our results demonstrate that aegyptin abolishes collagen-induced clot formation and thrombin generation in platelet-free plasma. Aegyptin has no antithrombotic activity in the arteriovenous shunt model (collagen-independent) but it prevents laser-induced collagen-mediated thrombus formation in rats. Furthermore, aegyptin protects mice from collagen and epinephrine-induced thromboembolism. Therefore, aegyptin has a dual antithrombotic mechanism: inhibition of platelet-collagen interaction and collagen's pro-coagulant activity. This is the first description of a collagen-binding protein that also inhibits collagen-mediated coagulant activity.
Subject(s)
Blood Coagulation/drug effects , Collagen/pharmacology , Insect Proteins/pharmacology , Pulmonary Embolism/prevention & control , Salivary Proteins and Peptides/pharmacology , Animals , Culicidae/metabolism , Dose-Response Relationship, Drug , Epinephrine/pharmacology , Female , HEK293 Cells , Humans , Insect Proteins/genetics , Male , Mice , Mice, Inbred BALB C , Rats , Rats, Wistar , Recombinant Proteins/pharmacology , Salivary Proteins and Peptides/genetics , Thrombin/metabolismABSTRACT
Phα1ß toxin is a peptide purified from the venom of the armed spider Phoneutria nigriventer, with markedly antinociceptive action in models of acute and persistent pain in rats. Similarly to ziconotide, its analgesic action is related to inhibition of high voltage activated calcium channels with more selectivity for N-type. In this study we evaluated the effect of Phα1ß when injected peripherally or intrathecally in a rat model of spontaneous pain induced by capsaicin. We also investigated the effect of Phα1ß on Ca²âº transients in cultured dorsal root ganglia (DRG) neurons and HEK293 cells expressing the TRPV1 receptor. Intraplantar or intrathecal administered Phα1ß reduced both nocifensive behavior and mechanical hypersensitivity induced by capsaicin similarly to that observed with SB366791, a specific TRPV1 antagonist. Peripheral nifedipine and mibefradil did also decrease nociceptive behavior induced by intraplantar capsaicin. In contrast, ω-conotoxin MVIIA (a selective N-type Ca²âº channel blocker) was effective only when administered intrathecally. Phα1ß, MVIIA and SB366791 inhibited, with similar potency, the capsaicin-induced Ca²âº transients in DRG neurons. The simultaneous administration of Phα1ß and SB366791 inhibited the capsaicin-induced Ca²âº transients that were additive suggesting that they act through different targets. Moreover, Phα1ß did not inhibit capsaicin-activated currents in patch-clamp recordings of HEK293 cells that expressed TRPV1 receptors. Our results show that Phα1ß may be effective as a therapeutic strategy for pain and this effect is not related to the inhibition of TRPV1 receptors.
Subject(s)
Analgesics, Non-Narcotic/therapeutic use , Disease Models, Animal , Ganglia, Spinal/drug effects , Membrane Transport Modulators/therapeutic use , Neuralgia/drug therapy , Neurons/drug effects , Spider Venoms/therapeutic use , Analgesics, Non-Narcotic/pharmacology , Animals , Behavior, Animal/drug effects , Calcium Signaling/drug effects , Capsaicin , Cells, Cultured , Ganglia, Spinal/cytology , Ganglia, Spinal/metabolism , Ganglia, Spinal/pathology , HEK293 Cells , Humans , Insect Proteins/pharmacology , Insect Proteins/therapeutic use , Male , Membrane Transport Modulators/pharmacology , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neuralgia/metabolism , Neuralgia/pathology , Neurons/cytology , Neurons/metabolism , Neurons/pathology , Peptides/pharmacology , Peptides/therapeutic use , Rats , Rats, Wistar , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Spider Venoms/pharmacology , TRPV Cation Channels/antagonists & inhibitors , TRPV Cation Channels/genetics , TRPV Cation Channels/metabolismABSTRACT
Peptides isolated from animal venoms have shown the ability to regulate pancreatic beta cell function. Characterization of wasp venoms is important, since some components of these venoms present large molecular variability, and potential interactions with different signal transduction pathways. For example, the well studied mastoparan peptides interact with a diversity of cell types and cellular components and stimulate insulin secretion via the inhibition of ATP dependent K(+) (K(ATP)) channels, increasing intracellular Ca(2+) concentration. In this study, the insulin secretion of isolated pancreatic islets from adult Swiss mice was evaluated in the presence of synthetic Agelaia MP-I (AMP-I) peptide, and some mechanisms of action of this peptide on endocrine pancreatic function were characterized. AMP-I was manually synthesized using the Fmoc strategy, purified by RP-HPLC and analyzed using ESI-IT-TOF mass spectrometry. Isolated islets were incubated at increasing glucose concentrations (2.8, 11.1 and 22.2 mM) without (Control group: CTL) or with 10 µM AMP-I (AMP-I group). AMP-I increased insulin release at all tested glucose concentrations, when compared with CTL (P < 0.05). Since molecular analysis showed a potential role of the peptide interaction with ionic channels, insulin secretion was also analyzed in the presence of 250 µM diazoxide, a K(ATP) channel opener and 10 µM nifedipine, a Ca(2+) channel blocker. These drugs abolished insulin secretion in the CTL group in the presence of 2.8 and 11.1 mM glucose, whereas AMP-I also enhanced insulin secretory capacity, under these glucose conditions, when incubated with diazoxide and nifedipine. In conclusion, AMP-I increased beta cell secretion without interfering in K(ATP) and L-type Ca(2+) channel function, suggesting a different mechanism for this peptide, possibly by G protein interaction, due to the structural similarity of this peptide with Mastoparan-X, as obtained by modeling.
Subject(s)
Hypoglycemic Agents/pharmacology , Insect Proteins/pharmacology , Insulin/metabolism , Islets of Langerhans/drug effects , Peptides/pharmacology , Wasp Venoms/chemistry , Animals , Calcium/metabolism , Cells, Cultured , Chromatography, High Pressure Liquid , Hypoglycemic Agents/chemical synthesis , Insect Proteins/chemical synthesis , Insect Proteins/metabolism , Insulin Secretion , Islets of Langerhans/metabolism , KATP Channels/drug effects , Male , Mice , Peptides/chemical synthesis , Spectrometry, Mass, Electrospray Ionization , Stereoisomerism , Wasp Venoms/chemical synthesis , Wasp Venoms/pharmacology , WaspsABSTRACT
Several studies have pointed out the immunomodulatory properties of the Salivary Gland Extract (SGE) from Lutzomyia longipalpis. We aimed to identify the SGE component (s) responsible for its effect on ovalbumin (OVA)-induced neutrophil migration (NM) and to evaluate the effect of SGE and components in the antigen-induced arthritis (AIA) model. We tested the anti-arthritic activities of SGE and the recombinant LJM111 salivary protein (rLJM111) by measuring the mechanical hypernociception and the NM into synovial cavity. Furthermore, we measured IL-17, TNF-α and IFN-γ released by lymph nodes cells stimulated with mBSA or anti-CD3 using enzyme-linked immunosorbent assay (ELISA). Additionally, we tested the effect of SGE and rLJM111 on co-stimulatory molecules expression (MHC-II and CD-86) by flow cytometry, TNF-α and IL-10 production (ELISA) of bone marrow-derived dendritic cells (BMDCs) stimulated with LPS, chemotaxis and actin polymerization from neutrophils. Besides, the effect of SGE on CXCR2 and GRK-2 expression on neutrophils was investigated. We identified one plasmid expressing the protein LJM111 that prevented NM in OVA-challenged immunized mice. Furthermore, both SGE and rLJM111 inhibited NM and pain sensitivity in AIA and reduced IL-17, TNF-α and IFN-γ. SGE and rLJM111 also reduced MHC-II and CD-86 expression and TNF-α whereas increased IL-10 release by LPS-stimulated BMDCs. SGE, but not LJM 111, inhibited neutrophils chemotaxis and actin polymerization. Additionally, SGE reduced neutrophil CXCR2 expression and increased GRK-2. Thus, rLJM111 is partially responsible for SGE mechanisms by diminishing DC function and maturation but not chemoattraction of neutrophils.
Subject(s)
Anti-Inflammatory Agents/pharmacology , Arthritis, Experimental/immunology , Insect Proteins/pharmacology , Psychodidae , Salivary Glands/immunology , Salivary Proteins and Peptides/pharmacology , Animals , Cell Movement , Cytokines/immunology , Dendritic Cells/immunology , Female , G-Protein-Coupled Receptor Kinase 2/immunology , Lymph Nodes/cytology , Male , Mice , Mice, Inbred BALB C , Neutrophils/immunology , Ovalbumin/immunology , Receptors, Interleukin-8B/immunology , Recombinant Proteins/pharmacology , Serum Albumin, Bovine/immunologyABSTRACT
The control of viral infections, mainly those caused by influenza viruses, is of great interest in Public Health. Several studies have shown the presence of active properties in the hemolymph of arthropods, some of which are of interest for the development of new pharmacological drugs. Recently, we have demonstrated the existence of a potent antiviral property in the hemolymph of Lonomia obliqua caterpillars. The aim of this study was to produce an antiviral protein in a baculovirus/Sf9 cell system. The resulting bacmid contains the sequence coding for the antiviral protein previously described by our group. Total RNA from L. obliqua caterpillars was extracted with Trizol and used in the reverse transcription assay with oligo(d)T primer followed by polymerase chain reactions (RT-PCR) with specific primers for the cDNA coding for the antiviral protein, based on the sequence deposited in the GenBank database. Restriction sites were inserted in the cDNA for ligation in the donor plasmid pFastBac1™. The recombinant plasmid was selected in Escherichia coli DH5α and subsequently used in the transformation of E. coli DH10Bac for the construction of the recombinant bacmid. This bacmid was used for the expression of the antiviral protein in the baculovirus/Sf9 cell system. After identifying the protein by western blot, activity tests were performed, showing that the purified recombinant protein was able to significantly reduce viral replication (about 4 logs). Studies on the optimization of the expression system for the production of this antiviral protein in insect cells are in progress.
Subject(s)
Antiviral Agents/pharmacology , Biological Products/pharmacology , Hemolymph/chemistry , Hemolymph/immunology , Insect Proteins/biosynthesis , Insect Proteins/pharmacology , Lepidoptera/immunology , Animals , Antiviral Agents/isolation & purification , Baculoviridae/genetics , Biological Products/isolation & purification , Cell Line , Cloning, Molecular , Escherichia coli/genetics , Gene Expression , Genetic Vectors , Insect Proteins/genetics , Lepidoptera/genetics , Recombinant Proteins/genetics , Recombinant Proteins/pharmacologyABSTRACT
Experiments were designed to determine if the vasodilatory peptides maxadilan and pituitary adenylate cyclase-activating peptide (PACAP-38) may cause plasma leakage through activation of leukocytes and to what extent these effects could be due to PAC1 and CXCR1/2 receptor stimulation. Intravital microscopy of hamster cheek pouches utilizing FITC-dextran and rhodamine, respectively, as plasma and leukocyte markers was used to measure arteriolar diameter, plasma leakage and leukocyte accumulation in a selected area (5mm(2)) representative of the hamster cheek pouch microcirculation. Our studies showed that the sand fly vasodilator maxadilan and PACAP-38 induced arteriolar dilation, leukocyte accumulation and plasma leakage in postcapillary venules. The recombinant mutant of maxadilan M65 and an antagonist of CXCR1/2 receptors, reparixin, and an inhibitor of CD11b/CD18 up-regulation, ropivacaine, inhibited all these effects as induced by maxadilan. Dextran sulfate, a complement inhibitor with heparin-like anti-inflammatory effects, inhibited plasma leakage and leukocyte accumulation but not arteriolar dilation as induced by maxadilan and PACAP-38. In vitro studies with isolated human neutrophils showed that maxadilan is a potent stimulator of neutrophil migration comparable with fMLP and leukotriene B(4) and that M65 and reparixin inhibited such migration. The data suggest that leukocyte accumulation and plasma leakage induced by maxadilan involves a mechanism related to PAC1- and CXCR1/2-receptors on leukocytes and endothelial cells.
Subject(s)
Capillary Permeability/drug effects , Cheek/blood supply , Insect Proteins/pharmacology , Psychodidae , Receptors, Interleukin-8A/drug effects , Receptors, Interleukin-8B/drug effects , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/drug effects , Signal Transduction/drug effects , Vasodilation/drug effects , Vasodilator Agents/pharmacology , Animals , Cells, Cultured , Chemotaxis, Leukocyte/drug effects , Cricetinae , Dextrans/metabolism , Dose-Response Relationship, Drug , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Fluorescein-5-isothiocyanate/analogs & derivatives , Fluorescein-5-isothiocyanate/metabolism , Fluorescent Dyes/metabolism , Humans , Insect Proteins/genetics , Insect Proteins/isolation & purification , Microscopy, Fluorescence , Microscopy, Video , Mutation , Neutrophils/drug effects , Neutrophils/metabolism , Pituitary Adenylate Cyclase-Activating Polypeptide/pharmacology , Psychodidae/chemistry , Receptors, Interleukin-8A/metabolism , Receptors, Interleukin-8B/metabolism , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Recombinant Proteins/pharmacology , Rhodamines/metabolism , Time Factors , Vasodilator Agents/isolation & purification , Venules/drug effects , Venules/metabolismABSTRACT
Ticks are blood-feeding arthropods that secrete anticoagulant molecules to maintain the fluidity of the blood during its feeding. Tick saliva has many compounds with biological activities that interact directly with host systems, such as blood clotting, platelet aggregation, cell death, among others. Some reports show that there are proteins with anticancer properties in tick saliva. This paper reports some of the biological roles of the Amblyomma cajennense tick saliva, including Factor Xa and thrombin inhibition, action on platelet aggregation, and also preliminary cytotoxic effects on tumor cell lines. The crude saliva was tested in the coagulation, fibrinolysis and platelet aggregation systems. The protein profile of the crude saliva was examined through anion exchange chromatography performed in a FPLC system. The chromatography separated seven protein fractions (Pools I to VII), which biological activities were evaluated. Moreover, the cytotoxic effects of the crude saliva were evaluated on SK-MEL-28 (melanoma cells) and MIA PaCa-2 (pancreas adenocarcinoma cells) using the MTT assay, flow cytometry and fluorescence microscopy. The crude saliva was able to induce cell death on both cancer cells lines, and, interestingly, the cytotoxic effects were not observed on human fibroblasts, which were used as control. The present work opens perspectives for the characterization and development of new molecules involved in the hemostatic system and in cancer control.
Subject(s)
Anticoagulants/pharmacology , Antineoplastic Agents/pharmacology , Drug Discovery , Hemostasis/drug effects , Ixodidae/metabolism , Neoplasms/drug therapy , Saliva/metabolism , Adenocarcinoma/drug therapy , Animals , Anticoagulants/adverse effects , Anticoagulants/isolation & purification , Antineoplastic Agents/adverse effects , Antineoplastic Agents/isolation & purification , Antithrombins/adverse effects , Antithrombins/isolation & purification , Antithrombins/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Factor Xa Inhibitors , Female , Humans , Insect Proteins/adverse effects , Insect Proteins/isolation & purification , Insect Proteins/pharmacology , Male , Melanoma/drug therapy , Neoplasms/pathology , Pancreatic Neoplasms/drug therapy , Platelet Aggregation Inhibitors/adverse effects , Platelet Aggregation Inhibitors/isolation & purification , Platelet Aggregation Inhibitors/pharmacologyABSTRACT
Digestive endoprotease activities of the coconut palm weevil, Homalinotus coriaceus (Coleoptera: Curculionidae), were characterized based on the ability of gut extracts to hydrolyze specific synthetic substrates, optimal pH, and hydrolysis sensitivity to protease inhibitors. Trypsin-like proteinases were major enzymes for H. coriaceus, with minor activity by chymotrypsin proteinases. More importantly, gut proteinases of H. coriaceus were inhibited by trypsin inhibitor from Inga laurina seeds. In addition, a serine proteinase inhibitor from I. laurina seeds demonstrated significant reduction of growth of H. coriaceus larvae after feeding on inhibitor incorporated artificial diets. Dietary utilization experiments show that 0.05% I. laurina trypsin inhibitor, incorporated into an artificial diet, decreases the consumption rate and fecal production of H. coriaceus larvae. Dietary utilization experiments show that 0.05% I. laurina trypsin inhibitor, incorporated into an artificial diet, decreases the consumption rate and fecal production of H. coriaceus larvae. We have constructed a three-dimensional model of the trypsin inhibitor complexed with trypsin. The model was built based on its comparative homology with soybean trypsin inhibitor. Trypsin inhibitor of I. laurina shows structural features characteristic of the Kunitz type trypsin inhibitor. In summary, these findings contribute to the development of biotechnological tools such as transgenic plants with enhanced resistance to insect pests.
Subject(s)
Fabaceae , Insect Proteins/physiology , Peptide Hydrolases/physiology , Peptides/pharmacology , Plant Proteins/pharmacology , Weevils/enzymology , Amino Acid Sequence , Animals , Digestive System/enzymology , Insect Proteins/isolation & purification , Insect Proteins/pharmacology , Larva/drug effects , Larva/growth & development , Models, Molecular , Molecular Sequence Data , Protease Inhibitors/pharmacology , Protein Structure, Tertiary , Sequence Alignment , Structural Homology, Protein , Weevils/drug effects , Weevils/growth & developmentABSTRACT
Envenoming by the contact of human skin with Lonomia obliqua caterpillars promotes a hemorrhagic syndrome characterized by a consumptive coagulopathy. Losac (Lonomia obliqua Stuart factor activator) is a component of the bristle of L. obliqua that is probably partially responsible for the observed syndrome because it activates factor X and is recognized by an effective antilonomic serum. Here we unveil the proteolytic activity of Losac and demonstrate the feasibility of its recombinant production. On the other hand, Losac has no homology to known proteases, but it can be inhibited by PMSF, a serine protease inhibitor. Instead, it shows closer homology to members of the hemolin family of proteins, a group of cell adhesion molecules. The recombinant protein (rLosac) shortened the coagulation time of normal and deficient plasmas, whereas it was ineffective in factor X-deficient plasma unless reconstituted with this protein. rLosac was able to activate factor X in a dose- and time-dependent manner but not γ-carboxyglutamic acid domainless factor X. Moreover, phospholipids and calcium ions increased rLosac activity. Also, rLosac had no effect on fibrin or fibrinogen, indicating its specificity for blood coagulation activation. Linear double reciprocal plots indicate that rLosac follows a Michaelis-Menten kinetics. Cleavage of factor X by rLosac resulted in fragments that are compatible with those generated by RVV-X (a well known factor X activator). Together, our results validate Losac as the first protein from the hemolin family exhibiting procoagulant activity through selective proteolysis on coagulation factor X.