Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
Add more filters










Publication year range
1.
J Pept Sci ; 25(5): e3172, 2019 May.
Article in English | MEDLINE | ID: mdl-31006945

ABSTRACT

Venoms have recently emerged as a promising field in drug discovery due to their good selectivity and affinity for a wide range of biological targets. Among their multiple potential applications, venoms are a rich source of blood-brain barrier (BBB) peptide shuttles. We previously described a short nontoxic derivative of apamin, MiniAp-4, which can transport a wide range of cargoes across the BBB. Here, we have studied the conformation of the proline residue of a range of MiniAp-4 analogues by high-field NMR techniques, with the aim to identify whether there is a direct relation between the cis/trans population and a range of features, such as the capacity to transport molecules across a human-based cellular model and stability in various media. The most promising candidate showed improved transport properties for a relevant small fluorophore.


Subject(s)
Apamin/metabolism , Blood-Brain Barrier/metabolism , Proline/metabolism , Apamin/chemistry , Apamin/isolation & purification , Biological Transport , Blood-Brain Barrier/chemistry , Cells, Cultured , Humans , Nuclear Magnetic Resonance, Biomolecular , Proline/chemistry , Protein Conformation , Stereoisomerism
2.
J Steroid Biochem Mol Biol ; 168: 118-126, 2017 04.
Article in English | MEDLINE | ID: mdl-28223151

ABSTRACT

Testosterone has endothelium-dependent vasodilatory effects on the coronary artery, with some reports suggesting endothelial ion channel involvement. This study employed the whole-cell patch clamp technique to investigate the effect of testosterone on ion channels in human coronary artery endothelial cells (HCAECs) and the mechanisms involved. We found that 0.03-3µM testosterone significantly induced a rapid, concentration-dependent increase in total HCAEC current (EC50, 71.96±1.66nM; maximum increase, 59.13±8.37%; mean±SEM). The testosterone-enhanced currents consisted of small- and large-conductance Ca2+-activated K+ currents (SKCa and BKCa currents), but not Cl- and nonselective cation currents. Either a non-permeant testosterone conjugate or the non-aromatizable androgen dihydrotestosterone (DHT) could increase HCAEC currents as well. The androgen receptor antagonist flutamide prevented this testosterone, testosterone conjugate, and DHT effect, while the estrogen receptor antagonist fulvestrant did not. Incubating HCAECs with pertussis toxin or protein kinase A inhibitor H-89 largely inhibited the testosterone effect, while pre-incubation with phospholipase C inhibitor U-73122, prostacyclin inhibitor indomethacin, nitric oxide synthase inhibitor L-NAME or cytochrome P450 inhibitor MS-PPOH, did not. Finally, testosterone application induced HCAEC hyperpolarization within minutes; this effect was prevented by SKCa and BKCa current inhibitors apamin and iberiotoxin. This is the first electrophysiological demonstration of androgen-induced KCa current increase, leading to hyperpolarization, in any endothelial cell, and the first report of SKCa as a testosterone target. Our data show that testosterone rapidly increased whole-cell HCAEC SKCa and BKCa currents via a surface androgen receptor, Gi/o protein, and protein kinase A. This mechanism may explain rapid testosterone-induced coronary vasodilation seen in vivo.


Subject(s)
Coronary Vessels/cytology , Endothelial Cells/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Potassium Channels, Calcium-Activated/metabolism , Testosterone/blood , Androgens/chemistry , Apamin/chemistry , Cell Line , Cyclic AMP-Dependent Protein Kinases/metabolism , Endothelial Cells/drug effects , Epoprostenol/antagonists & inhibitors , Estrenes/chemistry , Humans , Indomethacin/chemistry , NG-Nitroarginine Methyl Ester/chemistry , Nitric Oxide Synthase/chemistry , Pyrrolidinones/chemistry , Receptors, Androgen/metabolism , Signal Transduction , Testosterone/chemistry , Vasodilation
3.
J Microbiol ; 54(12): 853-866, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27888461

ABSTRACT

Bee venom (BV) from honey bee (Apis Melifera L.) contains at least 18 pharmacologically active components including melittin (MLT), phospholipase A2 (PLA2), and apamin etc. BV is safe for human treatments dose dependently and proven to possess different healing properties including antibacterial and antiparasitidal properties. Nevertheless, antiviral properties of BV have not well investigated. Hence, we identified the potential antiviral properties of BV and its component against a broad panel of viruses. Co-incubation of non-cytotoxic amounts of BV and MLT, the main component of BV, significantly inhibited the replication of enveloped viruses such as Influenza A virus (PR8), Vesicular Stomatitis Virus (VSV), Respiratory Syncytial Virus (RSV), and Herpes Simplex Virus (HSV). Additionally, BV and MLT also inhibited the replication of non-enveloped viruses such as Enterovirus-71 (EV-71) and Coxsackie Virus (H3). Such antiviral properties were mainly explained by virucidal mechanism. Moreover, MLT protected mice which were challenged with lethal doses of pathogenic influenza A H1N1 viruses. Therefore, these results provides the evidence that BV and MLT could be a potential source as a promising antiviral agent, especially to develop as a broad spectrum antiviral agent.


Subject(s)
Antiviral Agents/pharmacology , Bee Venoms/pharmacology , Orthomyxoviridae Infections/drug therapy , Viruses/drug effects , Animals , Antiviral Agents/administration & dosage , Antiviral Agents/chemistry , Apamin/chemistry , Bee Venoms/administration & dosage , Bee Venoms/chemistry , Cell Line , Enterovirus/drug effects , Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H1N1 Subtype/physiology , Influenza A virus/drug effects , Melitten/pharmacology , Mice , Orthomyxoviridae Infections/virology , Simplexvirus/drug effects , Virus Replication/drug effects
4.
Angew Chem Int Ed Engl ; 55(2): 572-5, 2016 Jan 11.
Article in English | MEDLINE | ID: mdl-26492861

ABSTRACT

Drug delivery across the blood-brain barrier (BBB) is a formidable challenge for therapies targeting the central nervous system. Although BBB shuttle peptides enhance transport into the brain non-invasively, their application is partly limited by lability to proteases. The present study proposes the use of cyclic peptides derived from venoms as an affordable way to circumvent this drawback. Apamin, a neurotoxin from bee venom, was minimized by reducing its complexity, toxicity, and immunogenicity, while preserving brain targeting, active transport, and protease resistance. Among the analogues designed, the monocyclic lactam-bridged peptidomimetic MiniAp-4 was the most permeable. This molecule is capable of translocating proteins and nanoparticles in a human-cell-based BBB model. Furthermore, MiniAp-4 can efficiently deliver a cargo across the BBB into the brain parenchyma of mice.


Subject(s)
Apamin/chemistry , Peptidomimetics/administration & dosage , Venoms/administration & dosage , Amino Acid Sequence , Blood-Brain Barrier , Humans , Models, Biological , Molecular Sequence Data , Peptidomimetics/chemistry , Peptidomimetics/pharmacokinetics
5.
Angew Chem Int Ed Engl ; 54(17): 5157-60, 2015 Apr 20.
Article in English | MEDLINE | ID: mdl-25727886

ABSTRACT

Inspired by the knowledge that most antibodies recognize a conformational epitope because of the epitope's specific three-dimensional shape rather than its linear structure, we combined scaffold-based peptide design and surface molecular imprinting to fabricate a novel nanocarrier harboring stable binding sites that captures a membrane protein. In this study, a disulfide-linked α-helix-containing peptide, apamin, was used to mimic the extracellular, structured N-terminal part of the protein p32 and then serve as an imprinting template for generating a sub-40 nm-sized polymeric nanoparticle that potently binds to the target protein, recognizes p32-positive tumor cells, and successfully mediates targeted photodynamic therapy in vivo. This could provide a promising alternative for currently used peptide-modified nanocarriers and may have a broad impact on the development of polymeric nanoparticle-based therapies for a wide range of human diseases.


Subject(s)
Epitopes/chemistry , Molecular Imprinting , Nanoparticles/chemistry , Oligopeptides/chemistry , Amino Acid Sequence , Animals , Apamin/chemistry , Apamin/metabolism , Cell Line, Tumor , Disease Models, Animal , Drug Carriers/chemistry , Drug Carriers/metabolism , Epitopes/administration & dosage , Humans , Mice, Nude , Molecular Sequence Data , Nanoparticles/administration & dosage , Nanoparticles/metabolism , Neoplasms/drug therapy , Oligopeptides/administration & dosage , Particle Size , Photochemotherapy , Protein Structure, Secondary , Transplantation, Heterologous
6.
Bioorg Med Chem Lett ; 24(24): 5616-5620, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25466186

ABSTRACT

An asparagine or a histidine are present in a similar position in the outer pore region of SK2 and SK3 channels, respectively. Therefore, this structural difference was targeted in order to develop selective blockers of SK channel subtypes. Following docking investigations, based on theoretical models of truncated SK2 and SK3 channels, the benzyl side chain of N-methyl-laudanosine (NML) was functionalized in order to target this specific amino-acid residues. Chiral butanamide and benzyloxy analogues were prepared, resolved and tested for their affinity for SK2 and SK3 channels. Isoquinolinium (NMIQ) derivatives have a higher affinity for both SK channel subtypes than the corresponding derivative with no functionalized side chain. This trend was observed also for the 1,2,3,4-tetrahydroisoquinoline (THIQ) analogues. A benzyloxy functionalized NML enantiomer has a higher affinity than NML stereoisomers. Otherwise, the conserved affinity of these analogues led to the opportunity to further investigate in terms of possible labeling for in vivo investigations of the role of SK channels.


Subject(s)
Isoquinolines/chemistry , Small-Conductance Calcium-Activated Potassium Channels/chemistry , Amino Acid Sequence , Apamin/chemistry , Binding Sites , Humans , Isoquinolines/metabolism , Molecular Docking Simulation , Molecular Sequence Data , Nitrogen/chemistry , Protein Binding , Protein Structure, Tertiary , Small-Conductance Calcium-Activated Potassium Channels/metabolism , Stereoisomerism , Tetrahydroisoquinolines/chemistry
7.
Mol Pharm ; 11(9): 3210-22, 2014 Sep 02.
Article in English | MEDLINE | ID: mdl-25098949

ABSTRACT

Faced with the complex medical challenge presented by spinal cord injuries (SCI) and considering the lack of any available curative therapy, the development of a novel method of delivering existing drugs or candidate agents can be perceived to be as important as the development of new therapeutic molecules. By combining three ingredients currently in clinical use or undergoing testing, we have designed a central nervous system targeted delivery system based on apamin-modified polymeric micelles (APM). Apamin, one of the major components of honey bee venom, serves as the targeting moiety, poly(ethylene glycol) (PEG) distearoylphosphatidylethanolamine (DSPE) serves as the drug-loaded material, and curcumin is used as the therapeutic agent. Apamin was conjugated with NHS (N-hydroxysuccinimide)-PEG-DSPE in a site-specific manner, and APM were prepared by a thin-film hydration method. A formulation comprising 0.5 mol % targeting ligand with 50 nm particle size showed strong targeting efficiency in vivo and was evaluated in pharmacodynamic assays. A 7-day treatment by daily intravenous administration of low doses of APM (corresponding to 5 mg/kg of curcumin) was performed. Significantly enhanced recovery and prolonged survival was found in the SCI mouse model, as compared to sham-treated groups, with no apparent toxicity. A single dose of apamin-conjugated polymers was about 700-fold lower than the LD50 amount, suggesting that APM and apamin have potential for clinical applications as spinal cord targeting ligand for delivery of agents in treatment of diseases of the central nervous system.


Subject(s)
Apamin/pharmacology , Spinal Cord Injuries/drug therapy , Animals , Apamin/chemistry , Chemistry, Pharmaceutical/methods , Curcumin/chemistry , Drug Carriers/chemistry , Drug Delivery Systems/methods , Mice , Micelles , Particle Size , Phosphatidylethanolamines/chemistry , Polyethylene Glycols/chemistry , Polymers/chemistry , Succinimides/chemistry
8.
PLoS One ; 9(5): e96691, 2014.
Article in English | MEDLINE | ID: mdl-24798465

ABSTRACT

BACKGROUND: Apamin is commonly used as a small-conductance Ca2+-activated K+ (SK) current inhibitor. However, the specificity of apamin in cardiac tissues remains unclear. OBJECTIVE: To test the hypothesis that apamin does not inhibit any major cardiac ion currents. METHODS: We studied human embryonic kidney (HEK) 293 cells that expressed human voltage-gated Na+, K+ and Ca2+ currents and isolated rabbit ventricular myocytes. Whole-cell patch clamp techniques were used to determine ionic current densities before and after apamin administration. RESULTS: Ca2+ currents (CACNA1c+CACNB2b) were not affected by apamin (500 nM) (data are presented as median [25th percentile;75th percentile] (from -16 [-20;-10] to -17 [-19;-13] pA/pF, P = NS), but were reduced by nifedipine to -1.6 [-3.2;-1.3] pA/pF (p = 0.008). Na+ currents (SCN5A) were not affected by apamin (from -261 [-282;-145] to -268 [-379;-132] pA/pF, P = NS), but were reduced by flecainide to -57 [-70;-47] pA/pF (p = 0.018). None of the major K+ currents (IKs, IKr, IK1 and Ito) were inhibited by 500 nM of apamin (KCNQ1+KCNE1, from 28 [20]; [37] to 23 [18]; [32] pA/pF; KCNH2+KCNE2, from 28 [24]; [30] to 27 [24]; [29] pA/pF; KCNJ2, from -46 [-48;-40] to -46 [-51;-35] pA/pF; KCND3, from 608 [505;748] to 606 [454;684]). Apamin did not inhibit the INa or ICaL in isolated rabbit ventricular myocytes (INa, from -67 [-75;-59] to -68 [-71;-59] pA/pF; ICaL, from -16 [-17;-14] to -14 [-15;-13] pA/pF, P = NS for both). CONCLUSIONS: Apamin does not inhibit human cardiac Na+ currents, L-type Ca2+ currents or other major K+ currents. These findings indicate that apamin is a specific SK current inhibitor in hearts as well as in other organs.


Subject(s)
Apamin/chemistry , Calcium Channels, L-Type/metabolism , Myocardium/metabolism , Myocytes, Cardiac/cytology , Potassium Channels, Voltage-Gated/metabolism , Voltage-Gated Sodium Channels/metabolism , Animals , Apamin/pharmacology , Calcium/metabolism , Calcium Channel Blockers/chemistry , Calcium Channel Blockers/pharmacology , HEK293 Cells , Heart/drug effects , Heart Ventricles/cytology , Humans , Ion Transport , Patch-Clamp Techniques , Potassium Channels, Voltage-Gated/antagonists & inhibitors , Rabbits , Voltage-Gated Sodium Channel Blockers/chemistry , Voltage-Gated Sodium Channel Blockers/pharmacology
9.
Chembiochem ; 12(12): 1808-12, 2011 Aug 16.
Article in English | MEDLINE | ID: mdl-21726033

ABSTRACT

Ion-channel function can be modified in various ways. For example, numerous studies have shown that currents through voltage-gated ion channels are affected by pore block or modification of voltage dependence of activation/inactivation. Recent experiments performed on various ion channels show that allosteric modulation is an important mechanism for affecting channel function. For instance, in K(Ca)2 (formerly SK) channels, the prototypic "blocker" apamin prevents conduction by an allosteric mechanism, while TRPV1 channels are prevented from closing by a tarantula toxin, DkTx, through an interaction with residues located away from the selectivity filter. The recent evidence, therefore, suggests that in several ion channels, the region around the outer mouth of the pore is rich in binding sites and could be exploited therapeutically. These discoveries also suggest that the pharmacological vocabulary should be adapted to define these various actions.


Subject(s)
Allosteric Regulation/physiology , Calcium Channel Blockers/metabolism , Calcium Channels/metabolism , Ion Transport/physiology , Potassium Channel Blockers/metabolism , Potassium Channels, Calcium-Activated/metabolism , Potassium Channels, Voltage-Gated/metabolism , Allosteric Site , Amino Acid Sequence , Apamin/chemistry , Apamin/metabolism , Apamin/pharmacology , Binding Sites , Biodiversity , Calcium/metabolism , Calcium Channel Blockers/chemistry , Calcium Channel Blockers/pharmacology , Calcium Channels/chemistry , Humans , Ion Channel Gating , Membrane Potentials , Models, Molecular , Molecular Sequence Data , Potassium/metabolism , Potassium Channel Blockers/chemistry , Potassium Channel Blockers/pharmacology , Potassium Channels, Calcium-Activated/chemistry , Potassium Channels, Voltage-Gated/chemistry , Protein Binding , Protein Conformation , Spider Venoms/chemistry , Spider Venoms/metabolism , Spider Venoms/pharmacology
10.
J Biol Chem ; 285(35): 27067-27077, 2010 Aug 27.
Article in English | MEDLINE | ID: mdl-20562108

ABSTRACT

Activation of small conductance calcium-activated potassium (K(Ca)2) channels can regulate neuronal firing and synaptic plasticity. They are characterized by their high sensitivity to the bee venom toxin apamin, but the mechanism of block is not understood. For example, apamin binds to both K(Ca)2.2 and K(Ca)2.3 with the same high affinity (K(D) approximately 5 pM for both subtypes) but requires significantly higher concentrations to block functional current (IC(50) values of approximately 100 pM and approximately 5 nM, respectively). This suggests that steps beyond binding are needed for channel block to occur. We have combined patch clamp and binding experiments on cell lines with molecular modeling and mutagenesis to gain more insight into the mechanism of action of the toxin. An outer pore histidine residue common to both subtypes was found to be critical for both binding and block by the toxin but not for block by tetraethylammonium (TEA) ions. These data indicated that apamin blocks K(Ca)2 channels by binding to a site distinct from that used by TEA, supported by a finding that the onset of block by apamin was not affected by the presence of TEA. Structural modeling of ligand-channel interaction indicated that TEA binds deep within the channel pore, which contrasted with apamin being modeled to interact with the channel outer pore by utilizing the outer pore histidine residue. This multidisciplinary approach suggested that apamin does not behave as a classical pore blocker but blocks using an allosteric mechanism that is consistent with observed differences between binding affinity and potency of block.


Subject(s)
Apamin/pharmacology , Models, Molecular , Potassium Channel Blockers/pharmacology , Small-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Small-Conductance Calcium-Activated Potassium Channels/metabolism , Allosteric Regulation/drug effects , Allosteric Regulation/genetics , Allosteric Site/genetics , Animals , Apamin/chemistry , Bees/chemistry , Cell Line , Dose-Response Relationship, Drug , Humans , Patch-Clamp Techniques , Potassium Channel Blockers/chemistry , Protein Binding/drug effects , Rats , Small-Conductance Calcium-Activated Potassium Channels/chemistry , Small-Conductance Calcium-Activated Potassium Channels/genetics , Tetraethylammonium/pharmacology
13.
J Am Soc Mass Spectrom ; 20(1): 112-23, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18849171

ABSTRACT

A new analytical approach using matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI MSI) for the study of honeybee venom is shown. In vitro and in vivo models simulating the bee sting have been developed using live honeybees and, as the envenomation sites, pig ears and rat legs; MALDI MSI has been used to map, over time, the diffusion and distribution of three venom allergens (Api m 1, Api m 4, and Api m 6) and two venom toxins (apamine and mast cell degranulating peptide). In conjunction with other classical biochemical techniques and high resolution mass spectrometry (HRMS), structural data have been obtained that contribute to current understanding of honeybee venom composition. Initial data have also been obtained demonstrating the feasibility of mapping the organism's response to the sting. The opportunity to monitor venom diffusion and the organism's response at the same time might open new pathways for in vivo preclinical studies in designing and testing new venom immunotherapy (VIT).


Subject(s)
Bee Venoms/analysis , Insect Bites and Stings/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Allergens/analysis , Allergens/chemistry , Animals , Antigens, Plant , Apamin/analysis , Apamin/chemistry , Bee Venoms/chemistry , Bees , Ear , Extremities , Insect Bites and Stings/pathology , Insect Proteins/analysis , Insect Proteins/chemistry , Models, Biological , Muscle, Skeletal/chemistry , Muscle, Skeletal/pathology , Peptides/analysis , Peptides/chemistry , Phospholipases A/analysis , Phospholipases A/chemistry , Rats , Sensitivity and Specificity , Swine
14.
Bioorg Med Chem Lett ; 18(20): 5694-7, 2008 Oct 15.
Article in English | MEDLINE | ID: mdl-18824351

ABSTRACT

An exploratory SAR study on a series of potent, non-apamin-displacing 4-(aminomethylaryl)pyrazolopyrimidine K(Ca) channel blockers is described and their selectivity against K(Ca) channel subtypes is reported. The most potent analog, 5-chloro-N-(thiophen-2-ylmethyl)pyrazolo[1,5-a]pyrimidin-7-amine (24) displayed sub-micromolar activity in both a thallium flux and whole-cell electrophysiology assay and did not displace apamin in a competitive binding study.


Subject(s)
Apamin/chemistry , Potassium Channel Blockers/chemistry , Pyrazoles/chemical synthesis , Pyrimidines/chemical synthesis , Binding, Competitive , Cell Line , Electrophysiology , Humans , Inhibitory Concentration 50 , Models, Chemical , Potassium Channels, Calcium-Activated/metabolism , Protein Isoforms , Pyrazoles/pharmacology , Pyrimidines/chemistry , Pyrimidines/pharmacology , Structure-Activity Relationship , Thallium/chemistry
15.
Bioorg Med Chem ; 15(16): 5457-79, 2007 Aug 15.
Article in English | MEDLINE | ID: mdl-17560109

ABSTRACT

The synthesis and pharmacological testing of a series of non-peptidic blockers of the SK(Ca) (SK-3) channel is described. Target compounds were designed to mimic the spatial relationships of selected key residues in the energy-minimised structure of the octadecapeptide apamin, which are a highly potent blocker of this channel. Structures consist of a central unit, either a fumaric acid or an aromatic ring, to which are attached two alkylguanidine or two to four alkylaminoquinoline substituents. Potency was tested by the ability to inhibit the SK(Ca) channel-mediated after-hyperpolarization (AHP) in cultured rat sympathetic neurones. It was found that bis-aminoquinoline derivatives are significantly more potent as channel blockers than are the corresponding guanidines. This adds to the earlier evidence that delocalisation of positive charge through the more extensive aminoquinolinium ring system is important for effective channel binding. It was also found that an increase in activity can be gained by the addition of a third aminoquinoline residue to give non-quaternized amines which have submicromolar potencies (IC(50)=0.13-0.36 microM). Extension to four aminoquinoline residues increased the potency to IC(50)=93 nM.


Subject(s)
Aminoquinolines/chemical synthesis , Aminoquinolines/pharmacology , Apamin/pharmacology , Potassium Channel Blockers/chemical synthesis , Potassium Channel Blockers/pharmacology , Potassium Channels, Calcium-Activated/metabolism , Aminoquinolines/chemistry , Animals , Apamin/chemistry , Guanidine/chemistry , Molecular Sequence Data , Molecular Structure , Neurons/drug effects , Potassium Channel Blockers/chemistry , Rats , Structure-Activity Relationship , Tissue Culture Techniques
16.
Biopolymers ; 86(5-6): 447-62, 2007.
Article in English | MEDLINE | ID: mdl-17486576

ABSTRACT

The X-ray structure of [N-acetyl]-apamin has been solved at 0.95 A resolution. It consists of an 1-7 N-terminal loop stabilized by an Asn-beta-turn motif (2-5 residues) and a helical structure spanning the 9-18 residues tightly linked together by two disulfide bonds. However, neither this accurate X-ray nor the available solution structures allowed us to rationally explain the unusual downfield shifts observed for the Asn(2) and Glu(7) amide signals upon Glu(7) carboxylic group ionization. Thus, apamin and its [N-acetyl], [Glu(7)Gln], [Glu(7)Asp], and [Asn(2)Abu] analogues and submitted to NMR structural studies as a function of pH. We first demonstrated that the Glu(7) carboxylate group is responsible for the large downfield shifts of the Asn(2) and Glu(7) amide signals. Then, molecular dynamics (MD) simulations suggested unexpected interactions between the carboxylate group and the Asn(2) and Glu(7) amide protons as well as the N-terminal alpha-amino group, through subtle conformational changes that do not alter the global fold of apamin. In addition, a structural study of the [Asn(2)Abu] analogue, revealed an essential role of Asn(2) in the beta-turn stability and the cis/trans isomerization of the Ala(5)-Pro(6) amide bond. Interestingly, this proline isomerization was shown to also depend on the ionization state of the Glu(7) carboxyl group. However, neither destabilization of the beta-turn nor proline isomerization drastically altered the helical structure that contains the residues essential for binding. Altogether, the Asn(2) and Glu(7) residues appeared essential for the N-terminal loop conformation and thus for the selective formation of the native disulfide bonds but not for the activity.


Subject(s)
Apamin/chemistry , Amino Acid Sequence , Animals , Apamin/analogs & derivatives , Asparagine/chemistry , Crystallography, X-Ray , Dimerization , Glutamic Acid/chemistry , Hydrogen-Ion Concentration , In Vitro Techniques , Models, Molecular , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Proline/chemistry , Protein Conformation , Protein Folding , Protein Structure, Quaternary , Solutions , Thermodynamics
17.
J Mol Biol ; 369(2): 553-66, 2007 Jun 01.
Article in English | MEDLINE | ID: mdl-17433819

ABSTRACT

Most animal toxins are short proteins that appear in venom and vary in sequence, structure and function. A common characteristic of many such toxins is their apparent structural stability. Sporadic instances of endogenous toxin-like proteins that function in non-venom context have been reported. We have utilized machine learning methodology, based on sequence-derived features and guided by the notion of structural stability, in order to conduct a large-scale search for toxin and toxin-like proteins. Application of the method to insect and mammalian sequences revealed novel families of toxin-like proteins. One of these proteins shows significant similarity to ion channel inhibitors that are expressed in cone snail and assassin bug venom, and is surprisingly expressed in the bee brain. A toxicity assay in which the protein was injected to fish induced a strong yet reversible paralytic effect. We suggest that the protein may function as an endogenous modulator of voltage-gated Ca(2+) channels. Additionally, we have identified a novel mammalian cluster of toxin-like proteins that are expressed in the testis. We suggest that these proteins might be involved in regulation of nicotinic acetylcholine receptors that affect the acrosome reaction and sperm motility. Finally, we highlight a possible evolutionary link between venom toxins and antibacterial proteins. We expect our methodology to enhance the discovery of additional novel protein families.


Subject(s)
Computer Simulation , Peptides/genetics , Toxins, Biological/chemistry , Toxins, Biological/genetics , Amino Acid Sequence , Animals , Antimicrobial Cationic Peptides/chemistry , Antimicrobial Cationic Peptides/genetics , Apamin/chemistry , Apamin/genetics , Base Sequence , Bees , Humans , Insect Proteins/chemistry , Insect Proteins/genetics , Insecta , Mice , Molecular Sequence Data , Neuropeptides/chemistry , Peptides/chemistry , Peptides/classification , Protein Conformation , Reproducibility of Results , Sequence Alignment , Toxins, Biological/classification
18.
Biochim Biophys Acta ; 1712(1): 52-61, 2005 Jun 15.
Article in English | MEDLINE | ID: mdl-15890311

ABSTRACT

The effect of a hyposmotic shock and extracellular ATP on the efflux of K(+)(Rb(+)) from human breast cancer cell lines (MDA-MB-231 and MCF-7) has been examined. A hyposmotic shock increased the fractional efflux of K(+)(Rb(+)) from MDA-MB-231 cells via a pathway which was unaffected by Cl(-) replacement. Apamin, charybdotoxin or removing extracellular Ca(2+) had no effect on volume-activated K(+)(Rb(+)) efflux MDA-MB-231 cells. An osmotic shock also stimulated K(+)(Rb(+)) efflux from MCF-7 cells but to a much lesser extent than found with MDA-MB-231 cells. ATP-stimulated K(+)(Rb(+)) efflux from MDA-MB-231 cells in a dose-dependent fashion but had little effect on K(+)(Rb(+)) release from MCF-7 cells. ATP-stimulated K(+)(Rb(+)) efflux was only inhibited slightly by replacing Cl(-) with NO(3)(-). Removal of external Ca(2+) during treatment with ATP reduced the fractional efflux of K(+)(Rb(+)) in a manner suggesting a role for cellular Ca(2+) stores. Charybdotoxin, but neither apamin nor iberiotoxin, inhibited ATP-stimulated K(+)(Rb(+)) release from MDA-MB-231 cells. Suramin inhibited the ATP-activated efflux of K(+)(Rb(+)). UTP also stimulated K(+)(Rb(+)) efflux from MDA-MB-231 cells whereas ADP, AMP and adenosine were without effect. A combination of an osmotic shock and ATP increased the fractional efflux of K(+)(Rb(+)) to a level greater than the sum of the individual treatments. It appears that the hyposmotically-activated and ATP-stimulated K(+) efflux pathways are separate entities. However, there may be a degree of 'crosstalk' between the two pathways.


Subject(s)
Breast Neoplasms/pathology , Receptors, Purinergic/metabolism , Adenosine Triphosphate/chemistry , Apamin/chemistry , Calcium/metabolism , Cell Line, Tumor , Charybdotoxin/pharmacology , Dose-Response Relationship, Drug , Fluorescent Dyes/pharmacology , Fura-2/pharmacology , Humans , Osmosis , Peptides/chemistry , Potassium/chemistry , Receptors, Estrogen/metabolism , Receptors, Purinergic/chemistry , Rubidium/chemistry , Spectrometry, Fluorescence , Suramin/chemistry , Taurine/chemistry , Time Factors
19.
J Enzyme Inhib Med Chem ; 20(6): 517-23, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16408787

ABSTRACT

Small conductance calcium-activated potassium channels (SK) are widely expressed throughout the central nervous system (CNS) and the periphery. Three subtypes of SK channels have so far been identified in different parts of the brain. Activation of the SK channels by a rise in intracellular calcium leads to the hyperpolarisation of the membrane, reducing cell excitability. Blocking the SK channels might be beneficial in the treatment of depression, Parkinson's disease and cognitive disorders. However, few blockers of SK channels have been characterized. In this study, a pharmacophoric model of SK channels blockers is presented. It is based on a series of nonpeptidic compounds and apamin, a peptidic blocker. To create the pharmacophore model, the conformational space of nonpeptidic blockers was investigated to generate a series of distance constraints applied to a simulated annealing study of apamin. The resulting conformation was superimposed with the nonpeptidic blockers to give a pharmacophore.


Subject(s)
Apamin/chemistry , Drug Design , Macrocyclic Compounds/chemistry , Potassium Channel Blockers/chemistry , Quinolinium Compounds/chemistry , Small-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Amino Acid Sequence , Animals , Apamin/pharmacology , Azepines/chemistry , Azepines/pharmacology , Bees , Cadaverine/analogs & derivatives , Cadaverine/chemistry , Cadaverine/pharmacology , Macrocyclic Compounds/pharmacology , Models, Molecular , Molecular Sequence Data , Molecular Structure , Potassium Channel Blockers/pharmacology , Protein Conformation , Protein Structure, Secondary , Quinolines/chemistry , Quinolines/pharmacology , Quinolinium Compounds/pharmacology , Small-Conductance Calcium-Activated Potassium Channels/chemistry , Structure-Activity Relationship
20.
Chembiochem ; 5(8): 1075-80, 2004 Aug 06.
Article in English | MEDLINE | ID: mdl-15300830

ABSTRACT

An 18-residue miniature enzyme, Apoxaldie-1, has been designed, based on the known structure of the neurotoxic peptide apamin. Three lysine residues were introduced on the solvent-exposed face of the apamin alpha-helix to serve as an active site for decarboxylation of oxaloacetate. The oxidised form of Apoxaldie-1, in which two disulfide bonds stabilise the alpha-helix, formed spontaneously. CD spectroscopy measurements revealed that, in its oxidised form, Apoxaldie-1 adopted a stably folded structure, which was lost upon reduction of the disulfide bonds. Despite its small size and the absence of a designed binding pocket, Apoxaldie-1 displayed saturation kinetics in its oxidised form and catalysed the decarboxylation of oxaloacetate at a rate that was almost four orders of magnitude faster than that observed with n-butylamine. This rivals the performance of the best synthetic oxaloacetate decarboxylases reported to date. Unlike those, however, Apoxaldie-1 displayed significant stability. It maintained its secondary structure at temperatures in excess of 75 degrees C, in the presence of high concentrations of guanidinium chloride and at pH values as low as 2.2. Apamin-based catalysts have potential for the generation of miniature peptides that display activity under nonphysiological conditions.


Subject(s)
Apamin/analogs & derivatives , Apamin/metabolism , Carboxy-Lyases/metabolism , Amino Acid Sequence , Apamin/chemistry , Carboxy-Lyases/chemistry , Circular Dichroism , Models, Molecular , Molecular Sequence Data , Oxidation-Reduction
SELECTION OF CITATIONS
SEARCH DETAIL
...