Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Infect Disord Drug Targets ; 12(4): 316-25, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22697129

RESUMEN

A new bacteriocin, lacticin LC14, produced by Lactococcus lactis BMG6.14, was isolated and characterized. It was purified to homogeneity from overnight broth culture by ammonium sulfate precipitation, Sep-Pak chromatography, and two steps of reversed-phase HPLC. Lacticin LC14 showed bactericidal-type antimicrobial activity against several lactic acid bacteria and pathogenic strains including Listeria monocytogenes. It was inactivated by proteinase K and pronase E, but was resistant to papain, lysozyme, lipase and catalase. Lacticin LC14 was heat resistant, stable over a wide range of pH (2-10) and after treatment by solvents and detergents. Its N-terminal end was found unreactive towards Edman sequencing. Based on MALDI-TOF mass spectrometry, its molecular mass was 3333.7 Da. LC14 amino acid composition revealed a high proportion of hydrophobic residues, but no modified ones. LC14 may be able to challenge other well known other bacteriocins in probiotic and therapeutic applications.


Asunto(s)
Bacteriocinas/aislamiento & purificación , Lactococcus lactis/metabolismo , Secuencia de Aminoácidos , Aminoácidos/análisis , Bacteriocinas/análisis , Bacteriocinas/química , Bacteriocinas/farmacología , Lactococcus lactis/aislamiento & purificación
2.
J Antibiot (Tokyo) ; 61(2): 89-93, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18408329

RESUMEN

Bacteriocin J46 is a 27-residue polypeptide produced by Lactococcus lactis subsp. cremoris J46 in fermented milk. The natural form of J46 (nJ46) exhibits a broad antimicrobial spectrum. Herein, we produced the synthetic form of J46 (sJ46) by solid-phase chemical synthesis. The biochemical and physico-chemical properties of sJ46, as well as its antimicrobial activity, were found to be identical to those of its natural counterpart nJ46. It showed a potent antimicrobial activity against both lactic acid bacteria and other Gram-positive microorganisms. (1)H-NMR conformational analysis of sJ46 indicates that it adopts a flexible random coil structure.


Asunto(s)
Antibacterianos/síntesis química , Bacteriocinas/síntesis química , Lactococcus lactis/química , Animales , Antibacterianos/química , Antibacterianos/farmacología , Bacteriocinas/química , Bacteriocinas/farmacología , Pruebas de Sensibilidad Microbiana , Leche/microbiología , Conformación Molecular
3.
Toxicon ; 51(3): 353-62, 2008 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-18054371

RESUMEN

KAaH1 and KAaH2 are non-toxic peptides, isolated from the venom of the Androctonus australis hector (Aah) scorpion. In a previous study, we showed these peptides to be the most abundant (approximately 10% each) in the toxic fraction (AahG50) of the Aah venom. KAaH1 and KAaH2 showed high sequence identities (approximately 60%) with birtoxin-like peptides, which likewise are the major peptidic components of Parabuthus transvaalicus scorpion venom. Here, we report the immunological characterization of KAaH1 and KAaH2. These peptides were found to be specifically recognized by polyclonal antibodies raised against AahII, the most toxic peptide of Aah venom, and represents the second antigenic group, including toxins from different scorpion species in the world. Moreover, KAaH1 partially inhibits AahII binding to its specific antibody, suggesting some common epitopes between these two peptides. The identification of possible key antigenic residues in KAaH1 was deduced from comparison of its 3-D model with the experimental structure of AahII. Two clusters of putative antigenically important residues were found at the exposed surface; one could be constituted of V3 and D53, the other of D10, T15 and Y16. Polyclonal antibodies raised against KAaH1 in mice were found to cross-react with both AahII and AahG50, and neutralizing 5LD(50)/ml of the toxic fraction. Mice vaccinated with KAaH1 were protected against a challenge of 2LD(50) of AahG50 fraction. All these data suggest that KAaH1 has clear advantages over the use of the whole or part of the venom. KAaH1 is not toxic and could produce sera-neutralizing scorpion toxins, not only from Aah venom, but also toxins of other venoms from Buthus, Leiurus, or Parabuthus scorpion species presenting antigenically related toxins.


Asunto(s)
Péptidos/inmunología , Péptidos/farmacología , Venenos de Escorpión/antagonistas & inhibidores , Venenos de Escorpión/toxicidad , Escorpiones , Secuencia de Aminoácidos , Animales , Epítopos/química , Epítopos/inmunología , Masculino , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Péptidos/química , Conformación Proteica , Venenos de Escorpión/química , Venenos de Escorpión/inmunología
4.
Protein Sci ; 17(1): 107-18, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18042681

RESUMEN

Agitoxin 2 (AgTx2) is a 38-residue scorpion toxin, cross-linked by three disulfide bridges, which acts on voltage-gated K(+) (Kv) channels. Maurotoxin (MTX) is a 34-residue scorpion toxin with an uncommon four-disulfide bridge reticulation, acting on both Ca(2+)-activated and Kv channels. A 39-mer chimeric peptide, named AgTx2-MTX, was designed from the sequence of the two toxins and chemically synthesized. It encompasses residues 1-5 of AgTx2, followed by the complete sequence of MTX. As established by enzyme cleavage, the new AgTx2-MTX molecule displays half-cystine pairings of the type C1-C5, C2-C6, C3-C7, and C4-C8, which is different from that of MTX. The 3D structure of AgTx2-MTX solved by (1)H-NMR, revealed both alpha-helical and beta-sheet structures, consistent with a common alpha/beta scaffold of scorpion toxins. Pharmacological assays of AgTx2-MTX revealed that this new molecule is more potent than both original toxins in blocking rat Kv1.2 channel. Docking simulations, performed with the 3D structure of AgTx2-MTX, confirmed this result and demonstrated the participation of the N-terminal domain of AgTx2 in its increased affinity for Kv1.2 through additional molecular contacts. Altogether, the data indicated that replacement of the N-terminal domain of MTX by the one of AgTx2 in the AgTx2-MTX chimera results in a reorganization of the disulfide bridge arrangement and an increase of affinity to the Kv1.2 channel.


Asunto(s)
Venenos de Crotálidos/química , Venenos de Crotálidos/síntesis química , Canal de Potasio Kv.1.2/antagonistas & inhibidores , Venenos de Escorpión/química , Venenos de Escorpión/síntesis química , Secuencia de Aminoácidos , Sitios de Unión , Línea Celular , Venenos de Crotálidos/farmacología , Humanos , Cinética , Espectroscopía de Resonancia Magnética/métodos , Modelos Moleculares , Datos de Secuencia Molecular , Conformación Proteica , Proteínas Recombinantes/antagonistas & inhibidores , Venenos de Escorpión/farmacología , Transfección
5.
J Biol Chem ; 281(30): 21332-21344, 2006 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-16679310

RESUMEN

Voltage sensing by voltage-gated sodium channels determines the electrical excitability of cells, but the molecular mechanism is unknown. beta-Scorpion toxins bind specifically to neurotoxin receptor site 4 and induce a negative shift in the voltage dependence of activation through a voltage sensor-trapping mechanism. Kinetic analysis showed that beta-scorpion toxin binds to the resting state, and subsequently the bound toxin traps the voltage sensor in the activated state in a voltage-dependent but concentration-independent manner. The rate of voltage sensor trapping can be fit by a two-step model, in which the first step is voltage-dependent and correlates with the outward gating movement of the IIS4 segment, whereas the second step is voltage-independent and results in shifted voltage dependence of activation of the channel. Mutations of Glu(779) in extracellular loop IIS1-S2 and both Glu(837) and Leu(840) in extracellular loop IIS3-S4 reduce the binding affinity of beta-scorpion toxin. Mutations of positively charged and hydrophobic amino acid residues in the IIS4 segment do not affect beta-scorpion toxin binding but alter voltage dependence of activation and enhance beta-scorpion toxin action. Structural modeling with the Rosetta algorithm yielded a three-dimensional model of the toxin-receptor complex with the IIS4 voltage sensor at the extracellular surface. Our results provide mechanistic and structural insight into the voltage sensor-trapping mode of scorpion toxin action, define the position of the voltage sensor in the resting state of the sodium channel, and favor voltage-sensing models in which the S4 segment spans the membrane in both resting and activated states.


Asunto(s)
Venenos de Escorpión/química , Canales de Sodio/química , Secuencia de Aminoácidos , Línea Celular , Humanos , Riñón/citología , Cinética , Modelos Moleculares , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Mutación , Unión Proteica , Homología de Secuencia de Aminoácido , Relación Estructura-Actividad
6.
Peptides ; 26(7): 1095-108, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15949626

RESUMEN

The small-conductance Ca2+-activated K+ (SKCa) channels modulate cytosolic Ca2+ concentration in excitable and non-excitable tissues by regulating the membrane potential and are responsible of slow action potential after hyperpolarization that inhibits cell firing. Among these, human SKCa2 and SKCa3 channels differ in the pore region by only two residues: Ala331 and Asn367 (human small-conductance calcium-activated potassium channel, hSKCa2) instead of Val485 and His521 (hSKCa3). To design highly selective blockers of hSKCa channels, a number of known hSKCa2 and/or hSKCa3-active peptides (i.e. scorpion toxins and analogs thereof) were analyzed for their interactions and selectivities toward these channels. Molecular models of hSKCa2 and hSKCa3 channels (S5-H5-S6 portion) were generated, and scorpion toxins/peptides of unsolved three-dimensional (3D) structures were modeled. Models of toxin-channel complexes were generated by the bimolecular complex generation with global evaluation, and ranking (BiGGER) docking software and selected by using a screening method of the docking solutions. A high degree of correlation was found to exist between docking energies and experimental Kd values of peptides that blocked hSKCa2 and/or hSKCa3 channels, suggesting it could be appropriate to predict Kd values of other bioactive peptides. The best scoring complexes were also used to identify key residues of both interacting partners, indicating that such an approach should help the design of more active and/or selective peptide blockers of targeted ion channels.


Asunto(s)
Modelos Moleculares , Neuropéptidos/química , Canales de Potasio Calcio-Activados/química , Canales de Potasio/química , Venenos de Escorpión/química , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Simulación por Computador , Humanos , Datos de Secuencia Molecular , Neuropéptidos/antagonistas & inhibidores , Péptidos/química , Canales de Potasio Calcio-Activados/antagonistas & inhibidores , Conformación Proteica , Canales de Potasio de Pequeña Conductancia Activados por el Calcio
7.
Proteins ; 60(3): 401-11, 2005 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-15971207

RESUMEN

Scorpion toxins interact with their target ion channels through multiple molecular contacts. Because a "gain of function" approach has never been described to evaluate the importance of the molecular contacts in defining toxin affinity, we experimentally examined whether increasing the molecular contacts between a toxin and an ion channel directly impacts toxin affinity. For this purpose, we focused on two scorpion peptides, the well-characterized maurotoxin with its variant Pi1-like disulfide bridging (MTX(Pi1)), used as a molecular template, and butantoxin (BuTX), used as an N-terminal domain provider. BuTX is found to be 60-fold less potent than MTX(Pi1) in blocking Kv1.2 (IC(50) values of 165 nM for BuTX versus 2.8 nM for MTX(Pi1)). Removal of its N-terminal domain (nine residues) further decreases BuTX affinity for Kv1.2 by 5.6-fold, which is in agreement with docking simulation data showing the importance of this domain in BuTX-Kv1.2 interaction. Transfer of the BuTX N-terminal domain to MTX(Pi1) results in a chimera with five disulfide bridges (BuTX-MTX(Pi1)) that exhibits 22-fold greater affinity for Kv1.2 than MTX(Pi1) itself, in spite of the lower affinity of BuTX as compared to MTX(Pi1). Docking experiments performed with the 3-D structure of BuTX-MTX(Pi1) in solution, as solved by (1)H-NMR, reveal that the N-terminal domain of BuTX participates in the increased affinity for Kv1.2 through additional molecular contacts. Altogether, the data indicate that acting on molecular contacts between a toxin and a channel is an efficient strategy to modulate toxin affinity.


Asunto(s)
Biología Computacional/métodos , Canal de Potasio Kv.1.2/química , Canales de Potasio con Entrada de Voltaje/química , Proteómica/métodos , Venenos de Escorpión/química , Secuencia de Aminoácidos , Animales , Dicroismo Circular , Cisteína/química , Disulfuros/química , Electrofisiología , Concentración 50 Inhibidora , Espectroscopía de Resonancia Magnética , Modelos Moleculares , Datos de Secuencia Molecular , Péptidos/química , Unión Proteica , Conformación Proteica , Estructura Terciaria de Proteína , Proteínas Recombinantes de Fusión/química , Escorpiones , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Ácido Trifluoroacético/química
8.
J Pept Sci ; 10(11): 666-77, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15568681

RESUMEN

Aah I is a 63-residue alpha-toxin isolated from the venom of the Buthidae scorpion Androctonus australis hector, which is considered to be the most dangerous species. We report here the first chemical synthesis of Aah I by the solid-phase method, using a Fmoc strategy. The synthetic toxin I (sAah I) was renatured in DMSO-Tris buffer, purified and subjected to thorough analysis and comparison with the natural toxin. The sAah I showed physico-chemical (CD spectrum, molecular mass, HPLC elution), biochemical (amino-acid composition, sequence), immunochemical and pharmacological properties similar to those of the natural toxin. The synthetic toxin was recognized by a conformation-dependent monoclonal anti-Aah I antibody, with an IC50 value close to that for the natural toxin. Following intracerebroventricular injection, the synthetic and the natural toxins were similarly lethal to mice. In voltage-clamp experiments, Na(v) 1.2 sodium channel inactivation was inhibited by the application of sAah I or of the natural toxin in a similar way. This work describes a simple protocol for the chemical synthesis of a scorpion alpha-toxin, making it possible to produce structural analogues in time.


Asunto(s)
Neurotoxinas/síntesis química , Venenos de Escorpión/síntesis química , Escorpiones/patogenicidad , Canales de Sodio/efectos de los fármacos , Fosfolipasas de Tipo C/síntesis química , Animales , Anticuerpos Monoclonales , Afinidad de Anticuerpos , Electrofisiología , Ratones , Neurotoxinas/farmacología , Renaturación de Proteína , Venenos de Escorpión/farmacología , Bloqueadores de los Canales de Sodio , Tasa de Supervivencia , Fosfolipasas de Tipo C/inmunología , Fosfolipasas de Tipo C/farmacología
9.
Nucleic Acids Res ; 31(21): e131, 2003 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-14576331

RESUMEN

Cre recombinase is extensively used to engineer the genome of experimental animals. However, its usefulness is still limited by the lack of an efficient temporal control over its activity. To overcome this, we have developed DiCre, a regulatable fragment complementation system for Cre. The enzyme was split into two moieties that were fused to FKBP12 (FK506-binding protein) and FRB (binding domain of the FKBP12-rapamycin-associated protein), respectively. These can be efficiently heterodimerized by rapamycin. Several variants, based on splitting Cre at different sites and using different linker peptides, were tested in an indicator cell line. The fusion proteins, taken separately, had no recombinase activity. Stable transformants, co-expressing complementing fragments based on splitting Cre between Asn59 and Asn60, displayed low background activity affecting 0.05-0.4% of the cells. Rapamycin induced a rapid recombination, reaching 100% by 48-72 h, with an EC50 of 0.02 nM. Thus, ligand-induced dimerization can efficiently regulate Cre, and should be useful to achieve a tight temporal control of its activity, such as in the case of the creation of conditional knock-out animals.


Asunto(s)
Integrasas/química , Integrasas/metabolismo , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Proteínas Virales/química , Proteínas Virales/metabolismo , Secuencia de Aminoácidos , Animales , Línea Celular , Dimerización , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Prueba de Complementación Genética , Integrasas/genética , Ligandos , Modelos Moleculares , Fragmentos de Péptidos/genética , Estructura Cuaternaria de Proteína/efectos de los fármacos , Ratas , Eliminación de Secuencia/genética , Sirolimus/farmacología , Transfección , Proteínas Virales/genética
10.
Eur J Biochem ; 270(17): 3583-92, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12919322

RESUMEN

Pi4 is a 38-residue toxin cross-linked by four disulfide bridges that has been isolated from the venom of the Chactidae scorpion Pandinus imperator. Together with maurotoxin, Pi1, Pi7 and HsTx1, Pi4 belongs to the alpha KTX6 subfamily of short four-disulfide-bridged scorpion toxins acting on K+ channels. Due to its very low abundance in venom, Pi4 was chemically synthesized in order to better characterize its pharmacology and structural properties. An enzyme-based cleavage of synthetic Pi4 (sPi4) indicated half-cystine pairings between Cys6-Cys27, Cys12-32, Cys16-34 and Cys22-37, which denotes a conventional pattern of scorpion toxin reticulation (Pi1/HsTx1 type). In vivo, sPi4 was lethal after intracerebroventricular injection to mice (LD50 of 0.2 microg per mouse). In vitro, addition of sPi4 onto Xenopus laevis oocytes heterologously expressing various voltage-gated K+ channel subtypes showed potent inhibition of currents from rat Kv1.2 (IC50 of 8 pm) and Shaker B (IC50 of 3 nm) channels, whereas no effect was observed on rat Kv1.1 and Kv1.3 channels. The sPi4 was also found to compete with 125I-labeled apamin for binding to small-conductance Ca(2+)-activated K+ (SK) channels from rat brain synaptosomes (IC50 value of 0.5 microm). sPi4 is a high affinity blocker of the Kv1.2 channel. The toxin was docked (BIGGER program) on the Kv channel using the solution structure of sPi4 and a molecular model of the Kv1.2 channel pore region. The model suggests a key role for residues Arg10, Arg19, Lys26 (dyad), Ile28, Lys30, Lys33 and Tyr35 (dyad) in the interaction and the associated blockage of the Kv1.2 channel.


Asunto(s)
Bloqueadores de los Canales de Potasio/síntesis química , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio/efectos de los fármacos , Venenos de Escorpión/síntesis química , Venenos de Escorpión/farmacología , Secuencia de Aminoácidos , Animales , Apamina/química , Apamina/metabolismo , Calcio/química , Calcio/metabolismo , Cisteína/química , Cisteína/genética , Disulfuros/química , Electrofisiología , Dosificación Letal Mediana , Ratones , Ratones Endogámicos C57BL , Modelos Moleculares , Datos de Secuencia Molecular , Bloqueadores de los Canales de Potasio/química , Bloqueadores de los Canales de Potasio/metabolismo , Canales de Potasio/fisiología , Unión Proteica , Ratas , Venenos de Escorpión/química , Venenos de Escorpión/metabolismo , Homología de Secuencia de Aminoácido , Sinaptosomas/metabolismo , Xenopus laevis
11.
J Biol Chem ; 278(33): 31095-104, 2003 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-12783861

RESUMEN

Maurotoxin (MTX) is a 34-residue toxin that has been isolated initially from the venom of the scorpion Scorpio maurus palmatus. It presents a large number of pharmacological targets, including small conductance Ca2+-activated and voltage-gated K+ channels. Contrary to other toxins of the alpha-KTx6 family (Pi1, Pi4, Pi7, and HsTx1), MTX exhibits a unique disulfide bridge organization of the type C1-C5, C2-C6, C3-C4, and C7-C8 (instead of the conventional C1-C5, C2-C6, C3-C7, and C4-C8, herein referred to as Pi1-like) that does not prevent its folding along the classic alpha/beta scaffold of scorpion toxins. Here, we developed an innovative strategy of chemical peptide synthesis to produce an MTX variant (MTXPi1) with a conventional pattern of disulfide bridging without any alteration of the toxin chemical structure. This strategy was used solely to address the impact of half-cystine pairings on MTX structural properties and pharmacology. The data indicate that MTXPi1 displays some marked changes in affinities toward the target K+ channels. Computed docking analyses using molecular models of both MTXPi1 and the various voltage-gated K+ channel subtypes (Shaker B, Kv1.2, and Kv1.3) were found to correlate with MTXPi1 pharmacology. A functional map detailing the interaction between MTXPi1 and Shaker B channel was generated in line with docking experiments.


Asunto(s)
Disulfuros/química , Venenos de Escorpión/química , Venenos de Escorpión/toxicidad , Escorpiones/química , Secuencia de Aminoácidos , Animales , Apamina/metabolismo , Apamina/farmacología , Sitios de Unión , Unión Competitiva , Radioisótopos de Yodo , Potenciales de la Membrana/efectos de los fármacos , Datos de Secuencia Molecular , Oocitos/fisiología , Canales de Potasio/química , Canales de Potasio/metabolismo , Estructura Terciaria de Proteína , Ratas , Venenos de Escorpión/metabolismo , Análisis de Secuencia de Proteína , Canales de Potasio de la Superfamilia Shaker , Sinaptosomas/efectos de los fármacos , Xenopus
12.
Eur J Biochem ; 269(12): 2831-41, 2002 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12071945

RESUMEN

BotXIV and LqhalphaIT are two structurally related long chain scorpion alpha-toxins that inhibit sodium current inactivation in excitable cells. However, while LqhalphaIT from Leiurus quinquestriatus hebraeus is classified as a true and strong insect alpha-toxin, BotXIV from Buthus occitanus tunetanus is characterized by moderate biological activities. To assess the possibility that structural differences between these two molecules could reflect the localization of particular functional topographies, we compared their sequences. Three structurally deviating segments located in three distinct and exposed loops were identified. They correspond to residues 8-10, 19-22, and 38-43. To evaluate their functional role, three BotXIV/LqhalphaIT chimeras were designed by transferring the corresponding LqhalphaIT sequences into BotXIV. Structural and antigenic characterizations of the resulting recombinant chimera show that BotXIV can accommodate the imposed modifications, confirming the structural flexibility of that particular alpha/beta fold. Interestingly, substitution of residues 8-10 yields to a new electrophysiological profile of the corresponding variant, partially comparable to that one of alpha-like scorpion toxins. Taken together, these results suggest that even limited structural deviations can reflect functional diversity, and also that the structure-function relationships between insect alpha-toxins and alpha-like scorpion toxins are probably more complex than expected.


Asunto(s)
Inmunotoxinas/química , Venenos de Escorpión/química , Secuencia de Aminoácidos , Animales , Electrofisiología , Inmunotoxinas/genética , Inmunotoxinas/inmunología , Insectos , Ratones , Ratones Endogámicos C57BL , Modelos Moleculares , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Venenos de Escorpión/genética , Venenos de Escorpión/inmunología , Electricidad Estática , Pruebas de Toxicidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...