Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros











Intervalo de año de publicación
1.
Channels (Austin) ; 13(1): 17-32, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-30477399

RESUMEN

Large-conductance Ca2+-activated K+ (BK) channels are composed of a pore-forming α and a variable number of auxiliary ß subunits and play important roles in regulating excitability, action potential waveforms and firing patterns, particularly in neurons and endocrine and cardiovascular cells. The ß2 subunits increase the diversity of gating and pharmacological properties. Its extracellular loop contains eight cysteine residues, which can pair to form a high-order structure, underlying the stability of the extracellular loop of ß2 subunits and the functional effects on BK channels. However, how these cysteines form disulfide bonds still remains unclear. To address this, based on the fact that the rectification and association of BK α to ß2 subunits are highly sensitive to disruption of the disulfide bonds in the extracellular loop of ß2, we developed a rectification ratio based assay by combining the site-directed mutagenesis, electrophysiology and enzymatic cleavage. Three disulfide bonds: C1(C84)-C5(C113), C3(C101)-C7(C148) and C6(C142)-C8C(174) are successfully deduced in ß2 subunit in complex with a BK α subunit, which are helpful to predict structural model of ß2 subunits through computational simulation and to understand the interface between the extracellular domain of the ß subunits and the pore-forming α subunit.


Asunto(s)
Disulfuros/análisis , Canales de Potasio de Gran Conductancia Activados por el Calcio/química , Animales , Ratones , Simulación de Dinámica Molecular
2.
Biochemistry ; 57(44): 6349-6355, 2018 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-30281282

RESUMEN

As an ω-conopeptide originally discovered from Conus striatus, SO-3 contains 25 amino acid residues and three disulfide bridges. Our previous study has shown that this peptide possesses potent analgesic activity in rodent pain models (mouse and rat), and it specifically inhibits an N-type calcium ion channel (Cav2.2). In the study presented here, we investigated the key amino acid residues for their inhibitory activity against Cav2.2 expressed in HEK 293 cells and analgesic activity in mice. To improve the inhibitory activity of SO-3, we also evaluated the effects of some amino acid residues derived from the corresponding residues of ω-peptide MVIIA, CVID, or GVIA. Our data reveal that Lys6, Ile11, and Asn14 are the important functional amino acid residues for SO-3. The replacement of some amino acid residues of SO-3 in loop 1 with the corresponding residues of CVID and GVIA improved the inhibitory activity of SO-3. The binding mode of Cav2.2 with SO-3 amino acids in loop 1 and loop 2 may be somewhat different from that of MVIIA. This study expanded our knowledge of the structure-activity relationship of ω-peptides and provided a new strategy for improving the potency of Cav2.2 inhibitors.


Asunto(s)
Analgésicos/farmacología , Conducta Animal/efectos de los fármacos , Canales de Calcio Tipo N/química , Canales de Calcio Tipo N/metabolismo , Dolor/tratamiento farmacológico , Péptidos/farmacología , Analgésicos/química , Animales , Células HEK293 , Humanos , Ratones , Modelos Moleculares , Dolor/metabolismo , Péptidos/química , Conformación Proteica , Ratas , Relación Estructura-Actividad
3.
Neuropharmacology ; 101: 137-45, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26344359

RESUMEN

MVIIA (ziconotide) is a specific inhibitor of N-type calcium channel, Cav2.2. It is derived from Cone snail and currently used for the treatment of severe chronic pains in patients unresponsive to opioid therapy. However, MVIIA produces severe side-effects, including dizziness, nystagmus, somnolence, abnormal gait, and ataxia, that limit its wider application. We previously identified a novel inhibitor of Cav2.2, ω-conopeptide SO-3, which possesses similar structure and analgesic activity to MVIIA's. To investigate the key residues for MVIIA toxicity, MVIIA/SO-3 hybrids and MVIIA variants carrying mutations in its loop 2 were synthesized. The substitution of MVIIA's loop 1 with the loop 1 of SO-3 resulted in significantly reduced Cav2.2 binding activity in vitro; the replacement of MVIIA loop 2 by the loop 2 of SO-3 not only enhanced the peptide/Cav2.2 binding but also decreased its toxicity on goldfish, attenuated mouse tremor symptom, spontaneous locomotor activity, and coordinated locomotion function. Further mutation analysis and molecular calculation revealed that the toxicity of MVIIA mainly arose from Met(12) in the loop 2, and this residue inserts into a hydrophobic hole (Ile(300), Phe(302) and Leu(305)) located between repeats II and III of Cav2.2. The combinative mutations of the loop 2 of MVIIA or other ω-conopeptides may be used for future development of more effective Cav2.2 inhibitors with lower side effects.


Asunto(s)
Bloqueadores de los Canales de Calcio/toxicidad , Canales de Calcio Tipo N/metabolismo , omega-Conotoxinas/toxicidad , Animales , Bloqueadores de los Canales de Calcio/metabolismo , Canales de Calcio Tipo N/genética , Carpa Dorada , Células HEK293 , Humanos , Locomoción/efectos de los fármacos , Locomoción/genética , Masculino , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/genética , Ratones , Ratones Endogámicos , Trastornos Motores/tratamiento farmacológico , Trastornos Motores/genética , Mutación , Neuralgia/tratamiento farmacológico , Neuralgia/etiología , Péptidos/farmacología , Conformación Proteica , Estructura Secundaria de Proteína , Ratas , Ratas Sprague-Dawley , Tiempo de Reacción/efectos de los fármacos , Homología de Secuencia de Aminoácido , Temblor/inducido químicamente , omega-Conotoxinas/química
4.
Acta Crystallogr F Struct Biol Commun ; 71(Pt 8): 1033-7, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26249695

RESUMEN

Nosiheptide is a member of the thiopeptide family of antibiotics which demonstrates potent activities against various bacterial pathogens. The formation of its C-terminal amide is catalysed by NosA in an unusual strategy for maturating certain thiopeptides by processing precursor peptides featuring a serine extension. Here, a recombinant C-terminally truncated selenomethionine-derivatized NosA1-111 variant from Streptomyces actuosus consisting of residues 1-111, named SeMet NosA1-111, was crystallized using the sitting-drop vapour-diffusion method. Diffraction data were collected to 2.40 Šresolution using synchrotron radiation. The crystals belonged to the primitive cubic space group P4132, with unit-cell parameters a = b = c = 143.3 Å. Assuming the presence of three molecules in the asymmetric unit, the calculated Matthews coefficient was 3.94 Å(3) Da(-1) and the corresponding solvent content was 40.3%.


Asunto(s)
Proteínas Bacterianas/química , Péptidos/química , Proteínas Recombinantes de Fusión/química , Streptomyces/química , Secuencia de Aminoácidos , Proteínas Bacterianas/genética , Clonación Molecular , Cristalización , Cristalografía por Rayos X , Escherichia coli/genética , Escherichia coli/metabolismo , Expresión Génica , Datos de Secuencia Molecular , Péptidos/genética , Proteínas Recombinantes de Fusión/genética , Selenometionina/química , Selenometionina/metabolismo , Alineación de Secuencia , Streptomyces/enzimología , Tiazoles/química , Tiazoles/metabolismo
5.
Biochem Biophys Res Commun ; 454(1): 151-6, 2014 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-25450372

RESUMEN

T-superfamily conotoxins have a typical cysteine pattern of "CC-CC", and are known to mainly target calcium or sodium ion channels. Recently, we screened the targets of a series of T-superfamily conotoxins and found that a new T-superfamily conotoxin TxVC (KPCCSIHDNSCCGL-NH2) from the venom of Conus textile. It selectively targeted the neuronal nicotinic acetylcholine receptor (nAChR) subtypes α4ß2 and α3ß2, with IC50 values of 343.4 and 1047.2nM, respectively, but did not exhibit obvious pharmacological effects on voltage-gated potassium, sodium or calcium channel in DRG cells, the BK channels expressed in HEK293 cells, or the Kv channels in LßT2 cells. The changes in the inhibitory activities of its Ala mutants, the NMR structure, and molecular simulation results based on other conotoxins targeting nAChR α4ß2, all demonstrated that the residues Ile(6) and Leu(14) were the main hydrophobic pharmacophores. To our best knowledge, this is the first T-superfamily conotoxin that inhibits neuronal nAChRs and possesses high binding affinity to α4ß2. This finding will expand the knowledge of the targets of T-superfamily conotoxins and the motif information could help the design of new nAChR inhibitors.


Asunto(s)
Conotoxinas/química , Conotoxinas/toxicidad , Caracol Conus/química , Receptores Nicotínicos/efectos de los fármacos , Secuencia de Aminoácidos , Animales , Sitios de Unión , Células Cultivadas , Conotoxinas/genética , Caracol Conus/genética , Femenino , Células HEK293 , Humanos , Modelos Moleculares , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Resonancia Magnética Nuclear Biomolecular , Oocitos/efectos de los fármacos , Oocitos/metabolismo , Conformación Proteica , Receptores Nicotínicos/metabolismo , Proteínas Recombinantes/efectos de los fármacos , Proteínas Recombinantes/metabolismo , Xenopus
6.
Biochem Biophys Res Commun ; 446(2): 519-22, 2014 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-24613831

RESUMEN

An 84-residue bactericidal peptide, PSK, was purified from a Chrysomya megacephala fly larvae preparation. Its amino acid sequence is similar to that of a previously reported larval peptide of the Drosophila genus (SK84) noticed for its anticancer and antimicrobial properties. The PSK sequence is also homologous to mitochondrial ATPase inhibitors from insects to humans (35-65% sequence identity), indicating an intracellular protein target and possible mechanism for PSK. It contains a cluster of six glycine residues, and has several two- and three-residue repeats. It is active against both Gram-positive and Gram-negative bacteria via a mechanism apparently involving cell membrane disintegration and inhibition of ATP hydrolysis. In addition, PSK induces an inward cationic current in pancreatic ß cells. Together, the findings identify a bioactive peptide of the ATPase inhibitor family with specific effects on both prokaryotic and mammalian cells.


Asunto(s)
Antibacterianos/farmacología , Fenómenos Fisiológicos Bacterianos/efectos de los fármacos , Dípteros/metabolismo , Células Secretoras de Insulina/fisiología , Activación del Canal Iónico/fisiología , Proteínas/farmacología , Secuencia de Aminoácidos , Animales , Antibacterianos/química , Línea Celular , Humanos , Hidrólisis , Células Secretoras de Insulina/efectos de los fármacos , Activación del Canal Iónico/efectos de los fármacos , Larva/metabolismo , Datos de Secuencia Molecular , Proteínas/química , Inhibidores de la Bomba de Protones/química , Inhibidores de la Bomba de Protones/farmacología , Relación Estructura-Actividad , Proteína Inhibidora ATPasa
7.
PLoS One ; 8(10): e77105, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24204745

RESUMEN

Computational modeling has emerged as an indispensable approach to resolve and predict the intricate interplay among the many ion channels underlying neuronal excitability. However, simulation results using the classic formula-based Hodgkin-Huxley (H-H) model or the superior Markov kinetic model of ion channels often deviate significantly from native cellular signals despite using carefully measured parameters. Here we found that the filters of patch-clamp amplifier not only delayed the signals, but also introduced ringing, and that the residual series resistance in experiments altered the command voltages, which had never been fully eliminated by improving the amplifier itself. To remove all the above errors, a virtual device with the parameters exactly same to that of amplifier was introduced into Markov kinetic modeling so as to establish a null-deviation model. We demonstrate that our novel null-deviation approach fully restores the native gating-kinetics of ion-channels with the data recorded at any condition, and predicts spike waveform and firing patterns clearly distinctive from those without correction.


Asunto(s)
Activación del Canal Iónico/fisiología , Canales Iónicos/fisiología , Modelos Neurológicos , Neuronas/fisiología , Potenciales de Acción/fisiología , Algoritmos , Animales , Células CHO , Simulación por Computador , Cricetinae , Cricetulus , Células HEK293 , Humanos , Canales Iónicos/metabolismo , Potenciales de la Membrana/fisiología , Neuronas/metabolismo , Técnicas de Placa-Clamp , Canales de Potasio Shaw/metabolismo , Canales de Potasio Shaw/fisiología
8.
Hypertension ; 62(2): 415-25, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23734009

RESUMEN

Human ß-defensin 2 (HBD2) is a cysteine-rich cationic antimicrobial peptide known for its important role in innate immune system. Intensive studies have demonstrated its antimicrobial and chemotactic activities in vitro. In this study, ELISA analysis showed that HBD2 was significantly downregulated in sera of patients with hypertension. It relaxed vessel smooth muscle by acting on the major regulatory pathways, contributing to vessel smooth muscle contraction. Electrophysiology analysis indicated that HBD2 acted as an opener of large-conductance Ca(2+)-activated potassium (BKCa)-mSlo+hß1 channels and increased BKCa currents. Mutation analysis revealed that HBD2 activated BKCa-mSlo+hß1 channels via interacting with Leu41 and Gln43 of ß1-loop. In vivo experiments suggested that HBD2 at 4 × to 6 × of physiological concentration exerted hypotensive effect in monkeys significantly, whereas the selective blocker of BKCa channels, Paxilline, inhibited the effect. HBD2 is the first peptide opener of BKCa-mSlo+hß1 channels. It may be a novel regulator of blood pressure and provides a new therapeutic target for the treatment of hypertension. The HBD2 blockade of the BKCa channels may represent a new type of cross-talk between immune and cardiovascular systems.


Asunto(s)
Hipotensión/etiología , Canales de Potasio Calcio-Activados/fisiología , Vasodilatación , beta-Defensinas/fisiología , Animales , Velocidad del Flujo Sanguíneo , Presión Sanguínea/efectos de los fármacos , Femenino , Humanos , Hipotensión/sangre , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/metabolismo , Macaca mulatta , Masculino , Persona de Mediana Edad , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , Cadenas Ligeras de Miosina/metabolismo , Fosfatasa de Miosina de Cadena Ligera/metabolismo , Fosforilación , Proteínas Recombinantes/farmacología , beta-Defensinas/sangre , beta-Defensinas/farmacología , Proteína de Unión al GTP rhoA/metabolismo
9.
J Proteome Res ; 11(12): 6197-212, 2012 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-23148443

RESUMEN

Centipedes have venom glands in their first pair of limbs, and their venoms contain a large number of components with different biochemical and pharmacological properties. However, information about the compositions and functions of their venoms is largely unknown. In this study, Scolopendra subspinipes dehaani venoms were systematically investigated by transcriptomic and proteomic analysis coupled with biological function assays. After random screening approximately 1500 independent clones, 1122 full length cDNA sequences, which encode 543 different proteins, were cloned from a constructed cDNA library using a pair of venom glands from a single centipede species. Neurotoxins, ion channel acting components and venom allergens were the main fractions of the crude venom as revealed by transcriptomic analysis. Meanwhile, 40 proteins/peptides were purified and characterized from crude venom of S. subspinipes dehaani. The N-terminal amino acid sequencing and mass spectrum results of 29 out of these 40 proteins or peptides matched well with their corresponding cDNAs. The purified proteins/peptides showed different pharmacological properties, including the following: (1) platelet aggregating activity; (2) anticoagulant activity; (3) phospholipase A(2) activity; (4) trypsin inhibiting activity; (5) voltage-gated potassium channel activities; (6) voltage-gated sodium channel activities; (7) voltage-gated calcium channel activities. Most of them showed no significant similarity to other protein sequences deposited in the known public database. This work provides the largest number of protein or peptide candidates with medical-pharmaceutical significance and reveals the toxin nature of centipede S. subspinipes dehaani venom.


Asunto(s)
Venenos de Artrópodos/análisis , Artrópodos/química , Perfilación de la Expresión Génica/métodos , Secuencia de Aminoácidos , Animales , Anticoagulantes/química , Anticoagulantes/aislamiento & purificación , Proteínas de Artrópodos/análisis , Proteínas de Artrópodos/química , Proteínas de Artrópodos/genética , Venenos de Artrópodos/química , Venenos de Artrópodos/genética , Clonación Molecular , Bases de Datos de Proteínas , Activación Enzimática , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/aislamiento & purificación , Glándulas Exocrinas/química , Biblioteca de Genes , Células HeLa , Hemolíticos/química , Hemolíticos/aislamiento & purificación , Humanos , Masculino , Datos de Secuencia Molecular , Neurotoxinas/análisis , Neurotoxinas/química , Neurotoxinas/genética , Péptidos/análisis , Péptidos/química , Fosfolipasas A2/química , Fosfolipasas A2/genética , Fosfolipasas A2/aislamiento & purificación , Agregación Plaquetaria , Proteómica/métodos , Ratas , Ratas Wistar , Análisis de Secuencia de Proteína , Especificidad de la Especie , Transcriptoma , Agonistas del Canal de Sodio Activado por Voltaje/química , Agonistas del Canal de Sodio Activado por Voltaje/aislamiento & purificación
10.
PLoS One ; 7(4): e35154, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22511981

RESUMEN

BACKGROUND: Although the basic scorpion K(+) channel toxins (KTxs) are well-known pharmacological tools and potential drug candidates, characterization the acidic KTxs still has the great significance for their potential selectivity towards different K(+) channel subtypes. Unfortunately, research on the acidic KTxs has been ignored for several years and progressed slowly. PRINCIPAL FINDINGS: Here, we describe the identification of nine new acidic KTxs by cDNA cloning and bioinformatic analyses. Seven of these toxins belong to three new α-KTx subfamilies (α-KTx28, α-KTx29, and α-KTx30), and two are new members of the known κ-KTx2 subfamily. ImKTx104 containing three disulfide bridges, the first member of the α-KTx28 subfamily, has a low sequence homology with other known KTxs, and its NMR structure suggests ImKTx104 adopts a modified cystine-stabilized α-helix-loop-ß-sheet (CS-α/ß) fold motif that has no apparent α-helixs and ß-sheets, but still stabilized by three disulfide bridges. These newly described acidic KTxs exhibit differential pharmacological effects on potassium channels. Acidic scorpion toxin ImKTx104 was the first peptide inhibitor found to affect KCNQ1 channel, which is insensitive to the basic KTxs and is strongly associated with human cardiac abnormalities. ImKTx104 selectively inhibited KCNQ1 channel with a K(d) of 11.69 µM, but was less effective against the basic KTxs-sensitive potassium channels. In addition to the ImKTx104 toxin, HeTx204 peptide, containing a cystine-stabilized α-helix-loop-helix (CS-α/α) fold scaffold motif, blocked both Kv1.3 and KCNQ1 channels. StKTx23 toxin, with a cystine-stabilized α-helix-loop-ß-sheet (CS-α/ß) fold motif, could inhibit Kv1.3 channel, but not the KCNQ1 channel. CONCLUSIONS/SIGNIFICANCE: These findings characterize the structural and functional diversity of acidic KTxs, and could accelerate the development and clinical use of acidic KTxs as pharmacological tools and potential drugs.


Asunto(s)
Canales de Potasio/química , Venenos de Escorpión/química , Escorpiones/química , Secuencia de Aminoácidos , Animales , Biología Computacional , Modelos Moleculares , Resonancia Magnética Nuclear Biomolecular , Filogenia , Canales de Potasio/fisiología , Alineación de Secuencia , Análisis de Secuencia de Proteína
11.
J Biol Chem ; 287(17): 13813-21, 2012 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-22354971

RESUMEN

The potassium channel Kv1.3 is an attractive pharmacological target for autoimmune diseases. Specific peptide inhibitors are key prospects for diagnosing and treating these diseases. Here, we identified the first scorpion Kunitz-type potassium channel toxin family with three groups and seven members. In addition to their function as trypsin inhibitors with dissociation constants of 140 nM for recombinant LmKTT-1a, 160 nM for LmKTT-1b, 124 nM for LmKTT-1c, 136 nM for BmKTT-1, 420 nM for BmKTT-2, 760 nM for BmKTT-3, and 107 nM for Hg1, all seven recombinant scorpion Kunitz-type toxins could block the Kv1.3 channel. Electrophysiological experiments showed that six of seven scorpion toxins inhibited ~50-80% of Kv1.3 channel currents at a concentration of 1 µM. The exception was rBmKTT-3, which had weak activity. The IC(50) values of rBmKTT-1, rBmKTT-2, and rHg1 for Kv1.3 channels were ~129.7, 371.3, and 6.2 nM, respectively. Further pharmacological experiments indicated that rHg1 was a highly selective Kv1.3 channel inhibitor with weak affinity for other potassium channels. Different from classical Kunitz-type potassium channel toxins with N-terminal regions as the channel-interacting interfaces, the channel-interacting interface of Hg1 was in the C-terminal region. In conclusion, these findings describe the first scorpion Kunitz-type potassium channel toxin family, of which a novel inhibitor, Hg1, is specific for Kv1.3 channels. Their structural and functional diversity strongly suggest that Kunitz-type toxins are a new source to screen and design potential peptides for diagnosing and treating Kv1.3-mediated autoimmune diseases.


Asunto(s)
Péptidos/química , Canales de Potasio/química , Venenos de Escorpión/farmacología , Secuencia de Aminoácidos , Animales , Enfermedades Autoinmunes/metabolismo , Bovinos , Electrofisiología/métodos , Biblioteca de Genes , Células HEK293 , Humanos , Concentración 50 Inhibidora , Canal de Potasio Kv1.3/química , Datos de Secuencia Molecular , Mapeo de Interacción de Proteínas/métodos , Venenos de Escorpión/química , Escorpiones , Homología de Secuencia de Aminoácido , Porcinos , Inhibidores de Tripsina/farmacología , Ponzoñas/metabolismo
12.
Peptides ; 28(12): 2306-12, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18006119

RESUMEN

LmKTx8, the first toxic gene isolated from the venom of scorpion Lychas mucronatus by constructing cDNA library method, was expressed and characterized physiologically. The mature peptide has 40 residues including six conserved cysteines, and is classified as one of alpha-KTx11 subfamily. Using patch-clamp recording, the recombinant LmKTx8 (rLmKTx8) was used to test the effect on voltage-gated K(+) channels (Kv1.3) stably expressed in COS7 cells and large conductance-Ca(2+)-activated K(+) (BK) channels expressed in HEK293. The results of electrophysiological experiments showed that the rLmKTx8 was a potent inhibitor of Kv1.3 channels with an IC(50)=26.40+/-1.62nM, but 100nM rLmKTx8 did not block the BK currents. LmKTx8 or its analogs might serve as a potential candidate for the development of new drugs for autoimmune diseases.


Asunto(s)
Bloqueadores de los Canales de Potasio/farmacología , Venenos de Escorpión/genética , Venenos de Escorpión/farmacología , Escorpiones/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Línea Celular , Clonación Molecular , Cartilla de ADN/genética , ADN Complementario/genética , Electrofisiología , Humanos , Canal de Potasio Kv1.3/antagonistas & inhibidores , Modelos Moleculares , Datos de Secuencia Molecular , Peso Molecular , Técnicas de Placa-Clamp , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/farmacología , Homología de Secuencia de Aminoácido , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
13.
J Cell Physiol ; 212(2): 348-57, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17523149

RESUMEN

Auxiliary beta-subunits associated with pore-forming Slo1 alpha-subunits play an essential role in regulating functional properties of large-conductance, voltage- and Ca(2+)-activated K(+) channels commonly termed BK channels. Even though both noninactivating and inactivating BK channels are thought to be regulated by beta-subunits (beta1, beta2, beta3, or beta4), the molecular determinants underlying inactivating BK channels in native cells have not been extensively demonstrated. In this study, rbeta2 (but not rbeta3-subunit) was identified as a molecular component in rat lumbar L4-6 dorsal root ganglia (DRG) by RT-PCR responsible for inactivating large-conductance Ca(2+)-dependent K(+) currents (BK(i) currents) in small sensory neurons. The properties of native BK(i) currents obtained from both whole-cell and inside-out patches are very similar to inactivating BK channels produced by co-expressing mSlo1 alpha- and hbeta2-subunits in Xenopus oocytes. Intracellular application of 0.5 mg/ml trypsin removes inactivation of BK(i) channels, and the specific blockers of BK channels, charybdotoxin (ChTX) and iberiotoxin (IbTX), inhibit these BK(i) currents. Single BK(i) channel currents derived from inside-out patches revealed that one BK(i) channel contained three rbeta2-subunits (on average), with a single-channel conductance about 217 pS under 160 K(+) symmetrical recording conditions. Blockade of BK(i) channels by 100 nM IbTX augmented firing frequency, broadened action potential waveform and reduced after-hyperpolarization. We propose that the BK(i) channels in small diameter DRG sensory neurons might play an important role in regulating nociceptive input to the central nervous system (CNS).


Asunto(s)
Ganglios Espinales/metabolismo , Activación del Canal Iónico , Subunidades beta de los Canales de Potasio de Gran Conductancia Activados por el Calcio/metabolismo , Neuronas/metabolismo , Potasio/metabolismo , Potenciales de Acción , Animales , Calcio/metabolismo , Células Cultivadas , Caribdotoxina/metabolismo , Ganglios Espinales/citología , Ganglios Espinales/efectos de los fármacos , Activación del Canal Iónico/efectos de los fármacos , Cinética , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/metabolismo , Subunidades beta de los Canales de Potasio de Gran Conductancia Activados por el Calcio/antagonistas & inhibidores , Subunidades beta de los Canales de Potasio de Gran Conductancia Activados por el Calcio/genética , Masculino , Neuronas/efectos de los fármacos , Dolor/metabolismo , Técnicas de Placa-Clamp , Péptidos/farmacología , Bloqueadores de los Canales de Potasio/farmacología , Ratas , Ratas Wistar , Factores de Tiempo , Tripsina/metabolismo
14.
J Neurosci ; 26(46): 11833-43, 2006 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-17108156

RESUMEN

Cytosolic N-terminal segments of many K+ channel subunits mediate rapid blockade of ion permeation by physical occlusion of the ion-conducting pore. For some channels with large cytosolic structures, access to the channel pore by inactivation domains may occur through lateral entry pathways or "side portals" that separate the pore domain and associated cytosolic structures covering the axis of the permeation pathway. However, the extent to which side portals control access of molecules to the channel or influence channel gating is unknown. Here we use removal of inactivation by trypsin as a tool to examine basic residue accessibility in both the N terminus of the native auxiliary beta2 subunit of Ca2+-activated, BK-type K+ channels and beta2 subunits with artificial inactivating N termini. The results show that, for BK channels, side portals define a protected space that precedes the channel permeation pathway and excludes small proteins such as trypsin but allows inactivation domains to enter. When channels are closed, inactivation domains readily pass through side portals, with a central antechamber preceding the permeation pathway occupied by an inactivation domain approximately half of the time under resting conditions. The restricted volume of the pathway through side portals is likely to influence kinetic properties of inactivation mechanisms, blockade by large pharmacological probes, and accessibility of modulatory factors to surfaces of the channel within the protected space.


Asunto(s)
Membrana Celular/metabolismo , Canales de Potasio de Gran Conductancia Activados por el Calcio/química , Canales de Potasio de Gran Conductancia Activados por el Calcio/metabolismo , Secuencia de Aminoácidos/fisiología , Animales , Citosol/química , Citosol/metabolismo , Femenino , Activación del Canal Iónico/fisiología , Potenciales de la Membrana/fisiología , Modelos Moleculares , Oocitos/metabolismo , Técnicas de Placa-Clamp , Péptidos/química , Péptidos/metabolismo , Estructura Terciaria de Proteína/fisiología , Tripsina/farmacología , Xenopus laevis
15.
Traffic ; 7(4): 429-39, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16536741

RESUMEN

Three different methods, membrane capacitance (C(m)) measurement, amperometry and FM dye labeling were used to investigate the role of extracellular ATP in insulin secretion from rat pancreatic beta cells. We found that extracellular application of ATP mobilized intracellular Ca(2+) stores and synchronously triggered vigorous exocytosis. No influence of ATP on the readily releasable pool of vesicles was observed, which argues against a direct modulation of the secretory machinery at a level downstream of Ca(2+) elevation. The stimulatory effects of ATP were greatly reduced by intracellular perfusion of BAPTA but not EGTA, suggesting a close spatial association of fusion sites with intracellular Ca(2+) releasing sites. ATP-induced Ca(2+) transients and exocytosis were not blocked by thapsigargin (TG), by a ryanodine receptor antagonist or by dissipation of pH in acidic stores by monensin alone, but they were greatly attenuated by IP(3) receptor inhibition as well as ionomycin plus monensin, suggesting involvement of IP(3)-sensitive acidic Ca(2+) stores. Taken together, our data suggest that extracellular ATP triggers exocytosis by mobilizing spatially limited acidic Ca(2+) stores through IP(3) receptors. This mechanism may explain how insulin secretion from the pancreas is coordinated through diffusible ATP that is co-released with insulin.


Asunto(s)
Adenosina Trifosfato/metabolismo , Calcio/metabolismo , Exocitosis/fisiología , Concentración de Iones de Hidrógeno , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Animales , Cafeína/metabolismo , Canales de Calcio/metabolismo , Células Cultivadas , Dantroleno/metabolismo , Electrofisiología , Inhibidores Enzimáticos/metabolismo , Colorantes Fluorescentes/metabolismo , Receptores de Inositol 1,4,5-Trifosfato , Células Secretoras de Insulina/citología , Masculino , Relajantes Musculares Centrales/metabolismo , Compuestos de Piridinio/metabolismo , Compuestos de Amonio Cuaternario/metabolismo , Ratas , Ratas Wistar , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores Purinérgicos P2/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Tapsigargina/metabolismo
16.
J Biol Chem ; 280(15): 14819-28, 2005 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-15695820

RESUMEN

A novel "long chain" toxin BmP09 has been purified and characterized from the venom of the Chinese scorpion Buthus martensi Karsch. The toxin BmP09 is composed of 66 amino acid residues, including eight cysteines, with a mass of 7721.0 Da. Compared with the B. martensi Karsch AS-1 as a Na(+) channel blocker (7704.8 Da), the BmP09 has an exclusive difference in sequence by an oxidative modification at the C terminus. The sulfoxide Met-66 at the C terminus brought the peptide a dramatic switch from a Na(+) channel blocker toaK(+) channel blocker. Upon probing the targets of the toxin BmP09 on the isolated mouse adrenal medulla chromaffin cells, where a variety of ion channels coexists, we found that the toxin BmP09 specifically blocked large conductance Ca(2+)- and voltage-dependent K(+) channels (BK) but not Na(+) channels at a range of 100 nm concentration. This was further confirmed by blocking directly the BK channels encoded with mSlo1 alpha-subunits in Xenopus oocytes. The half-maximum concentration EC(50) of BmP09 was 27 nm, and the Hill coefficient was 1.8. In outside-out patches, the 100 nm BmP09 reduced approximately 70% currents of BK channels without affecting the single-channel conductance. In comparison with the "short chain" scorpion peptide toxins such as Charybdotoxin, the toxin BmP09 behaves much better in specificity and reversibility, and thus it will be a more efficient tool for studying BK channels. A three-dimensional simulation between a BmP09 toxin and an mSlo channel shows that the Lys-41 in BmP09 lies at the center of the interface and plugs into the entrance of the channel pore. The stable binding between the toxin BmP09 and the BK channel is favored by aromatic pi -pi interactions around the center.


Asunto(s)
Péptidos/química , Canales de Potasio Calcio-Activados/antagonistas & inhibidores , Canales de Potasio Calcio-Activados/química , Venenos de Escorpión/química , Venenos de Escorpión/farmacología , Médula Suprarrenal/citología , Secuencia de Aminoácidos , Animales , Caribdotoxina/química , Cromatografía , Cisteína/química , Disulfuros , Relación Dosis-Respuesta a Droga , Electrofisiología , Enlace de Hidrógeno , Cinética , Canales de Potasio de Gran Conductancia Activados por el Calcio , Lisina/química , Metionina/química , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Mutagénesis , Oocitos/metabolismo , Técnicas de Placa-Clamp , Estructura Terciaria de Proteína , Escorpiones , Homología de Secuencia de Aminoácido , Sodio/química , Sulfóxidos/química , Factores de Tiempo , Xenopus
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA