Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Mar Drugs ; 20(2)2022 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-35200669

RESUMEN

(1) Background: G protein-coupled inward-rectifier potassium (GIRK) channels, especially neuronal GIRK1/2 channels, have been the focus of intense research interest for developing drugs against brain diseases. In this context, venom peptides that selectively activate GIRK channels can be seen as a new source for drug development. Here, we report on the identification and electrophysiological characterization of a novel activator of GIRK1/2 channels, AsKC11, found in the venom of the sea anemone Anemonia sulcata. (2) Methods: AsKC11 was purified from the sea anemone venom by reverse-phase chromatography and the sequence was identified by mass spectrometry. Using the two-electrode voltage-clamp technique, the activity of AsKC11 on GIRK1/2 channels was studied and its selectivity for other potassium channels was investigated. (3) Results: AsKC11, a Kunitz peptide found in the venom of A. sulcata, is the first peptide shown to directly activate neuronal GIRK1/2 channels independent from Gi/o protein activity, without affecting the inward-rectifier potassium channel (IRK1) and with only a minor effect on KV1.6 channels. Thus, AsKC11 is a novel activator of GIRK channels resulting in larger K+ currents because of an increased chord conductance. (4) Conclusions: These discoveries provide new insights into a novel class of GIRK activators.


Asunto(s)
Venenos de Cnidarios/química , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/efectos de los fármacos , Péptidos/farmacología , Animales , Cromatografía de Fase Inversa , Femenino , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/metabolismo , Espectrometría de Masas , Técnicas de Placa-Clamp , Péptidos/química , Péptidos/aislamiento & purificación , Xenopus laevis
2.
Molecules ; 26(5)2021 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-33670972

RESUMEN

Animal venoms are considered as a promising source of new drugs. Sea anemones release polypeptides that affect electrical activity of neurons of their prey. Voltage dependent sodium (Nav) channels are the common targets of Av1, Av2, and Av3 toxins from Anemonia viridis and CgNa from Condylactis gigantea. The toxins bind to the extracellular side of a channel and slow its fast inactivation, but molecular details of the binding modes are not known. Electrophysiological measurements on Periplaneta americana neuronal preparation revealed differences in potency of these toxins to increase nerve activity. Av1 and CgNa exhibit the strongest effects, while Av2 the weakest effect. Extensive molecular docking using a modern SMINA computer method revealed only partial overlap among the sets of toxins' and channel's amino acid residues responsible for the selectivity and binding modes. Docking positions support earlier supposition that the higher neuronal activity observed in electrophysiology should be attributed to hampering the fast inactivation gate by interactions of an anemone toxin with the voltage driven S4 helix from domain IV of cockroach Nav channel (NavPaS). Our modelling provides new data linking activity of toxins with their mode of binding in site 3 of NavPaS channel.


Asunto(s)
Péptidos/química , Canales de Sodio/química , Canales de Sodio/metabolismo , Ponzoñas/química , Secuencia de Aminoácidos , Aminoácidos/química , Animales , Sitios de Unión , Cucarachas , Fenómenos Electrofisiológicos , Conformación Molecular , Simulación del Acoplamiento Molecular , Neuronas/efectos de los fármacos , Anémonas de Mar
3.
Mar Drugs ; 15(9)2017 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-28902151

RESUMEN

The human ether-à-go-go channel (hEag1 or KV10.1) is a cancer-relevant voltage-gated potassium channel that is overexpressed in a majority of human tumors. Peptides that are able to selectively inhibit this channel can be lead compounds in the search for new anticancer drugs. Here, we report the activity-guided purification and electrophysiological characterization of a novel KV10.1 inhibitor from the sea anemone Anthopleura elegantissima. Purified sea anemone fractions were screened for inhibitory activity on KV10.1 by measuring whole-cell currents as expressed in Xenopus laevis oocytes using the two-microelectrode voltage clamp technique. Fractions that showed activity on Kv10.1 were further purified by RP-HPLC. The amino acid sequence of the peptide was determined by a combination of MALDI- LIFT-TOF/TOF MS/MS and CID-ESI-FT-ICR MS/MS and showed a high similarity with APETx1 and APETx3 and was therefore named APETx4. Subsequently, the peptide was electrophysiologically characterized on KV10.1. The selectivity of the toxin was investigated on an array of voltage-gated ion channels, including the cardiac human ether-à-go-go-related gene potassium channel (hERG or Kv11.1). The toxin inhibits KV10.1 with an IC50 value of 1.1 µM. In the presence of a similar toxin concentration, a shift of the activation curve towards more positive potentials was observed. Similar to the effect of the gating modifier toxin APETx1 on hERG, the inhibition of Kv10.1 by the isolated toxin is reduced at more positive voltages and the peptide seems to keep the channel in a closed state. Although the peptide also induces inhibitory effects on other KV and NaV channels, it exhibits no significant effect on hERG. Moreover, APETx4 induces a concentration-dependent cytotoxic and proapoptotic effect in various cancerous and noncancerous cell lines. This newly identified KV10.1 inhibitor can be used as a tool to further characterize the oncogenic channel KV10.1 or as a scaffold for the design and synthesis of more potent and safer anticancer drugs.


Asunto(s)
Antineoplásicos/farmacología , Venenos de Cnidarios/farmacología , Canales de Potasio con Entrada de Voltaje/antagonistas & inhibidores , Anémonas de Mar , Toxinas Biológicas/farmacología , Animales , Línea Celular Tumoral/efectos de los fármacos , Concentración 50 Inhibidora , Oocitos/efectos de los fármacos , Xenopus
4.
Mol Cancer ; 12: 12, 2013 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-23409748

RESUMEN

OBJECTIVES: Palytoxin (PTX), a marine toxin isolated from the Cnidaria (zooanthid) Palythoa caribaeorum is one of the most potent non-protein substances known. It is a very complex molecule that presents both lipophilic and hydrophilic areas. The effect of PTX was investigated in a series of experiments conducted in head and neck squamous cell carcinoma (HNSCC) cell lines and xenografts. MATERIALS AND METHODS: Cell viability, and gene expression of the sodium/potassium-transporting ATPase subumit alpha1 (ATP1AL1) and GAPDH were analyzed in HNSCC cells and normal epithelial cells after treatment with PTX using cytotoxicity-, clonogenic-, and enzyme inhibitor assays as well as RT-PCR and Northern Blotting. For xenograft experiments severe combined immunodeficient (SCID) mice were used to analyze tumor regression. The data were statistically analyzed using One-Way Annova (SPSS vs20). RESULTS: Significant toxic effects were observed in tumor cells treated with PTX (LD50 of 1.5 to 3.5 ng/ml) in contrast to normal cells. In tumor cells PTX affected both the release of LDH and the expression of the sodium/potassium-transporting ATPase subunit alpha1 gene suggesting loss of cellular integrity, primarily of the plasma membrane. Furthermore, strong repression of the c-Jun N-terminal kinase 3 (JNK3) mRNA expression was found in carcinoma cells which correlated with enhanced toxicity of PTX suggesting an essential role of the mitogen activated protein kinase (MAPK)/JNK signalling cascades pathway in the mechanisms of HNSCC cell resistance to PTX. In mice inoculated with carcinoma cells, injections of PTX into the xenografted tumors resulted within 24 days in extensive tumor destruction in 75% of the treated animals (LD50 of 68 ng/kg to 83 ng/kg) while no tumor regression occurred in control animals. CONCLUSIONS: These results clearly provide evidence that PTX possesses preferential toxicity for head and neck carcinoma cells and therefore it is worth further studying its impact which may extend our knowledge of the biology of head and neck cancer.


Asunto(s)
Acrilamidas/farmacología , Antineoplásicos/farmacología , Carcinoma de Células Escamosas/tratamiento farmacológico , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Proteína Quinasa 10 Activada por Mitógenos/metabolismo , Pirazoles/farmacología , Urea/análogos & derivados , Urea/farmacología , Acrilamidas/administración & dosificación , Animales , Antineoplásicos/administración & dosificación , Carcinoma de Células Escamosas/enzimología , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Forma de la Célula/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Venenos de Cnidarios , Sinergismo Farmacológico , Expresión Génica/efectos de los fármacos , ATPasa Intercambiadora de Hidrógeno-Potásio/genética , ATPasa Intercambiadora de Hidrógeno-Potásio/metabolismo , Neoplasias de Cabeza y Cuello/enzimología , Neoplasias de Cabeza y Cuello/patología , Humanos , Concentración 50 Inhibidora , Inyecciones Intralesiones , Inyecciones Intraperitoneales , Ratones , Ratones SCID , Proteína Quinasa 10 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 10 Activada por Mitógenos/genética , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
5.
FASEB J ; 26(12): 5141-51, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22972919

RESUMEN

APETx3, a novel peptide isolated from the sea anemone Anthopleura elegantissima, is a naturally occurring mutant from APETx1, only differing by a Thr to Pro substitution at position 3. APETx1 is believed to be a selective modulator of human ether-á-go-go related gene (hERG) potassium channels with a K(d) of 34 nM. In this study, APETx1, 2, and 3 have been subjected to an electrophysiological screening on a wide range of 24 ion channels expressed in Xenopus laevis oocytes: 10 cloned voltage-gated sodium channels (Na(V) 1.2-Na(V)1.8, the insect channels DmNa(V)1, BgNa(V)1-1a, and the arachnid channel VdNa(V)1) and 14 cloned voltage-gated potassium channels (K(V)1.1-K(V)1.6, K(V)2.1, K(V)3.1, K(V)4.2, K(V)4.3, K(V)7.2, K(V)7.4, hERG, and the insect channel Shaker IR). Surprisingly, the Thr3Pro substitution results in a complete abolishment of APETx3 modulation on hERG channels and provides this toxin the ability to become a potent (EC(50) 276 nM) modulator of voltage-gated sodium channels (Na(V)s) because it slows down the inactivation of mammalian and insect Na(V) channels. Our study also shows that the homologous toxins APETx1 and APETx2 display promiscuous properties since they are also capable of recognizing Na(V) channels with IC(50) values of 31 nM and 114 nM, respectively, causing an inhibition of the sodium conductance without affecting the inactivation. Our results provide new insights in key residues that allow these sea anemone toxins to recognize distinct ion channels with similar potency but with different modulatory effects. Furthermore, we describe for the first time the target promiscuity of a family of sea anemone toxins thus far believed to be highly selective.


Asunto(s)
Activación del Canal Iónico/efectos de los fármacos , Mutación Puntual , Anémonas de Mar/metabolismo , Toxinas Biológicas/farmacología , Animales , Venenos de Cnidarios/genética , Venenos de Cnidarios/metabolismo , Venenos de Cnidarios/farmacología , Relación Dosis-Respuesta a Droga , Electrofisiología , Femenino , Humanos , Insectos/genética , Insectos/metabolismo , Activación del Canal Iónico/fisiología , Oocitos/efectos de los fármacos , Oocitos/metabolismo , Oocitos/fisiología , Canales de Potasio/genética , Canales de Potasio/metabolismo , Canales de Potasio/fisiología , Anémonas de Mar/genética , Canales de Sodio/genética , Canales de Sodio/metabolismo , Canales de Sodio/fisiología , Toxinas Biológicas/genética , Toxinas Biológicas/metabolismo , Xenopus laevis
6.
Biochem J ; 406(1): 67-76, 2007 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-17506725

RESUMEN

CgNa (Condylactis gigantea neurotoxin) is a 47-amino-acid- residue toxin from the giant Caribbean sea anemone Condylactis gigantea. The structure of CgNa, which was solved by 1H-NMR spectroscopy, is somewhat atypical and displays significant homology with both type I and II anemone toxins. CgNa also displays a considerable number of exceptions to the canonical structural elements that are thought to be essential for the activity of this group of toxins. Furthermore, unique residues in CgNa define a characteristic structure with strong negatively charged surface patches. These patches disrupt a surface-exposed cluster of hydrophobic residues present in all anemone-derived toxins described to date. A thorough characterization by patch-clamp analysis using rat DRG (dorsal root ganglion) neurons indicated that CgNa preferentially binds to TTX-S (tetrodotoxin-sensitive) voltage-gated sodium channels in the resting state. This association increased the inactivation time constant and the rate of recovery from inactivation, inducing a significant shift in the steady state of inactivation curve to the left. The specific structural features of CgNa may explain its weaker inhibitory capacity when compared with the other type I and II anemone toxins.


Asunto(s)
Venenos de Cnidarios/química , Venenos de Cnidarios/metabolismo , Anémonas de Mar/química , Secuencia de Aminoácidos , Animales , Región del Caribe , Venenos de Cnidarios/clasificación , Venenos de Cnidarios/farmacología , Electrofisiología , Ganglios Espinales/efectos de los fármacos , Activación del Canal Iónico/efectos de los fármacos , Canales Iónicos/metabolismo , Modelos Moleculares , Datos de Secuencia Molecular , Neuronas/efectos de los fármacos , Resonancia Magnética Nuclear Biomolecular , Péptidos/química , Estructura Secundaria de Proteína , Ratas , Ratas Wistar , Soluciones , Tetrodotoxina/farmacología , Factores de Tiempo
7.
Toxicon ; 48(2): 211-20, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16814340

RESUMEN

A new peptide toxin exhibiting a molecular weight of 5043Da (av.) and comprising 47 amino acid residues was isolated from the sea anemone Condylactis gigantea. Purification of the peptide was achieved by a multistep chromatographic procedure monitoring its strong paralytic activity on crustacea (LD(50) approx. 1microg/kg). Complete sequence analysis of the toxic peptide revealed the isolation of a new member of type I sea anemone sodium channel toxins containing the typical pattern of the six cysteine residues. From 11kg of wet starting material, approximately 1g of the peptide toxin was isolated. The physiological action of the new toxin from C. gigantea CgNa was investigated on sodium currents of rat dorsal root ganglion neurons in culture using whole-cell patch clamp technique (n=60). Under current clamp condition (CgNa) increased action potential duration. This effect is due to slowing down of the TTX-S sodium current inactivation, without modifying the activation process. CgNa prolonged the cardiac action potential duration and enhanced contractile force albeit at 100-fold higher concentrations than the Anemonia sulcata toxin ATXII. The action on sodium channel inactivation and on cardiac excitation-contraction coupling resemble previous results with compounds obtained from this and other sea anemones [Shapiro, B.I., 1968. Purification of a toxin from tentacles of the anemone C. gigantea. Toxicon 5, 253-259; Pelhate, M., Zlotkin, E., 1982. Actions of insect toxin and other toxins derived from the venom of scorpion Androtonus australis on isolated giant axons of the cockroach Periplaneta americana. J. Exp. Biol. 97, 67-77; Salgado, V., Kem, W., 1992. Actions of three structurally distinct sea anemone toxins on crustacean and insect sodium channels. Toxicon 30, 1365-1381; Bruhn, T., Schaller, C., Schulze, C., Sanchez-Rodriquez, J., Dannmeier, C., Ravens, U., Heubach, J.F., Eckhardt, K., Schmidtmayer, J., Schmidt, H., Aneiros, A., Wachter, E., Béress, L., 2001. Isolation and characterization of 5 neurotoxic and cardiotoxic polypeptides from the sea anemone Anthopleura elegantissima. Toxicon, 39, 693-702]. Comprehensive analysis of the purified active fractions suggests that CgNa may represent the main peptide toxin of this sea anemone species.


Asunto(s)
Venenos de Cnidarios/química , Toxinas Marinas/toxicidad , Anémonas de Mar/metabolismo , Bloqueadores de los Canales de Sodio/toxicidad , Secuencia de Aminoácidos , Animales , Braquiuros/efectos de los fármacos , Braquiuros/fisiología , Células Cultivadas , Relación Dosis-Respuesta a Droga , Cobayas , Masculino , Toxinas Marinas/química , Toxinas Marinas/aislamiento & purificación , Datos de Secuencia Molecular , Peso Molecular , Contracción Muscular/efectos de los fármacos , Contracción Muscular/fisiología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Músculos Papilares/efectos de los fármacos , Músculos Papilares/fisiopatología , Parálisis/inducido químicamente , Parálisis/fisiopatología , Técnicas de Placa-Clamp , Bloqueadores de los Canales de Sodio/química , Bloqueadores de los Canales de Sodio/aislamiento & purificación , Canales de Sodio/efectos de los fármacos
8.
Artículo en Inglés | MEDLINE | ID: mdl-22824729

RESUMEN

Sea anemones are sources of biologically active proteins and peptides. However, up to date few peptidomic studies of these organisms are known; therefore most species and their peptide diversity remain unexplored. Contrasting to previous venom peptidomic works on sea anemones and other venomous animals, in the present study we combined pH gradient ion-exchange chromatography with gel filtration and reversed-phase chromatography, allowing the separation of the 1-10 kDa polypeptides from the secretion of the unexplored sea anemone Phymanthus crucifer (Cnidaria/Phymanthidae). This multidimensional chromatographic approach followed by MALDI-TOF-MS detection generated a peptide fingerprint comprising 504 different molecular mass values from acidic and basic peptides, being the largest number estimated for a sea anemone exudate. The peptide population within the 2.0-3.5 kDa mass range showed the highest frequency whereas the main biomarkers comprised acidic and basic peptides with molecular masses within 2.5-6.9 kDa, in contrast to the homogeneous group of 4-5 kDa biomarkers found in sea anemones such as B. granulifera and B. cangicum (Cnidaria/Actiniidae). Our study shows that sea anemone peptide fingerprinting can be greatly improved by including pH gradient ion-exchange chromatography into the multidimensional separation approach, complemented by MALDI-TOF-MS detection. This strategy allowed us to find the most abundant and unprecedented diversity of secreted components from a sea anemone exudate, indicating that the search for novel biologically active peptides from these organisms has much greater potential than previously predicted.


Asunto(s)
Cromatografía por Intercambio Iónico/métodos , Mapeo Peptídico/métodos , Péptidos/análisis , Péptidos/química , Anémonas de Mar/metabolismo , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción/métodos , Animales , Cromatografía en Gel , Concentración de Iones de Hidrógeno , Interacciones Hidrofóbicas e Hidrofílicas , Toxinas Marinas/análisis , Toxinas Marinas/química , Peso Molecular
9.
Biochem Pharmacol ; 82(1): 81-90, 2011 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-21477583

RESUMEN

Sea anemone venom is a known source of interesting bioactive compounds, including peptide toxins which are invaluable tools for studying structure and function of voltage-gated potassium channels. APEKTx1 is a novel peptide isolated from the sea anemone Anthopleura elegantissima, containing 63 amino acids cross-linked by 3 disulfide bridges. Sequence alignment reveals that APEKTx1 is a new member of the type 2 sea anemone peptides targeting voltage-gated potassium channels (K(V)s), which also include the kalicludines from Anemonia sulcata. Similar to the kalicludines, APEKTx1 shares structural homology with both the basic pancreatic trypsin inhibitor (BPTI), a very potent Kunitz-type protease inhibitor, and dendrotoxins which are powerful blockers of voltage-gated potassium channels. In this study, APEKTx1 has been subjected to a screening on a wide range of 23 ion channels expressed in Xenopus laevis oocytes: 13 cloned voltage-gated potassium channels (K(V)1.1-K(V)1.6, K(V)1.1 triple mutant, K(V)2.1, K(V)3.1, K(V)4.2, K(V)4.3, hERG, the insect channel Shaker IR), 2 cloned hyperpolarization-activated cyclic nucleotide-sensitive cation non-selective channels (HCN1 and HCN2) and 8 cloned voltage-gated sodium channels (Na(V)1.2-Na(V)1.8 and the insect channel DmNa(V)1). Our data show that APEKTx1 selectively blocks K(V)1.1 channels in a very potent manner with an IC(50) value of 0.9nM. Furthermore, we compared the trypsin inhibitory activity of this toxin with BPTI. APEKTx1 inhibits trypsin with a dissociation constant of 124nM. In conclusion, this study demonstrates that APEKTx1 has the unique feature to combine the dual functionality of a potent and selective blocker of K(V)1.1 channels with that of a competitive inhibitor of trypsin.


Asunto(s)
Venenos de Cnidarios/farmacología , Péptidos/farmacología , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio con Entrada de Voltaje/antagonistas & inhibidores , Inhibidores de Proteasas/farmacología , Anémonas de Mar/fisiología , Secuencia de Aminoácidos , Animales , Venenos de Cnidarios/química , Venenos de Cnidarios/aislamiento & purificación , Datos de Secuencia Molecular , Oocitos/efectos de los fármacos , Oocitos/fisiología , Péptidos/química , Péptidos/aislamiento & purificación , Bloqueadores de los Canales de Potasio/química , Bloqueadores de los Canales de Potasio/aislamiento & purificación , Inhibidores de Proteasas/química , Inhibidores de Proteasas/aislamiento & purificación , Alineación de Secuencia , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción/métodos , Xenopus
10.
Front Pharmacol ; 1: 133, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-21833172

RESUMEN

Because of their prominent role in electro-excitability, voltage-gated sodium (Na(V)) channels have become the foremost important target of animal toxins. These toxins have developed the ability to discriminate between closely related Na(V) subtypes, making them powerful tools to study Na(V) channel function and structure. CgNa is a 47-amino acid residue type I toxin isolated from the venom of the Giant Caribbean Sea Anemone Condylactis gigantea. Previous studies showed that this toxin slows the fast inactivation of tetrodotoxin-sensitive Na(V) currents in rat dorsal root ganglion neurons. To illuminate the underlying Na(V) subtype-selectivity pattern, we have assayed the effects of CgNa on a broad range of mammalian isoforms (Na(V)1.2-Na(V)1.8) expressed in Xenopus oocytes. This study demonstrates that CgNa selectively slows the fast inactivation of rNa(V)1.3/ß(1), mNa(V)1.6/ß(1) and, to a lesser extent, hNa(V)1.5/ß(1), while the other mammalian isoforms remain unaffected. Importantly, CgNa was also examined on the insect sodium channel DmNa(V)1/tipE, revealing a clear phyla-selectivity in the efficacious actions of the toxin. CgNa strongly inhibits the inactivation of the insect Na(V) channel, resulting in a dramatic increase in peak current amplitude and complete removal of fast and steady-state inactivation. Together with the previously determined solution structure, the subtype-selective effects revealed in this study make of CgNa an interesting pharmacological probe to investigate the functional role of specific Na(V) channel subtypes. Moreover, further structural studies could provide important information on the molecular mechanism of Na(V) channel inactivation.

11.
Mol Pharmacol ; 64(1): 59-69, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12815161

RESUMEN

A new peptide, APETx1, which specifically inhibits human ether-a-go-go-related gene (HERG) channels, was purified from venom of the sea anemone Anthopleura elegantissima. APETx1 is a 42-amino acid peptide cross-linked by three disulfide bridges and shares 54% homology with BDS-I, another sea anemone K+ channel inhibitor. Although they differ in their specific targets, circular dichroism spectra and molecular modeling indicate that APETx1 and BDS-I have a common molecular scaffold and belong to the same structural family of K+ channel blocking peptides. APETx1 inhibits HERG currents in a heterologous system with an IC50 value of 34 nM by modifying the voltage dependence of the channel gating. Central injections in mice failed to induce any neurotoxic symptoms. APETx1, which has no sequence homologies with scorpion toxins acting on HERG, defines a new structural group of HERG gating modifiers isolated from a sea anemone.


Asunto(s)
Venenos de Cnidarios/farmacología , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio con Entrada de Voltaje/antagonistas & inhibidores , Canales de Potasio/química , Anémonas de Mar/química , Secuencia de Aminoácidos , Animales , Dicroismo Circular , Venenos de Cnidarios/química , Venenos de Cnidarios/genética , Canal de Potasio ERG1 , Canales de Potasio Éter-A-Go-Go , Humanos , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Conformación Proteica , Homología de Secuencia de Aminoácido
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA