Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Front Pharmacol ; 13: 881732, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35754473

RESUMEN

A novel 4/8 subtype α-conotoxin, Vt1.27 (NCCMFHTCPIDYSRFNC-NH2), was identified from Conus vitulinus in the South China Sea by RACE methods. The peptide was synthesized and structurally characterized. Similar to other α-conotoxins that target neuronal nicotinic acetylcholine receptor (nAChR) subtypes, Vt1.27 inhibited the rat α3ß2 nAChR subtype (IC50 = 1160 nM) and was inactive at voltage-gated sodium and potassium channels in rat sensory neurons. However, Vt1.27 inhibited high voltage-activated N-type (CaV2.2) calcium channels expressed in HEK293T cells with an IC50 of 398 nM. An alanine scan of the peptide showed that residues Phe5, Pro9, Ile10, and Ser13 contribute significantly to the inhibitory activity of Vt1.27. The molecular dockings indicate that Vt1.27 inhibits the transmembrane region of CaV2.2, which is different from that of ω-conotoxins. Furthermore, Vt1.27 exhibited potent anti-allodynic effect in rat partial sciatic nerve injury (PNL) and chronic constriction injury (CCI) pain models at 10 nmol/kg level with the intramuscular injection. The pain threshold elevation of Vt1.27 groups was higher than that of α-conotoxin Vc1.1 in CCI rat models. These findings expand our knowledge of targets of α-conotoxins and potentially provide a potent, anti-allodynic peptide for the treatment of neuropathic pain.

2.
J Neurochem ; 159(1): 90-100, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34008858

RESUMEN

α-Conotoxins are small disulfide-rich peptides found in the venom of marine cone snails and are potent antagonists of nicotinic acetylcholine receptors (nAChRs). They are valuable pharmacological tools and have potential therapeutic applications for the treatment of chronic pain or neurological diseases and disorders. In the present study, we synthesized and functionally characterized a novel α-conotoxin Bt1.8, which was cloned from Conus betulinus. Bt1.8 selectively inhibited ACh-evoked currents in Xenopus oocytes expressing rat(r) α6/α3ß2ß3 and rα3ß2 nAChRs with an IC50 of 2.1 nM and 9.4 nM, respectively, and similar potency for human (h) α6/α3ß2ß3 and hα3ß2 nAChRs. Additionally, Bt1.8 had higher binding affinity with a slower dissociation rate for the rα6/α3ß2ß3 subtype compared to rα3ß2. The amino acid sequence of Bt1.8 is significantly different from other reported α-conotoxins targeting the two nAChR subtypes. Further Alanine scanning analyses demonstrated that residues Ile9, Leu10, Asn11, Asn12 and Asn14 are critical for its inhibitory activity at the α6/α3ß2ß3 and α3ß2 subtypes. Moreover, the NMR structure of Bt1.8 indicated the presence of a relatively larger hydrophobic zone than other α4/7-conotoxins which may explain its potent inhibition at α6/α3ß2ß3 nAChRs.


Asunto(s)
Conotoxinas/farmacología , Antagonistas Nicotínicos/farmacología , Receptores Nicotínicos/metabolismo , Animales , Conotoxinas/química , Conotoxinas/aislamiento & purificación , Caracol Conus , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Antagonistas Nicotínicos/química , Antagonistas Nicotínicos/aislamiento & purificación , Oocitos , Estructura Terciaria de Proteína , Subunidades de Proteína/antagonistas & inhibidores , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Ratas , Receptores Nicotínicos/genética , Xenopus laevis
3.
Mar Drugs ; 19(1)2021 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-33478061

RESUMEN

N-methyl-D-aspartate receptor (NMDAR) antagonists have been found to be effective to inhibit morphine dependence. However, the discovery of the selective antagonist for NMDAR GluN2B with low side-effects still remains challenging. In the present study, we report a selective NMDAR GluN2B antagonist con-T[M8Q](a conantokin-T variant) that potently inhibits the naloxone-induced jumping and conditioned place preference of morphine-dependent mice at nmol/kg level, 100-fold higher than ifenprodil, a classical NMDAR NR2B antagonist. Con-T[M8Q] displays no significant impacts on coordinated locomotion function, spontaneous locomotor activity, and spatial memory mice motor function at the dose used. Further molecular mechanism experiments demonstrate that con-T[M8Q] effectively inhibited the transcription and expression levels of signaling molecules related to NMDAR NR2B subunit in hippocampus, including NR2B, p-NR2B, CaMKII-α, CaMKII-ß, CaMKIV, pERK, and c-fos. The high efficacy and low side effects of con-T[M8Q] make it a good lead compound for the treatment of opiate dependence and for the reduction of morphine usage.


Asunto(s)
Conotoxinas/farmacología , Antagonistas de Aminoácidos Excitadores/farmacología , Dependencia de Morfina/tratamiento farmacológico , Receptores de N-Metil-D-Aspartato/efectos de los fármacos , Animales , Conotoxinas/administración & dosificación , Conotoxinas/toxicidad , Modelos Animales de Enfermedad , Antagonistas de Aminoácidos Excitadores/administración & dosificación , Antagonistas de Aminoácidos Excitadores/toxicidad , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Locomoción/efectos de los fármacos , Masculino , Ratones , Dependencia de Morfina/fisiopatología , Naloxona/farmacología , Piperidinas/farmacología , Memoria Espacial/efectos de los fármacos
4.
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
5.
J Med Chem ; 61(22): 10198-10205, 2018 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-30358401

RESUMEN

α-Conotoxins exhibiting analgesic activity, such as Vc1.1, have been shown to inhibit α9α10 nicotinic acetylcholine receptors (nAChRs) and GABAB-receptor (GABABR) coupled N-type (CaV2.2) calcium channels. Here, we report two Vc1.1 variants, Vc1.1[N9R] and benzoyl-Vc1.1[N9R], that selectively inhibit CaV2.2 channels via GABABR activation but exhibit reduced inhibitory activity at α9α10 and other neuronal nAChR subtypes compared with Vc1.1. Surprisingly, the analgesic activity of Vc1.1[N9R] and benzoyl-Vc1.1[N9R] was more potent than that of Vc1.1 when tested in partial sciatic nerve ligation injury and chronic constriction injury models. Vc1.1[N9R] and benzoyl-Vc1.1[N9R] exhibited either similar or tenfold higher activity of GABABR-mediated CaV2.2 inhibition but no activity at CaV2.2 alone; however, the mechanism of increased analgesic activity is unknown. The effects on analgesic activity and α9α10 nAChR of other Vc1.1 variations at position 9 and the N-terminus were also determined. Our findings provide new insights for designing potent inhibitors for GABABR-coupled N-type (CaV2.2) calcium channels.


Asunto(s)
Analgésicos/química , Analgésicos/farmacología , Canales de Calcio Tipo N/metabolismo , Conotoxinas/química , Conotoxinas/farmacología , Receptores de GABA-B/metabolismo , Animales , Bloqueadores de los Canales de Calcio/química , Bloqueadores de los Canales de Calcio/farmacología , Descubrimiento de Drogas , Células HEK293 , Humanos , Masculino , Ratas , Ratas Sprague-Dawley , Relación Estructura-Actividad
6.
Mar Drugs ; 16(4)2018 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-29614714

RESUMEN

α-Conotoxins (α-CTxs) are small peptides composed of 11 to 20 amino acid residues with two disulfide bridges. Most of them potently and selectively target nicotinic acetylcholine receptor (nAChR) subtypes, and a few were found to inhibit the GABAB receptor (GABABR)-coupled N-type calcium channels (Cav2.2). However, in all of α-CTxs targeting both receptors, the disulfide connectivity arrangement "C¹-C³, C²-C4" is present. In this work, a novel α4/7-CTx named Lt1.3 (GCCSHPACSGNNPYFC-NH2) was cloned from the venom ducts of Conus litteratus (C. litteratus) in the South China Sea. Lt1.3 was then chemically synthesized and two isomers with disulfide bridges "C¹-C³, C²-C4" and "C¹-C4, C²-C³" were found and functionally characterized. Electrophysiological experiments showed that Lt1.3 containing the common disulfide bridges "C¹-C³, C²-C4" potently and selectively inhibited α3ß2 nAChRs and not GABABR-coupled Cav2.2. Surprisingly, but the isomer with the disulfide bridges "C¹-C4, C²-C³" showed exactly the opposite inhibitory activity, inhibiting only GABABR-coupled Cav2.2 and not α3ß2 nAChRs. These findings expand the knowledge of the targets and selectivity of α-CTxs and provide a new structural motif to inhibit the GABABR-coupled Cav2.2.


Asunto(s)
Bloqueadores de los Canales de Calcio/farmacología , Conotoxinas/farmacología , Caracol Conus/química , Antagonistas Nicotínicos/farmacología , Receptores Nicotínicos/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Bloqueadores de los Canales de Calcio/química , Canales de Calcio Tipo N/metabolismo , Conotoxinas/química , Células HEK293 , Humanos , Concentración 50 Inhibidora , Antagonistas Nicotínicos/química , Océanos y Mares , Oocitos , Técnicas de Placa-Clamp , Relación Estructura-Actividad , Xenopus
7.
Sci Rep ; 8(1): 1004, 2018 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-29343689

RESUMEN

We here describe a novel α-conopeptide, Eu1.6 from Conus eburneus, which exhibits strong anti-nociceptive activity by an unexpected mechanism of action. Unlike other α-conopeptides that largely target nicotinic acetylcholine receptors (nAChRs), Eu1.6 displayed only weak inhibitory activity at the α3ß4 and α7 nAChR subtypes and TTX-resistant sodium channels, and no activity at TTX-sensitive sodium channels in rat dorsal root ganglion (DRG) neurons, or opiate receptors, VR1, KCNQ1, L- and T-type calcium channels expressed in HEK293 cells. However, Eu1.6 inhibited high voltage-activated N-type calcium channel currents in isolated mouse DRG neurons which was independent of GABAB receptor activation. In HEK293 cells expressing CaV2.2 channels alone, Eu1.6 reversibly inhibited depolarization-activated Ba2+ currents in a voltage- and state-dependent manner. Inhibition of CaV2.2 by Eu1.6 was concentration-dependent (IC50 ~1 nM). Significantly, systemic administration of Eu1.6 at doses of 2.5-5.0 µg/kg exhibited potent analgesic activities in rat partial sciatic nerve injury and chronic constriction injury pain models. Furthermore, Eu1.6 had no significant side-effect on spontaneous locomotor activity, cardiac and respiratory function, and drug dependence in mice. These findings suggest α-conopeptide Eu1.6 is a potent analgesic for the treatment of neuropathic and chronic pain and opens a novel option for future analgesic drug design.


Asunto(s)
Analgésicos/farmacología , Canales de Calcio Tipo N/metabolismo , Dolor Crónico/tratamiento farmacológico , Conotoxinas/farmacología , Péptidos/farmacología , Neuropatía Ciática/tratamiento farmacológico , Secuencia de Aminoácidos , Analgésicos/síntesis química , Analgésicos/aislamiento & purificación , Animales , Calcio/metabolismo , Dolor Crónico/metabolismo , Dolor Crónico/fisiopatología , Conotoxinas/síntesis química , Conotoxinas/aislamiento & purificación , Caracol Conus/química , Ganglios Espinales/citología , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/metabolismo , Células HEK293 , Humanos , Inyecciones Intramusculares , Inyecciones Intravenosas , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/citología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Oocitos/efectos de los fármacos , Oocitos/metabolismo , Técnicas de Placa-Clamp , Péptidos/síntesis química , Péptidos/aislamiento & purificación , Ratas , Ratas Sprague-Dawley , Neuropatía Ciática/metabolismo , Neuropatía Ciática/fisiopatología , Técnicas de Síntesis en Fase Sólida , Xenopus laevis
8.
Peptides ; 94: 64-70, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28666820

RESUMEN

As a class of peptides with 10 cysteine residues (-C-CC-C-CC-C-C-C-C-), D-superfamily conotoxins (D-conotoxins) can specifically act on nicotinic acetylcholine receptors (nAChRs). According to the conserved signal peptides of D-conotoxins, seven D-conotoxin precursor sequences with a previously undescribed Cys arrangement (-C-C-C-CC-C-C-C-C-C-) were identified by PCR-RACE methodology in the present study. The alignment of sequences revealed that signal peptide regions were same as D-VxXXA from Conus vexillum, and their mature peptides were almost different from the D-conotoxins. Analyses of the evolutionary tree demonstrated that they had low homology to those reported conotoxins with 10 cysteine residues (less than 35%) and lied in a separate branch in the evolutionary tree. Furthermore, a previously undescribed D-superfamily conotoxin Lt28.1 was further expressed in Pichia pastoris and then functionally characterized. The results showed that the recombinant Lt28.1 targeted α9α10 nAChRs but not other nAChRs subtypes. These findings defined a new branch of D-superfamily and expanded our knowledge of targets and potential application of D-conotoxins.


Asunto(s)
Conotoxinas/metabolismo , Caracol Conus/metabolismo , Secuencia de Aminoácidos , Animales , Clonación Molecular , Conotoxinas/genética , Caracol Conus/química , Filogenia , Pichia/genética , Análisis de Secuencia de Proteína
9.
Peptides ; 81: 15-20, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27131596

RESUMEN

In the present study, we isolated, synthesized and NMR structurally characterized a novel conopeptide Im10A consisting of 11 amino acids (NTICCEGCMCY-NH2) from Conus imperialis. Unlike other conopeptides with four cysteine residues, Im10A had only two residues in loop 1 and one residue in loop 2 (CC-loop1-C-loop2-C), which formed a stable disulfide connectivity "I-IV, II- III" (framework X) with a type I ß-turn. Interestingly, Im10A exhibited 50.7% analgesic activity on rat partial sciatic nerve ligation (PNL) at 2h after Im10A administration. However, 10µM Im10A exhibited no apparent effect on neuronal nicotinic acetylcholine receptor, and it did not target DRG voltage-dependent sodium, potassium and calcium ion channels and opioid receptor. To our knowledge, Im10A had the most concentrated disulfide bridges among conopeptides with four cysteine residues. This finding provided a new motif for the future development of biomimetic compounds.


Asunto(s)
Analgésicos/química , Analgésicos/farmacología , Conotoxinas/química , Conotoxinas/farmacología , Caracol Conus/química , Secuencia de Aminoácidos , Analgésicos/síntesis química , Analgésicos/aislamiento & purificación , Análisis de Varianza , Animales , Conotoxinas/síntesis química , Conotoxinas/aislamiento & purificación , Cisteína/química , Disulfuros/química , Masculino , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Resonancia Magnética Nuclear Biomolecular , Oligopéptidos/síntesis química , Oligopéptidos/química , Oligopéptidos/aislamiento & purificación , Oligopéptidos/farmacología , Ratas , Ratas Sprague-Dawley , Receptores Nicotínicos/metabolismo , Nervio Ciático/efectos de los fármacos
10.
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
11.
Acta Biochim Biophys Sin (Shanghai) ; 47(12): 1023-8, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26511093

RESUMEN

Conotoxins (CTxs) are typically small peptides composed of 12-50 amino acid residues with 2-5 disulfide bridges. Most of them potently and selectively target a wide variety of ion channels and membrane receptors. They are highly valued as neuropharmacological probes and in pharmaceutical development. In this work, a novel α4/6-CTx named ViIA (RDCCSNPPCAHNNPDC-NH2) was identified from a cDNA library of the venom ducts of Conus virgo (C. virgo). ViIA was then synthesized chemically and its disulfide connectivity was identified as 'C(1)-C(3), C(2)-C(4)'. Its molecular targets were further assessed using two-electrode voltage clamping. The results indicated that ViIA selectively inhibited nicotinic acetylcholine receptor (nAChR) α3ß2 subtype with an IC50 of 845.5 nM, but did not target dorsal root ganglion sodium (Na(+))-, potassium (K(+))- or calcium (Ca(2+))-ion channels. Further structure-activity relationship analysis demonstrated that Arg(1) and His(11) but not Asp(2) were the functional residues. To the best of our knowledge, ViIA is the first 4/6 α-CTx that selectively inhibits nAChR α3ß2 subtype. This finding expands the knowledge of targets of α4/6-family CTxs.


Asunto(s)
Conotoxinas/química , Regulación de la Expresión Génica , Antagonistas Nicotínicos/química , Receptores Nicotínicos/metabolismo , Acetilcolina/química , Secuencias de Aminoácidos , Animales , Caracol Conus , Disulfuros , Biblioteca de Genes , Concentración 50 Inhibidora , Oocitos/citología , Técnicas de Placa-Clamp , Ratas , Relación Estructura-Actividad , Xenopus
12.
Sci Rep ; 5: 14349, 2015 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-26400022

RESUMEN

A novel hirudin isoform 3 mimetic peptide, named peptide S2, has been prepared by introduction of a stearic acid modification. Peptide S2 exhibited superior inhibitory activity to hirulog-1 (Bivariludin) and showed significantly higher anticoagulant potency in vivo. Peptide S2 elevated the thrombin time, prothrombin time and activated partial thromboplastin time of rat and human plasma more efficiently than hirulog-1 and the unmodified form of peptide S2 (peptide 1). Furthermore, peptide S2 inhibited arterial thrombosis and inferior vena cava in rat model 8 h after administration, and was 10-fold more potent than hirulog-1 300 min after administration of 0.1 µmol/kg peptide. The enhanced antithrombotic activity could be attributed to its long half-life (T1/2 = 212.2 ± 58.4 min), which was 13.1 and 14.7-fold longer than those of hirulog-1 (T1/2 = 15.1 ± 1.3 min) and peptide 1 (T1/2 = 13.5 ± 2.6 min), respectively. Further enzymatic degradation and binding assay with human serum albumin (HSA) demonstrated that the longer duration time should be originated from the slowing of trypsin or thrombin-mediated degradation, as well as its binding to HSA. The improved pharmacokinetic properties observed for peptide S2 has made it a promising therapeutic agent for the treatment of thrombi-related diseases.


Asunto(s)
Anticoagulantes/química , Anticoagulantes/farmacología , Hirudinas/química , Fragmentos de Péptidos/química , Fragmentos de Péptidos/farmacología , Peptidomiméticos , Ácidos Esteáricos/química , Animales , Anticoagulantes/síntesis química , Coagulación Sanguínea/efectos de los fármacos , Pruebas de Coagulación Sanguínea , Arterias Carótidas/patología , Humanos , Masculino , Fragmentos de Péptidos/síntesis química , Unión Proteica , Ratas , Albúmina Sérica/metabolismo , Trombina/antagonistas & inhibidores , Trombosis/tratamiento farmacológico , Trombosis/etiología , Vena Cava Inferior/patología
13.
Mar Drugs ; 13(6): 3259-75, 2015 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-26023835

RESUMEN

In the present study, we synthesized and, structurally and functionally characterized a novel α4/7-conotoxin Mr1.7 (PECCTHPACHVSHPELC-NH2), which was previously identified by cDNA libraries from Conus marmoreus in our lab. The NMR solution structure showed that Mr1.7 contained a 310-helix from residues Pro7 to His10 and a type I ß-turn from residues Pro14 to Cys17. Electrophysiological results showed that Mr1.7 selectively inhibited the α3ß2, α9α10 and α6/α3ß2ß3 neuronal nicotinic acetylcholine receptors (nAChRs) with an IC50 of 53.1 nM, 185.7 nM and 284.2 nM, respectively, but showed no inhibitory activity on other nAChR subtypes. Further structure-activity studies of Mr1.7 demonstrated that the PE residues at the N-terminal sequence of Mr1.7 were important for modulating its selectivity, and the replacement of Glu2 by Ala resulted in a significant increase in potency and selectivity to the α3ß2 nAChR. Furthermore, the substitution of Ser12 with Asn in the loop2 significantly increased the binding of Mr1.7 to α3ß2, α3ß4, α2ß4 and α7 nAChR subtypes. Taken together, this work expanded our knowledge of selectivity and provided a new way to improve the potency and selectivity of inhibitors for nAChR subtypes.


Asunto(s)
Conotoxinas/farmacología , Antagonistas Nicotínicos/farmacología , Receptores Nicotínicos/efectos de los fármacos , Animales , Conotoxinas/administración & dosificación , Conotoxinas/química , Caracol Conus/química , Concentración 50 Inhibidora , Espectroscopía de Resonancia Magnética , Antagonistas Nicotínicos/administración & dosificación , Antagonistas Nicotínicos/química , Receptores Nicotínicos/metabolismo , Relación Estructura-Actividad
14.
Neurosci Lett ; 597: 38-42, 2015 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-25896730

RESUMEN

As a variant of peptide conantokin-T (con-T), con-T[M8Q] is derived from the venom of Conus tulipa. Our previous study has demonstrated that con-T[M8Q] selectively targets N-methyl-d-aspartate receptor (NMDAR) NR2B subunit. In the present study, we determined the effects of con-T[M8Q] on the expression and development of morphine tolerance using hot plate test and acetic acid writhing test. Our results demonstrated that con-T[M8Q] could efficiently attenuate the expression and development of morphine analgesic tolerance in mice at low doses (5-20nmol/kg), and it exhibited more potent effects compared with ifenprodil, a typical small-molecule antagonist of NMDAR. In addition, low doses of con-T[M8Q] (5-20nmol/kg) did not cause drug resistance and apparent analgesic activity compared with morphine. Taken together, con-T[M8Q] could be a promising new candidate in attenuating morphine tolerance.


Asunto(s)
Analgésicos Opioides/efectos adversos , Venenos de Moluscos/farmacología , Morfina/efectos adversos , Péptidos/farmacología , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Animales , Conotoxinas , Tolerancia a Medicamentos , Femenino , Péptidos y Proteínas de Señalización Intercelular , Masculino , Ratones , Naloxona/farmacología , Antagonistas de Narcóticos/farmacología , Dimensión del Dolor , Umbral del Dolor/efectos de los fármacos , Síndrome de Abstinencia a Sustancias/psicología
15.
Lab Chip ; 15(3): 680-9, 2015 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-25406521

RESUMEN

The reconstruction of neural activity across complete neural circuits, or brain activity mapping, has great potential in both fundamental and translational neuroscience research. Larval zebrafish, a vertebrate model, has recently been demonstrated to be amenable to whole brain activity mapping in behaving animals. Here we demonstrate a microfluidic array system ("Fish-Trap") that enables high-throughput mapping of brain-wide activity in awake larval zebrafish. Unlike the commonly practiced larva-processing methods using a rigid gel or a capillary tube, which are laborious and time-consuming, the hydrodynamic design of our microfluidic chip allows automatic, gel-free, and anesthetic-free processing of tens of larvae for microscopic imaging with single-cell resolution. Notably, this system provides the capability to directly couple pharmaceutical stimuli with real-time recording of neural activity in a large number of animals, and the local and global effects of pharmacoactive drugs on the nervous system can be directly visualized and evaluated by analyzing drug-induced functional perturbation within or across different brain regions. Using this technology, we tested a set of neurotoxin peptides and obtained new insights into how to exploit neurotoxin derivatives as therapeutic agents. The novel and versatile "Fish-Trap" technology can be readily unitized to study other stimulus (optical, acoustic, or physical) associated functional brain circuits using similar experimental strategies.


Asunto(s)
Conducta Animal/efectos de los fármacos , Mapeo Encefálico , Encéfalo/efectos de los fármacos , Encéfalo/fisiología , Ensayos Analíticos de Alto Rendimiento , Técnicas Analíticas Microfluídicas , Vigilia/fisiología , Animales , Conducta Animal/fisiología , Larva , Neurotoxinas/farmacología , Péptidos/farmacología , Pez Cebra
16.
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
17.
Virus Res ; 189: 254-61, 2014 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-24930448

RESUMEN

In order to produce polyvalent vaccines based on single rVSV vector, we investigated the immunogenicity, antibody neutralizing activity, and antigenic determinant domain of Zaire Ebola's fragment MFL (aa 393-556) that contains furin site and internal fusion loop. Both the recombinant protein and the recombinant plasmid of fragment MFL elicited high levels of antibody, similar to those of Zaire Ebola GP (ZGP). The MFL fragment of ZGP also elicited high levels of neutralizing antibody and induced moderate cellular immune response in mice, as revealed by the proliferation and cytokine secretion of splenocytes. Through the analysis of the induction of neutralizing antibody by pVAX1-based recombinant plasmids that expressed truncated fragments of MFL, we found that the domain containing the internal fusion loop and the furin site was the major contributor of fragment MFL's immunogenicity. Furthermore, the rVSV-based bivalent vaccine expressing Sudan Ebola GP (SGP) and MFL fragment elicited efficient cross-immunity against ZGP and SGP with high levels of neutralizing antibody. Our results indicate that fragment MFL is an effective and novel antigen for the production of neutralizing antibody and polyvalent vaccines of Ebola virus.


Asunto(s)
Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Antígenos Virales/inmunología , Vacunas contra el Virus del Ébola/inmunología , Ebolavirus/inmunología , Vacunas de ADN/inmunología , Proteínas del Envoltorio Viral/inmunología , Animales , Proliferación Celular , Citocinas/metabolismo , Vacunas contra el Virus del Ébola/administración & dosificación , Vacunas contra el Virus del Ébola/genética , Mapeo Epitopo , Femenino , Leucocitos Mononucleares/inmunología , Ratones Endogámicos BALB C , Estructura Terciaria de Proteína , Bazo/inmunología , Vacunas de ADN/administración & dosificación , Vacunas de ADN/genética
18.
Protein Pept Lett ; 21(1): 69-74, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24354772

RESUMEN

The RGD sequence was used to design potent hirudin isoform 3 mimetic peptides with both antithrombin activity and antiplatelet aggregation activity. The RGD and proline were inserted between the catalytic active binding domain (D-Phe-Pro-Arg-Pro) on the N-terminus and the anion-binding exosite binding domain (QGDFEPIPEDAYDE) on the Cterminus. Thrombin titration assay and ATP-induced platelet aggregation test revealed that the peptide with the linker RGDWP or RGDGP possessed potent antithrombin and antiplatelet activities, while other peptides without the Pro residue in the linker only showed antithrombin activity. Similar results were obtained in the RGD-containing hirulog-1 variants. Our study indicates that the inserted Pro residue facilitates the exposure of RGD and the binding of the peptide to glycoprotein IIb/IIIa (GPIIb/IIIa). The strategy of combining the RGD sequence and the Pro residue may be used for future designs of bifunctional antithrombotic agents.


Asunto(s)
Proteínas Antitrombina/síntesis química , Antitrombinas/síntesis química , Hirudinas/química , Fragmentos de Péptidos/química , Inhibidores de Agregación Plaquetaria/síntesis química , Animales , Antitrombinas/química , Dominio Catalítico , Hirudinas/síntesis química , Integrina beta3/metabolismo , Masculino , Oligopéptidos , Fragmentos de Péptidos/síntesis química , Glicoproteína IIb de Membrana Plaquetaria/metabolismo , Isoformas de Proteínas/síntesis química , Isoformas de Proteínas/química , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/síntesis química , Proteínas Recombinantes/química , Tiempo de Coagulación de la Sangre Total
19.
Toxicon ; 60(6): 982-9, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22781954

RESUMEN

The venom peptides of cone snails are encoded by a large gene family, and can selectively bind to voltage-gated ion channels (Na⁺, K⁺ and Ca²âº channels) and to membrane receptors (nAChR, 5-HT3R, NMDAR). To identify novel conotoxin genes and analyze the evolution of typical conotoxin superfamily genes from different Conus species, we have constructed cDNA libraries derived from the venom ducts of Conus virgo, Conus eburneus, Conus imperialis and Conus marmoreus, which were collected from the South China Sea. 1312 transcripts from four Conus venom duct cDNA libraries were analyzed and 38.7-49.6% of the transcripts encoded conotoxin sequences. In addition to known conotoxins, 34 novel conotoxins have been identified and can be classified into eleven superfamilies, some of which showed unique patterns of cysteines or different signal peptide sequences. The evolutionary trees of T- and A-superfamily conotoxins were analyzed. Likelihood approaches revealed that T-superfamily conotoxins from the four Conus species undergo positive selection, mostly located in the mature toxin region. These findings contribute to a better understanding of the diversity and evolution of conotoxins from the South China Sea, and some novel conotoxins are valuable for further functional investigations.


Asunto(s)
Conotoxinas/genética , Conotoxinas/aislamiento & purificación , Caracol Conus/química , Caracol Conus/clasificación , Evolución Molecular , Secuencia de Aminoácidos , Animales , China , Clonación Molecular , Conotoxinas/química , ADN Complementario/genética , Biblioteca de Genes , Funciones de Verosimilitud , Datos de Secuencia Molecular , Océanos y Mares , Péptidos/química , Filogenia , Señales de Clasificación de Proteína
20.
J Inorg Biochem ; 105(1): 52-7, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21134602

RESUMEN

We have previously elucidated a new paradigm for the metal ion-induced helix-helix assembly in the natural γ-carboxyglutamic acid (Gla)-containing class of conantokin (con) peptides, typified by con-G and a variant of con-T, con-T[K7Gla], independent of the hydrophobic effect. In these "metallo-zipper" structures, Gla residues spaced at i, i+4, i+7, i+11 intervals, which is similar to the arrangement of a and d residues in typical heptads of coiled-coils, coordinate with Ca(2+) and form specific antiparallel helical dimers. In order to evaluate the common role of Gla residues in peptide self-assembly, we extend herein the same Gla arrangement to designed peptides: NH(2)-(γLSγEAK)(3)-CONH(2) (peptide 1) and NH(2)-γLSγEAKγLSγQANγLSγKAE-CONH(2) (peptide 2). Peptide 1 and peptide 2 exhibit no helicity alone, but undergo structural transitions to helical conformations in the presence of a variety of divalent cations. Sedimentation equilibrium ultracentrifugation analyses showed that peptide 1 and peptide 2 form helical dimers in the presence of Ca(2+), but not Mg(2+). Folding and thiol-disulfide rearrangement assays with Cys-containing peptide variants indicated that the helical dimers are mixtures of antiparallel and parallel dimers, which is different from the strict antiparallel strand orientations of con-G and con-T[K7γGla] dimers. These findings suggest that the Gla arrangement, i, i+4, i+7, i+11, i+14, plays a key role in helix formation, without a strict adherence to strand orientation of the helical dimer.


Asunto(s)
Ácido 1-Carboxiglutámico/química , Calcio/química , Péptidos/química , Péptidos/síntesis química , Calorimetría , Dicroismo Circular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA