Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros

Bases de datos
Tipo de estudio
Tipo del documento
Asunto de la revista
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 119(5)2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-35074873

RESUMEN

The King Baboon spider, Pelinobius muticus, is a burrowing African tarantula. Its impressive size and appealing coloration are tempered by reports describing severe localized pain, swelling, itchiness, and muscle cramping after accidental envenomation. Hyperalgesia is the most prominent symptom after bites from P. muticus, but the molecular basis by which the venom induces pain is unknown. Proteotranscriptomic analysis of P. muticus venom uncovered a cysteine-rich peptide, δ/κ-theraphotoxin-Pm1a (δ/κ-TRTX-Pm1a), that elicited nocifensive behavior when injected into mice. In small dorsal root ganglion neurons, synthetic δ/κ-TRTX-Pm1a (sPm1a) induced hyperexcitability by enhancing tetrodotoxin-resistant sodium currents, impairing repolarization and lowering the threshold of action potential firing, consistent with the severe pain associated with envenomation. The molecular mechanism of nociceptor sensitization by sPm1a involves multimodal actions over several ion channel targets, including NaV1.8, KV2.1, and tetrodotoxin-sensitive NaV channels. The promiscuous targeting of peptides like δ/κ-TRTX-Pm1a may be an evolutionary adaptation in pain-inducing defensive venoms.


Asunto(s)
Nociceptores/efectos de los fármacos , Papio/metabolismo , Péptidos/farmacología , Venenos de Araña/farmacología , Arañas/metabolismo , Potenciales de Acción/efectos de los fármacos , Animales , Ganglios Espinales/efectos de los fármacos , Hiperalgesia/tratamiento farmacológico , Canales Iónicos/metabolismo , Ratones , Dolor/tratamiento farmacológico , Tetrodotoxina/farmacología
2.
J Nat Prod ; 71(6): 1099-103, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18444683

RESUMEN

Cancer cell toxicity-guided fractionation of extracts of the Papua New Guinea marine cyanobacteria Lyngbya majuscula and Lyngbya sordida led to the isolation of apratoxin D (1). Compound 1 contains the same macrocycle as apratoxins A and C but possesses the novel 3,7-dihydroxy-2,5,8,10,10-pentamethylundecanoic acid as the polyketide moiety. The planar structures and stereostructures of compound 1 were determined by extensive 1D and 2D NMR and MS data analyses and by comparison with the spectroscopic data of apratoxins A and C. Apratoxin D (1) showed potent in vitro cytotoxicity against H-460 human lung cancer cells with an IC 50 value of 2.6 nM.


Asunto(s)
Antineoplásicos/aislamiento & purificación , Antineoplásicos/farmacología , Cianobacterias/química , Depsipéptidos/aislamiento & purificación , Depsipéptidos/farmacología , Toxinas de Lyngbya/aislamiento & purificación , Toxinas de Lyngbya/farmacología , Toxinas Marinas/aislamiento & purificación , Toxinas Marinas/farmacología , Antineoplásicos/química , Depsipéptidos/química , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Concentración 50 Inhibidora , Toxinas de Lyngbya/química , Toxinas Marinas/química , Estructura Molecular , Resonancia Magnética Nuclear Biomolecular , Papúa Nueva Guinea
3.
PLoS One ; 12(9): e0182848, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28880874

RESUMEN

Spider venoms are rich sources of peptidic ion channel modulators with important therapeutical potential. We screened a panel of 60 spider venoms to find modulators of ion channels involved in pain transmission. We isolated, synthesized and pharmacologically characterized Cd1a, a novel peptide from the venom of the spider Ceratogyrus darlingi. Cd1a reversibly paralysed sheep blowflies (PD50 of 1318 pmol/g) and inhibited human Cav2.2 (IC50 2.6 µM) but not Cav1.3 or Cav3.1 (IC50 > 30 µM) in fluorimetric assays. In patch-clamp electrophysiological assays Cd1a inhibited rat Cav2.2 with similar potency (IC50 3 µM) without influencing the voltage dependence of Cav2.2 activation gating, suggesting that Cd1a doesn't act on Cav2.2 as a classical gating modifier toxin. The Cd1a binding site on Cav2.2 did not overlap with that of the pore blocker ω-conotoxin GVIA, but its activity at Cav2.2-mutant indicated that Cd1a shares some molecular determinants with GVIA and MVIIA, localized near the pore region. Cd1a also inhibited human Nav1.1-1.2 and Nav1.7-1.8 (IC50 0.1-6.9 µM) but not Nav1.3-1.6 (IC50 > 30 µM) in fluorimetric assays. In patch-clamp assays, Cd1a strongly inhibited human Nav1.7 (IC50 16 nM) and produced a 29 mV depolarising shift in Nav1.7 voltage dependence of activation. Cd1a (400 pmol) fully reversed Nav1.7-evoked pain behaviours in mice without producing side effects. In conclusion, Cd1a inhibited two anti-nociceptive targets, appearing to interfere with Cav2.2 inactivation gating, associated with the Cav2.2 α-subunit pore, while altering the activation gating of Nav1.7. Cd1a was inactive at some of the Nav and Cav channels expressed in skeletal and cardiac muscles and nodes of Ranvier, apparently contributing to the lack of side effects at efficacious doses, and suggesting potential as a lead for development of peripheral pain treatments.


Asunto(s)
Analgésicos/farmacología , Venenos de Araña/química , Arañas/química , Analgésicos/química , Animales , Sitios de Unión/efectos de los fármacos , Canales de Calcio Tipo N/metabolismo , Electrofisiología , Fluorometría , Humanos , Ratones , Canal de Sodio Activado por Voltaje NAV1.1/metabolismo , Canal de Sodio Activado por Voltaje NAV1.7/metabolismo , Péptidos/química , Péptidos/farmacología , Ratas
4.
Sci Rep ; 7(1): 974, 2017 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-28428547

RESUMEN

Voltage-gated sodium (NaV) channels are essential for the transmission of pain signals in humans making them prime targets for the development of new analgesics. Spider venoms are a rich source of peptide modulators useful to study ion channel structure and function. Here we describe ß/δ-TRTX-Pre1a, a 35-residue tarantula peptide that selectively interacts with neuronal NaV channels inhibiting peak current of hNaV1.1, rNaV1.2, hNaV1.6, and hNaV1.7 while concurrently inhibiting fast inactivation of hNaV1.1 and rNaV1.3. The DII and DIV S3-S4 loops of NaV channel voltage sensors are important for the interaction of Pre1a with NaV channels but cannot account for its unique subtype selectivity. Through analysis of the binding regions we ascertained that the variability of the S1-S2 loops between NaV channels contributes substantially to the selectivity profile observed for Pre1a, particularly with regards to fast inactivation. A serine residue on the DIV S2 helix was found to be sufficient to explain Pre1a's potent and selective inhibitory effect on the fast inactivation process of NaV1.1 and 1.3. This work highlights that interactions with both S1-S2 and S3-S4 of NaV channels may be necessary for functional modulation, and that targeting the diverse S1-S2 region within voltage-sensing domains provides an avenue to develop subtype selective tools.


Asunto(s)
Péptidos/farmacología , Venenos de Araña/química , Arañas/química , Canales de Sodio Activados por Voltaje/química , Canales de Sodio Activados por Voltaje/efectos de los fármacos , Animales , Sitios de Unión , Regulación de la Expresión Génica , Células HEK293 , Humanos , Modelos Moleculares , Péptidos/química , Unión Proteica , Estructura Secundaria de Proteína , Venenos de Araña/farmacología , Canales de Sodio Activados por Voltaje/metabolismo
5.
Sci Rep ; 7: 40883, 2017 01 20.
Artículo en Inglés | MEDLINE | ID: mdl-28106092

RESUMEN

Human genetic studies have implicated the voltage-gated sodium channel NaV1.7 as a therapeutic target for the treatment of pain. A novel peptide, µ-theraphotoxin-Pn3a, isolated from venom of the tarantula Pamphobeteus nigricolor, potently inhibits NaV1.7 (IC50 0.9 nM) with at least 40-1000-fold selectivity over all other NaV subtypes. Despite on-target activity in small-diameter dorsal root ganglia, spinal slices, and in a mouse model of pain induced by NaV1.7 activation, Pn3a alone displayed no analgesic activity in formalin-, carrageenan- or FCA-induced pain in rodents when administered systemically. A broad lack of analgesic activity was also found for the selective NaV1.7 inhibitors PF-04856264 and phlotoxin 1. However, when administered with subtherapeutic doses of opioids or the enkephalinase inhibitor thiorphan, these subtype-selective NaV1.7 inhibitors produced profound analgesia. Our results suggest that in these inflammatory models, acute administration of peripherally restricted NaV1.7 inhibitors can only produce analgesia when administered in combination with an opioid.

6.
Toxins (Basel) ; 8(3)2016 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-26999206

RESUMEN

Loss-of-function mutations of Na(V)1.7 lead to congenital insensitivity to pain, a rare condition resulting in individuals who are otherwise normal except for the inability to sense pain, making pharmacological inhibition of Na(V)1.7 a promising therapeutic strategy for the treatment of pain. We characterized a novel mouse model of Na(V)1.7-mediated pain based on intraplantar injection of the scorpion toxin OD1, which is suitable for rapid in vivo profiling of Na(V)1.7 inhibitors. Intraplantar injection of OD1 caused spontaneous pain behaviors, which were reversed by co-injection with Na(V)1.7 inhibitors and significantly reduced in Na(V)1.7(-/-) mice. To validate the use of the model for profiling Na(V)1.7 inhibitors, we determined the Na(V) selectivity and tested the efficacy of the reported Na(V)1.7 inhibitors GpTx-1, PF-04856264 and CNV1014802 (raxatrigine). GpTx-1 selectively inhibited Na(V)1.7 and was effective when co-administered with OD1, but lacked efficacy when delivered systemically. PF-04856264 state-dependently and selectively inhibited Na(V)1.7 and significantly reduced OD1-induced spontaneous pain when delivered locally and systemically. CNV1014802 state-dependently, but non-selectively, inhibited Na(V) channels and was only effective in the OD1 model when delivered systemically. Our novel model of Na(V)1.7-mediated pain based on intraplantar injection of OD1 is thus suitable for the rapid in vivo characterization of the analgesic efficacy of Na(V)1.7 inhibitors.


Asunto(s)
Canal de Sodio Activado por Voltaje NAV1.7/fisiología , Dolor/tratamiento farmacológico , Péptidos/uso terapéutico , Éteres Fenílicos/uso terapéutico , Prolina/análogos & derivados , Venenos de Escorpión/uso terapéutico , Bloqueadores de los Canales de Sodio/uso terapéutico , Venenos de Araña/uso terapéutico , Analgésicos , Animales , Conducta Animal/efectos de los fármacos , Células CHO , Cricetulus , Modelos Animales de Enfermedad , Células HEK293 , Humanos , Masculino , Ratones Endogámicos C57BL , Canal de Sodio Activado por Voltaje NAV1.7/genética , Fibras Nerviosas/efectos de los fármacos , Fibras Nerviosas/fisiología , Dolor/inducido químicamente , Prolina/uso terapéutico , Vena Safena/inervación , Sulfonamidas/uso terapéutico
7.
Biochem Pharmacol ; 83(11): 1562-71, 2012 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-22410003

RESUMEN

The human neuroblastoma cell line SH-SY5Y is a potentially useful model for the identification and characterisation of Na(v) modulators, but little is known about the pharmacology of their endogenously expressed Na(v)s. The aim of this study was to determine the expression of endogenous Na(v) α and ß subunits in SH-SY5Y cells using PCR and immunohistochemical approaches, and pharmacologically characterise the Na(v) isoforms endogenously expressed in this cell line using electrophysiological and fluorescence approaches. SH-SY5Y human neuroblastoma cells were found to endogenously express several Na(v) isoforms including Na(v)1.2 and Na(v)1.7. Activation of endogenously expressed Na(v)s with veratridine or the scorpion toxin OD1 caused membrane depolarisation and subsequent Ca(2+) influx through voltage-gated L- and N-type calcium channels, allowing Na(v) activation to be detected with membrane potential and fluorescent Ca(2) dyes. µ-Conotoxin TIIIA and ProTxII identified Na(v)1.2 and Na(v)1.7 as the major contributors of this response. The Na(v)1.7-selective scorpion toxin OD1 in combination with veratridine produced a Na(v)1.7-selective response, confirming that endogenously expressed human Na(v)1.7 in SH-SY5Y cells is functional and can be synergistically activated, providing a new assay format for ligand screening.


Asunto(s)
Regulación Enzimológica de la Expresión Génica/fisiología , Neuroblastoma/enzimología , Canales de Sodio/clasificación , Canales de Sodio/metabolismo , Línea Celular Tumoral , Fluorescencia , Humanos , Técnicas de Placa-Clamp , Isoformas de Proteínas , Bloqueadores de los Canales de Sodio/farmacología , Tetrodotoxina/farmacología , Veratridina/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA