Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 3 de 3
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Mar Drugs ; 15(9)2017 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-28867800

RESUMEN

Ciguatoxins (CTXs) are marine toxins that cause ciguatera fish poisoning, a debilitating disease dominated by sensory and neurological disturbances that include cold allodynia and various painful symptoms as well as long-lasting pruritus. Although CTXs are known as the most potent mammalian sodium channel activator toxins, the etiology of many of its neurosensory symptoms remains unresolved. We recently described that local application of 1 nM Pacific Ciguatoxin-1 (P-CTX-1) into the skin of human subjects induces a long-lasting, painful axon reflex flare and that CTXs are particularly effective in releasing calcitonin-gene related peptide (CGRP) from nerve terminals. In this study, we used mouse and rat skin preparations and enzyme-linked immunosorbent assays (ELISA) to study the molecular mechanism by which P-CTX-1 induces CGRP release. We show that P-CTX-1 induces CGRP release more effectively in mouse as compared to rat skin, exhibiting EC50 concentrations in the low nanomolar range. P-CTX-1-induced CGRP release from skin is dependent on extracellular calcium and sodium, but independent from the activation of various thermosensory transient receptor potential (TRP) ion channels. In contrast, lidocaine and tetrodotoxin (TTX) reduce CGRP release by 53-75%, with the remaining fraction involving L-type and T-type voltage-gated calcium channels (VGCC). Using transgenic mice, we revealed that the TTX-resistant voltage-gated sodium channel (VGSC) NaV1.9, but not NaV1.8 or NaV1.7 alone and the combined activation of the TTX-sensitive VGSC subtypes NaV1.7 and NaV1.1 carry the largest part of the P-CTX-1-caused CGRP release of 42% and 34%, respectively. Given the contribution of CGRP to nociceptive and itch sensing pathways, our findings contribute to a better understanding of sensory symptoms of acute and chronic ciguatera that may help in the identification of potential therapeutics.


Asunto(s)
Ciguatoxinas/farmacología , Canal de Sodio Activado por Voltaje NAV1.1/efectos de los fármacos , Canal de Sodio Activado por Voltaje NAV1.7/efectos de los fármacos , Canal de Sodio Activado por Voltaje NAV1.9/efectos de los fármacos , Receptores de Péptido Relacionado con el Gen de Calcitonina/efectos de los fármacos , Animales , Péptido Relacionado con Gen de Calcitonina/efectos de los fármacos , Calcio/metabolismo , Intoxicación por Ciguatera/metabolismo , Ciguatoxinas/química , Ensayo de Inmunoadsorción Enzimática , Humanos , Hiperalgesia/inducido químicamente , Lidocaína/farmacología , Masculino , Toxinas Marinas/farmacología , Potenciales de la Membrana/efectos de los fármacos , Ratones , Ratones Transgénicos , Ratas , Tetrodotoxina/farmacología
2.
J Neurosci ; 33(42): 16627-41, 2013 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-24133266

RESUMEN

Topically applied camphor elicits a sensation of cool, but nothing is known about how it affects cold temperature sensing. We found that camphor sensitizes a subpopulation of menthol-sensitive native cutaneous nociceptors in the mouse to cold, but desensitizes and partially blocks heterologously expressed TRPM8 (transient receptor potential cation channel subfamily M member 8). In contrast, camphor reduces potassium outward currents in cultured sensory neurons and, in cold nociceptors, the cold-sensitizing effects of camphor and menthol are additive. Using a membrane potential dye-based screening assay and heterologously expressed potassium channels, we found that the effects of camphor are mediated by inhibition of Kv7.2/3 channels subtypes that generate the M-current in neurons. In line with this finding, the specific M-current blocker XE991 reproduced the cold-sensitizing effect of camphor in nociceptors. However, the M-channel blocking effects of XE991 and camphor are not sufficient to initiate cold transduction but require a cold-activated inward current generated by TRPM8. The cold-sensitizing effects of XE991 and camphor are largest in high-threshold cold nociceptors. Low-threshold corneal cold thermoreceptors that express high levels of TRPM8 and lack potassium channels are not affected by camphor. We also found that menthol--like camphor--potently inhibits Kv7.2/3 channels. The apparent functional synergism arising from TRPM8 activation and M-current block can improve the effectiveness of topical coolants and cooling lotions, and may also enhance TRPM8-mediated analgesia.


Asunto(s)
Nociceptores/fisiología , Transducción de Señal/fisiología , Canales Catiónicos TRPM/metabolismo , Termorreceptores/fisiología , Sensación Térmica/fisiología , Animales , Alcanfor/farmacología , Frío , Femenino , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/metabolismo , Masculino , Mentol/farmacología , Ratones , Ratones Endogámicos C57BL , Fibras Nerviosas Amielínicas/efectos de los fármacos , Fibras Nerviosas Amielínicas/metabolismo , Nociceptores/metabolismo , Transducción de Señal/efectos de los fármacos , Canales Catiónicos TRPM/genética , Termorreceptores/metabolismo , Sensación Térmica/efectos de los fármacos
3.
EMBO J ; 31(19): 3795-808, 2012 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-22850668

RESUMEN

Ciguatoxins are sodium channel activator toxins that cause ciguatera, the most common form of ichthyosarcotoxism, which presents with peripheral sensory disturbances, including the pathognomonic symptom of cold allodynia which is characterized by intense stabbing and burning pain in response to mild cooling. We show that intraplantar injection of P-CTX-1 elicits cold allodynia in mice by targeting specific unmyelinated and myelinated primary sensory neurons. These include both tetrodotoxin-resistant, TRPA1-expressing peptidergic C-fibres and tetrodotoxin-sensitive A-fibres. P-CTX-1 does not directly open heterologously expressed TRPA1, but when co-expressed with Na(v) channels, sodium channel activation by P-CTX-1 is sufficient to drive TRPA1-dependent calcium influx that is responsible for the development of cold allodynia, as evidenced by a large reduction of excitatory effect of P-CTX-1 on TRPA1-deficient nociceptive C-fibres and of ciguatoxin-induced cold allodynia in TRPA1-null mutant mice. Functional MRI studies revealed that ciguatoxin-induced cold allodynia enhanced the BOLD (Blood Oxygenation Level Dependent) signal, an effect that was blunted in TRPA1-deficient mice, confirming an important role for TRPA1 in the pathogenesis of cold allodynia.


Asunto(s)
Ciguatoxinas/toxicidad , Dolor/inducido químicamente , Animales , Frío , Hiperalgesia/inducido químicamente , Imagen por Resonancia Magnética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratas , Ratas Wistar , Células Receptoras Sensoriales/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Canal Catiónico TRPA1 , Canales de Potencial de Receptor Transitorio/efectos de los fármacos , Canales de Potencial de Receptor Transitorio/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...