Your browser doesn't support javascript.
loading
Bradykinin-induced chemotaxis of human gliomas requires the activation of KCa3.1 and ClC-3.
Cuddapah, Vishnu Anand; Turner, Kathryn L; Seifert, Stefanie; Sontheimer, Harald.
Afiliación
  • Cuddapah VA; Department of Neurobiology and Center for Glial Biology in Medicine, University of Alabama at Birmingham, Birmingham, Alabama 35294, USA.
J Neurosci ; 33(4): 1427-40, 2013 Jan 23.
Article en En | MEDLINE | ID: mdl-23345219
ABSTRACT
Previous reports demonstrate that cell migration in the nervous system is associated with stereotypic changes in intracellular calcium concentration ([Ca(2+)](i)), yet the target of these changes are essentially unknown. We examined chemotactic migration/invasion of human gliomas to study how [Ca(2+)](i) regulates cellular movement and to identify downstream targets. Gliomas are primary brain cancers that spread exclusively within the brain, frequently migrating along blood vessels to which they are chemotactically attracted by bradykinin. Using simultaneous fura-2 Ca(2+) imaging and amphotericin B perforated patch-clamp electrophysiology, we find that bradykinin raises [Ca(2+)](i) and induces a biphasic voltage response. This voltage response is mediated by the coordinated activation of Ca(2+)-dependent, TRAM-34-sensitive K(Ca)3.1 channels, and Ca(2+)-dependent, 4,4'-diisothiocyanato-stilbene-2,2'-disulfonic acid (DIDS)-sensitive and gluconate-sensitive Cl(-) channels. A significant portion of these Cl(-) currents can be attributed to Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) activation of ClC-3, a voltage-gated Cl(-) channel/transporter, because pharmacological inhibition of CaMKII or shRNA-mediated knockdown of ClC-3 inhibited Ca(2+)-activated Cl(-) currents. Western blots show that K(Ca)3.1 and ClC-3 are expressed in tissue samples obtained from patients diagnosed with grade IV gliomas. Both K(Ca)3.1 and ClC-3 colocalize to the invading processes of glioma cells. Importantly, inhibition of either channel abrogates bradykinin-induced chemotaxis and reduces tumor expansion in mouse brain slices in situ. These channels should be further explored as future targets for anti-invasive drugs. Furthermore, these data elucidate a novel mechanism placing cation and anion channels downstream of ligand-mediated [Ca(2+)](i) increases, which likely play similar roles in other migratory cells in the nervous system.
Asunto(s)

Texto completo: 1 Colección: 01-internacional Base de datos: MEDLINE Asunto principal: Bradiquinina / Quimiotaxis / Canales de Cloruro / Canales de Potasio de Conductancia Intermedia Activados por el Calcio / Glioma / Invasividad Neoplásica Límite: Humans Idioma: En Revista: J Neurosci Año: 2013 Tipo del documento: Article País de afiliación: Estados Unidos

Texto completo: 1 Colección: 01-internacional Base de datos: MEDLINE Asunto principal: Bradiquinina / Quimiotaxis / Canales de Cloruro / Canales de Potasio de Conductancia Intermedia Activados por el Calcio / Glioma / Invasividad Neoplásica Límite: Humans Idioma: En Revista: J Neurosci Año: 2013 Tipo del documento: Article País de afiliación: Estados Unidos