Peripheral KV7 channels regulate visceral sensory function in mouse and human colon

Abstract

Background\textbf{Background} Chronic visceral pain is a defining symptom of many gastrointestinal disorders. The KV_V7 family (KV_V7.1-KV_V7.5) of voltage-gated potassium channels mediates the M current that regulates excitability in peripheral sensory nociceptors and central pain pathways. Here, we use a combination of immunohistochemistry, gut-nerve electrophysiological recordings in both mouse and human tissues, and single-cell qualitative real-time polymerase chain reaction of gut-projecting sensory neurons, to investigate the contribution of peripheral KV_V7 channels to visceral nociception. Results\textbf{Results} Immunohistochemical staining of mouse colon revealed labelling of KV_V7 subtypes (KV_V7.3 and KV_V7.5) with CGRP around intrinsic enteric neurons of the myenteric plexuses and within extrinsic sensory fibres along mesenteric blood vessels. Treatment with the KV_V7 opener retigabine almost completely abolished visceral afferent firing evoked by the algogen bradykinin, in agreement with significant co-expression of mRNA transcripts by single-cell qualitative real-time polymerase chain reaction for KCNQ subtypes and the B2_2 bradykinin receptor in retrogradely labelled extrinsic sensory neurons from the colon. Retigabine also attenuated responses to mechanical stimulation of the bowel following noxious distension (0-80 mmHg) in a concentration-dependent manner, whereas the KV_V7 blocker XE991 potentiated such responses. In human bowel tissues, KV_V7.3 and KV_V7.5 were expressed in neuronal varicosities co-labelled with synaptophysin and CGRP, and retigabine inhibited bradykinin-induced afferent activation in afferent recordings from human colon. Conclusions\textbf{Conclusions} We show that KV_V7 channels contribute to the sensitivity of visceral sensory neurons to noxious chemical and mechanical stimuli in both mouse and human gut tissues. As such, peripherally restricted KV_V7 openers may represent a viable therapeutic modality for the treatment of gastrointestinal pathologies.This work was supported by an EFIC-Grunenthal grant awarded to Madusha Peiris, CAG/CIHR/CCFR Fellowship to David E Reed, Rosetrees Postdoctoral Grant (A1296) to James RF Hockley/Ewan St. John Smith, Medical Research Council Grant (G0900907) to David C Bulmer and a Wellcome Trust University Award to L Ashley Blackshaw

    Similar works