MetaTOC stay on top of your field, easily

Genetic alteration of the metal/redox modulation of Cav3.2 T‐type calcium channel reveals its role in neuronal excitability

, ,

The Journal of Physiology

Published online on

Abstract

Key points In this study, we describe a new knock‐in (KI) mouse model that allows the study of the H191‐dependent regulation of T‐type Cav3.2 channels. Sensitivity to zinc, nickel and ascorbate of native Cav3.2 channels is significantly impeded in the dorsal root ganglion (DRG) neurons of this KI mouse. Importantly, we describe that this H191‐dependent regulation has discrete but significant effects on the excitability properties of D‐hair (down‐hair) cells, a sub‐population of DRG neurons in which Cav3.2 currents prominently regulate excitability. Overall, this study reveals that the native H191‐dependent regulation of Cav3.2 channels plays a role in the excitability of Cav3.2‐expressing neurons. This animal model will be valuable in addressing the potential in vivo roles of the trace metal and redox modulation of Cav3.2 T‐type channels in a wide range of physiological and pathological conditions. Abstract Cav3.2 channels are T‐type voltage‐gated calcium channels that play important roles in controlling neuronal excitability, particularly in dorsal root ganglion (DRG) neurons where they are involved in touch and pain signalling. Cav3.2 channels are modulated by low concentrations of metal ions (nickel, zinc) and redox agents, which involves the histidine 191 (H191) in the channel's extracellular IS3–IS4 loop. It is hypothesized that this metal/redox modulation would contribute to the tuning of the excitability properties of DRG neurons. However, the precise role of this H191‐dependent modulation of Cav3.2 channel remains unresolved. Towards this goal, we have generated a knock‐in (KI) mouse carrying the mutation H191Q in the Cav3.2 protein. Electrophysiological studies were performed on a subpopulation of DRG neurons, the D‐hair cells, which express large Cav3.2 currents. We describe an impaired sensitivity to zinc, nickel and ascorbate of the T‐type current in D‐hair neurons from KI mice. Analysis of the action potential and low‐threshold calcium spike (LTCS) properties revealed that, contrary to that observed in WT D‐hair neurons, a low concentration of zinc and nickel is unable to modulate (1) the rheobase threshold current, (2) the afterdepolarization amplitude, (3) the threshold potential necessary to trigger an LTCS or (4) the LTCS amplitude in D‐hair neurons from KI mice. Together, our data demonstrate that this H191‐dependent metal/redox regulation of Cav3.2 channels can tune neuronal excitability. This study validates the use of this Cav3.2‐H191Q mouse model for further investigations of the physiological roles thought to rely on this Cav3.2 modulation.