Skip Navigation

Publication Detail

Title: Dopaminergic neurotoxicants cause biphasic inhibition of purinergic calcium signaling in astrocytes.

Authors: Streifel, Karin M; Gonzales, Albert L; De Miranda, Briana; Mouneimne, Rola; Earley, Scott; Tjalkens, Ronald

Published In PLoS One, (2014)

Abstract: Dopaminergic nuclei in the basal ganglia are highly sensitive to damage from oxidative stress, inflammation, and environmental neurotoxins. Disruption of adenosine triphosphate (ATP)-dependent calcium (Ca2+) transients in astrocytes may represent an important target of such stressors that contributes to neuronal injury by disrupting critical Ca2+-dependent trophic functions. We therefore postulated that plasma membrane cation channels might be a common site of inhibition by structurally distinct cationic neurotoxicants that could modulate ATP-induced Ca2+ signals in astrocytes. To test this, we examined the capacity of two dopaminergic neurotoxicants to alter ATP-dependent Ca2+ waves and transients in primary murine striatal astrocytes: MPP+, the active metabolite of 1-methyl 4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), and 6-hydroxydopamine (6-OHDA). Both compounds acutely decreased ATP-induced Ca2+ transients and waves in astrocytes and blocked OAG-induced Ca2+ influx at micromolar concentrations, suggesting the transient receptor potential channel, TRPC3, as an acute target. MPP+ inhibited 1-oleoyl-2-acetyl-sn-glycerol (OAG)-induced Ca2+ transients similarly to the TRPC3 antagonist, pyrazole-3, whereas 6-OHDA only partly suppressed OAG-induced transients. RNAi directed against TRPC3 inhibited the ATP-induced transient as well as entry of extracellular Ca2+, which was augmented by MPP+. Whole-cell patch clamp experiments in primary astrocytes and TRPC3-overexpressing cells demonstrated that acute application of MPP+ completely blocked OAG-induced TRPC3 currents, whereas 6-OHDA only partially inhibited OAG currents. These findings indicate that MPP+ and 6-OHDA inhibit ATP-induced Ca2+ signals in astrocytes in part by interfering with purinergic receptor mediated activation of TRPC3, suggesting a novel pathway in glia that could contribute to neurotoxic injury.

PubMed ID: 25365260 Exiting the NIEHS site

MeSH Terms: 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology; Adenosine Triphosphate/metabolism; Animals; Astrocytes/drug effects*; Astrocytes/metabolism*; Calcium Signaling/drug effects*; Calcium/metabolism; Cell Line; Corpus Striatum/cytology; Corpus Striatum/drug effects; Corpus Striatum/metabolism; Dopamine Agents/pharmacology*; Humans; Mice; Neurotoxins/pharmacology*; Oxidopamine/pharmacology; Purinergic Agents/pharmacology*; Receptors, G-Protein-Coupled/metabolism; TRPC Cation Channels/metabolism

Back
to Top