Your browser doesn't support javascript.
loading
Alpha6-Containing Nicotinic Acetylcholine Receptors Mediate Nicotine-Induced Structural Plasticity in Mouse and Human iPSC-Derived Dopaminergic Neurons.
Collo, Ginetta; Cavalleri, Laura; Zoli, Michele; Maskos, Uwe; Ratti, Emiliangelo; Merlo Pich, Emilio.
  • Collo G; Section of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy.
  • Cavalleri L; Department of Biomedicine, University of Basel, Basel, Switzerland.
  • Zoli M; Section of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy.
  • Maskos U; Department of Biomedical, Metabolic and Neural Sciences, Center for Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, Modena, Italy.
  • Ratti E; Unité de Neurobiologie Intégrative des Systèmes Cholinergiques, CNRS UMR 3571, Institut Pasteur, Paris, France.
  • Merlo Pich E; Neuroscience Therapeutic Area Unit, Takeda Pharmaceuticals International Co., Cambridge, MA, United States.
Front Pharmacol ; 9: 572, 2018.
Article en En | MEDLINE | ID: mdl-29910731
ABSTRACT
Midbrain dopamine (DA) neurons are considered a critical substrate for the reinforcing and sensitizing effects of nicotine and tobacco dependence. While the role of the α4 and ß2 subunit containing nicotinic acetylcholine receptors (α4ß2∗nAChRs) in mediating nicotine effects on DA release and DA neuron activity has been widely explored, less information is available on their role in the morphological adaptation of the DA system to nicotine, eventually leading to dysfunctional behaviors observed in nicotine dependence. In particular, no information is available on the role of α6∗nAChRs in nicotine-induced structural plasticity in rodents and no direct evidence exists regarding the occurrence of structural plasticity in human DA neurons exposed to nicotine. To approach this problem, we used two parallel in vitro systems, mouse primary DA neuron cultures from E12.5 embryos and human DA neurons differentiated from induced pluripotent stem cells (iPSCs) of healthy donors, identified using TH+ immunoreactivity. In both systems, nicotine 1-10 µM produced a dose-dependent increase of maximal dendrite length, number of primary dendrites, and soma size when measured after 3 days in culture. These effects were blocked by pretreatments with the α6∗nAChR antagonists α-conotoxin MII and α-conotoxin PIA, as well as by the α4ß2nAChR antagonist dihydro-ß-erythroidine (DHßE) in both mouse and human DA neurons. Nicotine was also ineffective when the primary DA neurons were obtained from null mutant mice for either the α6 subunit or both the α4 and α6 subunits of nAChR. When pregnant mice were exposed to nicotine from gestational day 15, structural plasticity was also observed in the midbrain DA neurons of postnatal day 1 offspring only in wild-type mice and not in both null mutant mice. This study confirmed the critical role of α4α6∗nAChRs in mediating nicotine-induced structural plasticity in both mouse and human DA neurons, supporting the translational relevance of neurons differentiated from human iPSCs for pharmacological studies.
Palabras clave