Abstract

The noradrenergic and p38 mitogen-activated protein kinase (p38 MAPK) systems are implicated in cocaine-elicited behaviors. Previously, we demonstrated a role for p38 MAPK-mediated norepinephrine transporter (NET) Thr(30) phosphorylation in cocaine-induced NET up-regulation (Mannangatti, P., Arapulisamy, O., Shippenberg, T. S., Ramamoorthy, S., and Jayanthi, L. D. (2011) J. Biol. Chem. 286, 20239-20250). The present study explored the functional interaction between p38 MAPK-mediated NET regulation and cocaine-induced behaviors. In vitro cocaine treatment of mouse prefrontal cortex synaptosomes resulted in enhanced NET function, surface expression, and phosphorylation. Pretreatment with PD169316, a p38 MAPK inhibitor, completely blocked cocaine-mediated NET up-regulation and phosphorylation. In mice, in vivo administration of p38 MAPK inhibitor SB203580 completely blocked cocaine-induced NET up-regulation and p38 MAPK activation in the prefrontal cortex and nucleus accumbens. When tested for cocaine-induced locomotor sensitization and conditioned place preference (CPP), mice receiving SB203580 on cocaine challenge day or on postconditioning test day exhibited significantly reduced cocaine sensitization and CPP. A transactivator of transcription (TAT) peptide strategy was utilized to test the involvement of the NET-Thr(30) motif. In vitro treatment of synaptosomes with TAT-NET-Thr(30) (wild-type peptide) completely blocked cocaine-mediated NET up-regulation and phosphorylation. In vivo administration of TAT-NET-Thr(30) peptide but not TAT-NET-T30A (mutant peptide) completely blocked cocaine-mediated NET up-regulation and phosphorylation. In the cocaine CPP paradigm, mice receiving TAT-NET-Thr(30) but not TAT-NET-T30A on postconditioning test day exhibited significantly reduced cocaine CPP. Following extinction, TAT-NET-Thr(30) when given prior to cocaine challenge significantly reduced reinstatement of cocaine CPP. These results demonstrate that the direct inhibition of p38 MAPK or the manipulation of NET-Thr(30) motif/phosphorylation via a TAT peptide strategy prevents cocaine-induced NET up-regulation, locomotor sensitization, and CPP, suggesting a role for Thr(30)-linked NET regulation in cocaine-elicited behaviors.

Highlights

  • Cocaine-activated p38 MAPK-mediated norepinephrine transporter (NET) threonine 30 phosphorylation up-regulates NET

  • Similar to the observations made from our previous study [1], incubation of mouse prefrontal cortex (PFC) synaptosomes with 50 ␮M cocaine for 1 h produced a significant increase in NE transport capacity compared with vehicle controls (Fig. 1A)

  • Earlier studies indicated little evidence for NE playing a primary role in the reinforcing properties of psychomotor stimulants (44 – 46), more recent studies have documented that NET inhibition can play a significant role in cocaine-induced reinstatement [43]

Read more

Summary

Background

Cocaine-activated p38 MAPK-mediated norepinephrine transporter (NET) threonine 30 phosphorylation up-regulates NET. TATNET-Thr when given prior to cocaine challenge significantly reduced reinstatement of cocaine CPP These results demonstrate that the direct inhibition of p38 MAPK or the manipulation of NET-Thr motif/phosphorylation via a TAT peptide strategy prevents cocaine-induced NET up-regulation, locomotor sensitization, and CPP, suggesting a role for Thr30-linked NET regulation in cocaine-elicited behaviors. Initial in vitro experiments showed cocaine-mediated NET up-regulation and enhanced NET phosphorylation in mouse PFC synaptosomes These effects were completely blocked by prior treatment with either the p38 MAPK inhibitor PD169316 or TAT-NET-Thr. In vivo administration of the p38 MAPK inhibitor SB203580 or TAT-NET-Thr peptide significantly attenuated cocaine-induced locomotor sensitization and CPP These results suggested that p38 MAPK-mediated NET up-regulation is linked to cocaine-induced behaviors and that this regulation may have implications in the neuronal adaptations underlying cocaine addiction

EXPERIMENTAL PROCEDURES
RESULTS
DISCUSSION
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call