cocaine reward
Recently Published Documents


TOTAL DOCUMENTS

131
(FIVE YEARS 26)

H-INDEX

36
(FIVE YEARS 3)

Neuron ◽  
2021 ◽  
Author(s):  
Song-Jun Xu ◽  
Sonia I. Lombroso ◽  
Delaney K. Fischer ◽  
Marco D. Carpenter ◽  
Dylan M. Marchione ◽  
...  

2021 ◽  
pp. 108753
Author(s):  
Pablo Giménez-Gómez ◽  
Raúl Ballestín ◽  
Leticia Gil de Biedma-Elduayen ◽  
Rebeca Vidal ◽  
Carmen Ferrer-Pérez ◽  
...  

2021 ◽  
Vol 7 (8) ◽  
pp. eabc8310
Author(s):  
Yoon-Jin Kim ◽  
Qingyao Kong ◽  
Soh Yamamoto ◽  
Kenta Kuramoto ◽  
Mei Huang ◽  
...  

Drug abuse is a foremost public health problem. Cocaine is a widely abused drug worldwide that produces various reward-related behaviors. The mechanisms that underlie cocaine-induced disorders are unresolved, and effective treatments are lacking. Here, we found that an autophagy-related protein Becn2 is a previously unidentified regulator of cocaine reward behaviors. Becn2 deletion protects mice from cocaine-stimulated locomotion and reward behaviors, as well as cocaine-induced dopamine accumulation and signaling, by increasing presynaptic dopamine receptor 2 (D2R) autoreceptors in dopamine neurons. Becn2 regulates D2R endolysosomal trafficking, degradation, and cocaine-induced behaviors via interacting with a D2R-bound adaptor GASP1. Inactivating Becn2 by upstream autophagy inhibitors stabilizes striatal presynaptic D2R, reduces dopamine release and signaling, and prevents cocaine reward in normal mice. Thus, the autophagy protein Becn2 is essential for cocaine psychomotor stimulation and reward through regulating dopamine neurotransmission, and targeting Becn2 by autophagy inhibitors is a potential strategy to prevent cocaine-induced behaviors.


Author(s):  
Yukie Kawahara ◽  
Yoshinori Ohnishi ◽  
Yoko Ohnishi ◽  
Hiroshi Kawahara ◽  
Akinori Nishi
Keyword(s):  

2020 ◽  
Author(s):  
Diptendu Mukherjee ◽  
Ben Jerry Gonzales ◽  
Reut Ashwal-Fluss ◽  
Hagit Turm ◽  
Maya Groysman ◽  
...  

AbstractDrug addiction develops due to brain-wide plasticity within neuronal ensembles, mediated by dynamic gene expression. Though the most common approach to identify such ensembles relies on immediate early gene expression, little is known of how the activity of these genes is linked to modified behavior observed following repeated drug exposure. To address this gap, we present a broad-to-specific approach, beginning with a comprehensive investigation of brain-wide cocaine-driven gene expression, through the description of dynamic spatial patterns of gene induction in subregions of the striatum, and finally address functionality of region-specific gene induction in the development of cocaine preference. Our findings reveal differential cell-type specific dynamic transcriptional recruitment patterns within two subdomains of the dorsal striatum following repeated cocaine exposure. Furthermore, we demonstrate that induction of the IEG Egr2 in the ventrolateral striatum, as well as the cells within which it is expressed, are required for the development of cocaine seeking.Impact statementVLS ensembles are dynamically recruited by cocaine experiences to mediate cocaine reward.


2020 ◽  
Vol 177 (20) ◽  
pp. 4796-4807 ◽  
Author(s):  
Chloe J. Jordan ◽  
Yi He ◽  
Guo‐Hua Bi ◽  
Zhi‐Bing You ◽  
Jianjing Cao ◽  
...  

Author(s):  
Morgan E. Zipperly ◽  
Faraz A. Sultan ◽  
Guan-En Graham ◽  
Andrew C. Brane ◽  
Natalie A. Simpkins ◽  
...  

Abstract Exposure to drugs of abuse produces robust transcriptional and epigenetic reorganization within brain reward circuits that outlives the direct effects of the drug and may contribute to addiction. DNA methylation is a covalent epigenetic modification that is altered following stimulant exposure and is critical for behavioral and physiological adaptations to drugs of abuse. Although activity-related loss of DNA methylation requires the Gadd45 (Growth arrest and DNA-damage-inducible) gene family, very little is known about how this family regulates activity within the nucleus accumbens or behavioral responses to drugs of abuse. Here, we combined genome-wide transcriptional profiling, pharmacological manipulations, electrophysiological measurements, and CRISPR tools with traditional knockout and behavioral approaches in rodent model systems to dissect the role of Gadd45b in dopamine-dependent epigenetic regulation and cocaine reward. We show that acute cocaine administration induces rapid upregulation of Gadd45b mRNA in the rat nucleus accumbens, and that knockout or site-specific CRISPR/Cas9 gene knockdown of Gadd45b blocks cocaine conditioned place preference. In vitro, dopamine treatment in primary striatal neurons increases Gadd45b mRNA expression through a dopamine receptor type 1 (DRD1)-dependent mechanism. Moreover, shRNA-induced Gadd45b knockdown decreases expression of genes involved in psychostimulant addiction, blocks induction of immediate early genes by DRD1 stimulation, and prevents DRD1-mediated changes in DNA methylation. Finally, we demonstrate that Gadd45b knockdown decreases striatal neuron action potential burst duration in vitro, without altering other electrophysiological characteristics. These results suggest that striatal Gadd45b functions as a dopamine-induced gene that is necessary for cocaine reward memory and DRD1-mediated transcriptional activity.


2020 ◽  
Vol 221 ◽  
pp. 112913
Author(s):  
Carmen Ferrer-Pérez ◽  
Marina D. Reguilón ◽  
José Miñarro ◽  
Marta Rodríguez-Arias

2020 ◽  
Vol 237 (9) ◽  
pp. 2633-2648
Author(s):  
Britahny M. Baskin ◽  
Jia Jia Mai ◽  
Susan M. Dymecki ◽  
Kathleen M. Kantak
Keyword(s):  

Sign in / Sign up

Export Citation Format

Share Document