Tobacco use causes five million deaths worldwide annually and is the leading cause of preventable mortality in the world. Nicotine is the addictive component of tobacco that binds to and activates a family of ligand-gated ion channels, nicotinic acetylcholine receptors (nAChRs). Activation of the receptors in the dopaminergic mesocorticolimbic reward pathway is thought to underlie the initiation of addiction whereas signaling through nAChRs in the habenulo-interpeduncular pathway that feeds into the reward pathway is thought to play a key role in eliciting nicotine withdrawal symptoms. One of the challenges in understanding nicotine's effect on nAChRs arises from the existence of multiple nAChR subtypes, each exhibiting unique electrophysiological properties and varying affinities for nicotine. Eleven mammalian neuronal nAChR subunits have been identified (?2-??7, ?9, ?10 and ?2-??4). Five subunits co-assemble to form receptors with the subunit composition of each channel determining its pharmacological and biophysical properties. Chronic nicotine exposure alters the expression of nAChR subtypes, which likely contributes to nicotine dependence;however, the underlying mechanisms regulating these changes remain unclear, although transcriptional mechanisms are not thought to be involved. A growing body of evidence indicates that nicotine and cigarette smoke alters the expression of small 21-24 nucleotide long regulatory molecules, referred to as microRNAs (miRNAs). miRNAs are predicted to regulate the majority of all mammalian protein coding genes, typically by binding to complementary sites in the 3'- untranslated regions (3'-UTRs) of target mRNAs and guiding them to an RNA-induced silencing complex (RISC). To date, one miRNA, miR-1, has been reported to target nAChRs in C. elegans, leading to changes in nAChR function. Little is known regarding miRNA regulation in the context of mammalian nAChRs or nicotine addiction. We have preliminary data from a miRNA analysis using laser-captured material from mice chronically treated with nicotine that suggest: 1) a decrease in expression of two miRNAs, miR-7a and miR-9, in the ventral tegmental area (VTA) and 2) an increase in expression of the same two miRNAs in the medial habenula when compared to saline-treated animals. There was no change in their expression in the interpeduncular nucleus. Interestingly, potential targets of miR-7a and miR-9 include members of the nAChR family as well as other components important in the reward pathway. Our preliminary studies indicate that miR-7a regulates expression of the nAChR ?6 subunit via interactions with a single miR-7a recognition element in the 3'-UTR of the ?6 gene. These data form the basis of this application with the over-arching hypothesis that post-transcriptional regulation of nAChRs by miRNAs modulates nicotine-associated behaviors.
The first Aim will test the hypothesis that miR-7a and/or miR-9 modulates nicotine reward-associated behaviors as well as nAChR expression and function.
The second Aim will test the hypothesis that miR-7a and/or miR-9 modulates nicotine withdrawal-associated behaviors.
The final Aim will test the hypothesis that miR-7a and/or miR-9 interacts with the 3'-UTRs of nAChR subunit genes in a functionally relevant manner. The results from these experiments will provide unique insight to the molecular underpinnings of nicotine addiction.

Public Health Relevance

The goal of the proposed work is to understand the role of small regulatory RNA molecules in nicotine-mediated behaviors. Despite evidence indicating that expression of these molecules is altered by nicotine treatment, little is known regarding their function in this context. The proposed work will substantially close this gap in knowledge.

Agency
National Institute of Health (NIH)
Institute
National Institute on Drug Abuse (NIDA)
Type
Exploratory/Developmental Grants (R21)
Project #
5R21DA031952-02
Application #
8416976
Study Section
Molecular Neuropharmacology and Signaling Study Section (MNPS)
Program Officer
Satterlee, John S
Project Start
2012-02-01
Project End
2014-01-31
Budget Start
2013-02-01
Budget End
2014-01-31
Support Year
2
Fiscal Year
2013
Total Cost
$236,880
Indirect Cost
$92,880
Name
University of Massachusetts Medical School Worcester
Department
Psychiatry
Type
Schools of Medicine
DUNS #
603847393
City
Worcester
State
MA
Country
United States
Zip Code
01655
Rashnonejad, Afrooz; Chermahini, Gholamhossein Amini; Li, Shaoyong et al. (2016) Large-Scale Production of Adeno-Associated Viral Vector Serotype-9 Carrying the Human Survival Motor Neuron Gene. Mol Biotechnol 58:30-6
Wang, Dan; Gao, Guangping (2014) State-of-the-art human gene therapy: part I. Gene delivery technologies. Discov Med 18:67-77
Pang, Xueyan; Hogan, Eric M; Casserly, Alison et al. (2014) Dicer expression is essential for adult midbrain dopaminergic neuron maintenance and survival. Mol Cell Neurosci 58:22-8
Hogan, Eric M; Casserly, Alison P; Scofield, Michael D et al. (2014) miRNAome analysis of the mammalian neuronal nicotinic acetylcholine receptor gene family. RNA 20:1890-9
Wang, Dan; Gao, Guangping (2014) State-of-the-art human gene therapy: part II. Gene therapy strategies and clinical applications. Discov Med 18:151-61