Although opioid drugs are effective for short-term pain relief, major obstacles remain with long-term use of these drugs. Specifically, the many side effects are associated with the drugs, such as increasing incidents of drug abuse leading to addiction, have hampered opioid drug usage. A probable mechanism for drug addiction involves the activation and alteration in the neural circuitry that normally is involved in pleasure, incentive motivation, and learning, during chronic drug exposure. In addition to dopaminergic inputs from the ventral tegmental area and substantia nigra to the nucleus accumbens and striatum, glutaminergic inputs from the prefrontal cortex, amygdala, and hippocampus also have important roles in chronic drug action. Since the hippocampus is the structure involved in the storage, consolidation, and retrieval of decorative, spatial, and long-term memory, understanding its roles in drug acquisition and relapse, as well as drug reward experiences, has gained importance. Both electrophysiological and morphological plasticity have been observed with the various hippocampal structures during the course of drug exposure. In addition, integration of newborn neurons to the existing circuit within the hippocampus may have pronounced effects on the drug experience. Considering that all addictive drugs have been shown to alter adult neurogenesis, elucidating the mechanism by which opioid drugs regulate adult neurogenesis, and identifying the specific aspect of the drug experience that adult neurogenesis participates in will have a significant impact in understanding the long-term use of opioid drugs. During the course of our studies on ?-opioid receptor (OPRM1) biased agonism, we observed that morphine and fentanyl, two highly prescribed opioids, regulate the microRNA-190 level differentially, leading to differences in NeuroD levels within primary hippocampal neuron cultures. Since NeuroD is the transcription factor involved in differentiation and maturation of neurons, we hypothesize that OPRM1, by controlling miR-190/NeuroD pathway activity, regulates adult neurogenesis in the hippocampus. We further hypothesize that, since morphine and fentanyl are both addictive, differential control of miR-190/NeuroD activity by these two agonists is not involved in the acquisition of addictive behavior, but rather in the consolidation and retrieval of the context memory associated with drug reward. Therefore, the proposed studies are designed: (A) to understand the molecular mechanism involved in morphine and fentanyl biased agonism so as to manipulate the outcomes of this biased agonism;(B) to establish that miR-190/NeuroD regulation is central to the agonists differential regulation of adult neurogenesis in the hippocampus;and (C) to link the regulation of NeuroD activities and neurogenesis with the extinction of conditioned place preference induced by opiate agonists. From these studies, we anticipate that, by manipulating the miR-190/NeuroD pathway activity, the extinction of the opioid drug reward experience and subsequent drug relapse, can be regulated, and a future treatment paradigm can be developed.

Public Health Relevance

With the recent increase in prescriptions written, leading to increased assess to opioids, elucidation of the mechanism in which long-term use of opioid medication leads to addiction and relapse, is critical. Upon completion, our studies will provide important information on whether, by regulating the various aspects of the differentiation and maturation of newborn neurons in the brain, the drug experience can be modulated. This information will be essential in the future development of eventual treatment paradigms for opioid addiction and relapse.

Agency
National Institute of Health (NIH)
Institute
National Institute on Drug Abuse (NIDA)
Type
Research Project (R01)
Project #
1R01DA031442-01A1
Application #
8250218
Study Section
Molecular Neuropharmacology and Signaling Study Section (MNPS)
Program Officer
Hillery, Paul
Project Start
2012-09-15
Project End
2017-08-31
Budget Start
2012-09-15
Budget End
2013-08-31
Support Year
1
Fiscal Year
2012
Total Cost
$424,517
Indirect Cost
$130,898
Name
University of Minnesota Twin Cities
Department
Pharmacology
Type
Schools of Medicine
DUNS #
555917996
City
Minneapolis
State
MN
Country
United States
Zip Code
55455
Fan, Wenxiang; Wang, Helei; Zhang, Yue et al. (2018) Morphine regulates adult neurogenesis and contextual memory extinction via the PKC?/Prox1 pathway. Neuropharmacology 141:126-138
Song, Kyu Young; Choi, Hack Sun; Law, Ping-Yee et al. (2017) Post-Transcriptional Regulation of the Human Mu-Opioid Receptor (MOR) by Morphine-Induced RNA Binding Proteins hnRNP K and PCBP1. J Cell Physiol 232:576-584
Kibaly, Cherkaouia; Lin, Hong-Yiou; Loh, Horace H et al. (2017) Spinal or supraspinal phosphorylation deficiency at the MOR C-terminus does not affect morphine tolerance in vivo. Pharmacol Res 119:153-168
Zhang, Lei; Kibaly, Cherkaouia; Wang, Yu-Jun et al. (2017) Src-dependent phosphorylation of ?-opioid receptor at Tyr336 modulates opiate withdrawal. EMBO Mol Med 9:1521-1536
Zhang, Yue; Xu, Chi; Zheng, Hui et al. (2016) Morphine Modulates Adult Neurogenesis and Contextual Memory by Impeding the Maturation of Neural Progenitors. PLoS One 11:e0153628
Kibaly, Cherkaouia; Kam, Angel Y F; Loh, Horace H et al. (2016) Naltrexone Facilitates Learning and Delays Extinction by Increasing AMPA Receptor Phosphorylation and Membrane Insertion. Biol Psychiatry 79:906-16
Xu, Chi; Loh, Horace H; Law, Ping-Yee (2016) Effects of addictive drugs on adult neural stem/progenitor cells. Cell Mol Life Sci 73:327-48
Xu, Chi; Zheng, Hui; Loh, Horace H et al. (2015) Morphine Promotes Astrocyte-Preferential Differentiation of Mouse Hippocampal Progenitor Cells via PKC?-Dependent ERK Activation and TRBP Phosphorylation. Stem Cells 33:2762-72
Li, Wen; He, Songwei; Zhou, Yuye et al. (2014) Neurod1 modulates opioid antinociceptive tolerance via two distinct mechanisms. Biol Psychiatry 76:775-84
Xu, Chi; Zhang, Yue; Zheng, Hui et al. (2014) Morphine modulates mouse hippocampal progenitor cell lineages by upregulating miR-181a level. Stem Cells 32:2961-72

Showing the most recent 10 out of 13 publications