The purpose of this proposal is to continue our ongoing investigation on the regulatory mechanisms of the mouse kappa opioid receptor (KOR) gene expression which is currently funded by an R01. Following the same direction as proposed in the original project, this renewal application further serves to understand the fundamental problems in the control of opioid receptor expression, by using both tissue culture and transgenic mouse models. It is known that opiates exert extensive pharmacological and physiological effects in animals. Like many other drugs, opioids act through specific receptors on target cells. Their very restricted expression in the adult nervous system and during developmental stages suggests that the specificity and the level of opioid receptor expression must be tightly regulated, and raises an important question of the control for their expression in a homeostatic state. In the previous award period, we have selected the KOR gene as a model to address this question, and our data indicate that KOR gene is under a multi-level regulatory circuit that involves positive and negative transcriptional regulatory mechanisms and several post-transcriptional events such as alternative splicing and translation. The negative transcriptional control is mediated through the action of vitamin A hormone (the retinoic acid), whereas alternative splicing generates KOR mRNA isoforms that have distinct RNA stability, and translation efficiency, and can be differentially transported to neuron cell bodies and fibers. In the same direction as the original project, we will continue to address our principle hypothesis that KOR gene is maintained at a homeostatic state by integrating different levels of regulatory events. At the transcriptional level, KOR is regulated in two phases during stem cell differentiation, with an initial negative effect exerted by an endocrine factor, the retinoic acid, followed by unknown positive mechanisms to reactivate KOR gene in neurons. Ultimately, we will learn: a) the molecular/genetic basis of KOR gene transcriptional regulation, particularly in the context of chromatin, b) KOR gene post-transcriptional control with regard to differential mRNA stability and transport, and c) KOR gene regulatory mechanisms extended from cell cultures to animals (transgenic mice). One important feature of this renewal proposal is that we will test our hypotheses in a more physiologically relevant condition by also studying the regulatory events at the chromatin level. Additionally, the application of transgenic mouse models is essential for understanding the genetic basis of pharmacological problems that are related to the opioid receptor system.

Agency
National Institute of Health (NIH)
Institute
National Institute on Drug Abuse (NIDA)
Type
Research Project (R01)
Project #
2R01DA011190-06
Application #
6469450
Study Section
Special Emphasis Panel (ZRG1-MDCN-5 (01))
Program Officer
Satterlee, John S
Project Start
1997-09-30
Project End
2007-06-30
Budget Start
2002-09-01
Budget End
2003-06-30
Support Year
6
Fiscal Year
2002
Total Cost
$256,786
Indirect Cost
Name
University of Minnesota Twin Cities
Department
Pharmacology
Type
Schools of Medicine
DUNS #
168559177
City
Minneapolis
State
MN
Country
United States
Zip Code
55455
Hwang, Cheol Kyu; Wagley, Yadav; Law, Ping-Yee et al. (2017) Phosphorylation of poly(rC) binding protein 1 (PCBP1) contributes to stabilization of mu opioid receptor (MOR) mRNA via interaction with AU-rich element RNA-binding protein 1 (AUF1) and poly A binding protein (PABP). Gene 598:113-130
Hwang, Cheol Kyu; Wagley, Yadav; Law, Ping-Yee et al. (2015) Analysis of epigenetic mechanisms regulating opioid receptor gene transcription. Methods Mol Biol 1230:39-51
Feng, X; Wu, C-Y; Burton, F H et al. (2014) ?-arrestin protects neurons by mediating endogenous opioid arrest of inflammatory microglia. Cell Death Differ 21:397-406
Wagley, Yadav; Hwang, Cheol Kyu; Lin, Hong-Yiou et al. (2013) Inhibition of c-Jun NH2-terminal kinase stimulates mu opioid receptor expression via p38 MAPK-mediated nuclear NF-?B activation in neuronal and non-neuronal cells. Biochim Biophys Acta 1833:1476-88
Wu, Qifang; Hwang, Cheol Kyu; Zheng, Hui et al. (2013) MicroRNA 339 down-regulates ?-opioid receptor at the post-transcriptional level in response to opioid treatment. FASEB J 27:522-35
Song, Kyu Young; Choi, Hack Sun; Law, Ping-Yee et al. (2013) Vimentin interacts with the 5'-untranslated region of mouse mu opioid receptor (MOR) and is required for post-transcriptional regulation. RNA Biol 10:256-66
Ho, Ping-Chih; Tsui, Yao-Chen; Feng, Xudong et al. (2012) NF-?B-mediated degradation of the coactivator RIP140 regulates inflammatory responses and contributes to endotoxin tolerance. Nat Immunol 13:379-86
Kang, Duk-Hee; Song, Kyu Young; Choi, Hack Sun et al. (2012) Novel dual-binding function of a poly (C)-binding protein 3, transcriptional factor which binds the double-strand and single-stranded DNA sequence. Gene 501:33-8
Ho, Ping-Chih; Tsui, Yao-Chen; Lin, Yi-Wei et al. (2012) Endothelin-1 promotes cytoplasmic accumulation of RIP140 through a ET(A)-PLCýý-PKCýý pathway. Mol Cell Endocrinol 351:176-83
Song, Kyu Young; Choi, Hack Sun; Law, Ping-Yee et al. (2012) Post-transcriptional regulation of mu-opioid receptor: role of the RNA-binding proteins heterogeneous nuclear ribonucleoprotein H1 and F. Cell Mol Life Sci 69:599-610

Showing the most recent 10 out of 104 publications