Morphine acts on mu opioid receptors in the brain and spinal cord to produce dramatic pain relief, but mu-associated side effects (including tolerance and substance abuse) limit its use. Despite substantial information on mu signaling, the mechanisms by which morphine activates brain stem analgesic circuits remain unknown. The goal of this proposal is to validate a new mechanism for morphine analgesia and to investigate new analgesic agents which may mimic morphine actions but lack side effects. Cytochrome P450s are a family of drug-metabolizing enzymes that also perform endogenous metabolic oxidations in the brain. Many P450s have arachidonic acid (AA) epoxygenase activity, i.e. they convert AA to epoxyeicosatrienoic acids (EETs). Based on three new findings and a literature review, an epoxygenase hypothesis is proposed for opioid analgesic action in the brain stem: Morphine ->? receptor ->? AA ->(P450) ->? EET s ->? K+ ->? Analgesic Circuits Experiments to validate and exploit this hypothesis include: 1) Pilot studies show that newly-developed brain P450-deficient transgenic mice (BCPRN) have defective morphine antinociception. To validate this opioid analgesic phenotype, morphine experiments with BCPRN and control mice will investigate the importance of morphine dose, routes of administration, nociceptive tests and gender. 2) P450 epoxygenase inhibitors block morphine antinociception. Since morphine acts in the ventrolateral periaqueductal gray, the rostral ventromedial medulla, and the spinal dorsal horn, microinjections of P450 inhibitors and morphine into these regions of the rat CNS will identify the P450-relevant sites. Similar microinjections of morphine into BCPRN and control mice will confirm results from rats. 3) Morphine's mu-mediated side effects include modulation of respiration, locomotor activity, rotorod performance, and body temperature. Experiments with morphine and P450 inhibitors in rats and with morphine in BCPRN mice will discover which of these side effects utilize P450 mechanisms. 4) Additional experiments in rats with P450 inhibitors and with inhibitors of EET metabolism will further confirm and characterize the role of P450 expoxygenases in morphine analgesia. 5) Pilot results show that EETs, the products of AA epoxidation, have antinociceptive properties. Experiments to validate EET analgesia, to search for EET side effects, and to characterize EET mechanisms may reveal EETs to be a new class of experimental analgesic agents. The proposed studies will increase understanding of the mechanisms by which morphine relieves pain and produces unwanted side effects. Confirmation of the epoxygenase hypothesis may lead to new approaches for developing non-opioid, non-addicting pain relievers.

Public Health Relevance

There is an urgent need to discover new kinds of pain-relievers that lack addictive properties. This proposal will uncover new biochemical mechanisms for pain relief, and test a new group of chemicals for pain-relieving properties. This research could lead to the development of new, non-addicting pain relievers.

Agency
National Institute of Health (NIH)
Institute
National Institute on Drug Abuse (NIDA)
Type
Research Project (R01)
Project #
1R01DA027835-01
Application #
7773318
Study Section
Special Emphasis Panel (ZRG1-IFCN-E (02))
Program Officer
Purohit, Vishnudutt
Project Start
2010-03-01
Project End
2015-02-28
Budget Start
2010-03-01
Budget End
2011-02-28
Support Year
1
Fiscal Year
2010
Total Cost
$353,496
Indirect Cost
Name
Albany Medical College
Department
Pharmacology
Type
Schools of Medicine
DUNS #
190592162
City
Albany
State
NY
Country
United States
Zip Code
12208
Hough, Lindsay B; Nalwalk, Julia W; Yang, Weizhu et al. (2015) Neuronal cytochrome P450 activity and opioid analgesia: relevant sites and mechanisms. Brain Res 1616:10-8
Hough, Lindsay B; Nalwalk, Julia W; Cleary, Rachel A et al. (2014) Deficits in neuronal cytochrome P450 activity attenuate opioid analgesia but not opioid side effects. Eur J Pharmacol 740:255-62
Hough, Lindsay B; Nalwalk, Julia W; Yang, Weizhu et al. (2014) Significance of neuronal cytochrome P450 activity in opioid-mediated stress-induced analgesia. Brain Res 1578:30-7
Conroy, Jennie L; Nalwalk, Julia W; Phillips, James G et al. (2013) CC12, a P450/epoxygenase inhibitor, acts in the rat rostral, ventromedial medulla to attenuate morphine antinociception. Brain Res 1499:1-11
Agarwal, Jay J; Zhu, Yi; Zhang, Qing-Yu et al. (2013) TRAM-34, a putatively selective blocker of intermediate-conductance, calcium-activated potassium channels, inhibits cytochrome P450 activity. PLoS One 8:e63028
Hoerbelt, Paul; Nalwalk, Julia W; Phillips, James G et al. (2013) Antinociceptive activity of CC44, a biotinylated improgan congener. Eur J Pharmacol 714:464-71
Yang, Jun; VanAlstine, Melissa A; Phillips, James G et al. (2012) Cytochrome P450 2C24: Expression, Tissue Distribution, High-Throughput Assay, and Pharmacological Inhibition. Acta Pharm Sin B 2:137-145
Fleck, Mark W; Thomson, Jeffrey L; Hough, Lindsay B (2012) Histamine-gated ion channels in mammals? Biochem Pharmacol 83:1127-35
Phillips, Ryan S; Cleary, Daniel R; Nalwalk, Julia W et al. (2012) Pain-facilitating medullary neurons contribute to opioid-induced respiratory depression. J Neurophysiol 108:2393-404
VanAlstine, Melissa A; Hough, Lindsay B (2011) Effects of acetylenic epoxygenase inhibitors on recombinant cytochrome p450s. Drug Metab Dispos 39:1221-6

Showing the most recent 10 out of 15 publications