Endomorphins fully activate a cloned human mu opioid receptor

Similar documents
D. Nishizawa 1, N. Gajya 2 and K. Ikeda 1, * Global Research & Development, Nagoya Laboratories, Pfizer Japan Inc, Nagoya, Japan

Supporting Information

nachr α 4 β 2 CHO Cell Line

Human TRPC6 Ion Channel Cell Line

Constitutive activity ofthe d-opioid receptor expressed in C6 glioma cells: identi cation of non-peptide d-inverse agonists

Drug addiction is a complex process that includes tolerance

CHO α 1 β 2 γ 2 GABAA Cell Line

Interaction of Co-Expressed - and -Opioid Receptors in Transfected Rat Pituitary GH 3 Cells

Agonist-Induced Desensitization and Down-regulation of the Human Kappa Opioid Receptor Expressed in Chinese Hamster Ovary Cells 1

Acta Physiologica Sinica

STEIN IN-TERM EXAM -- BIOLOGY FEBRUARY 16, PAGE

Enhancement of synaptic transmission by cyclic AMP modulation of presynaptic I h channels. Vahri Beaumont and Robert S. Zucker

Tsutomu Fujimura, Kimie Murayama, Shunsuke Kawamura, Takumi Sato, Chikai Sakurada,

Neurotransmitter Systems II Receptors. Reading: BCP Chapter 6

Physiology Unit 1 CELL SIGNALING: CHEMICAL MESSENGERS AND SIGNAL TRANSDUCTION PATHWAYS

Leon F. Tseng* Keywords: Endomorphin-1, Endomorphin-2, Antinociception, -Opioid receptor, Descending pain control system

VaTx1 VaTx2 VaTx3. VaTx min Retention Time (min) Retention Time (min)

Phys 173 / BGGN 266. LPA Induced Cl - Oscillations in Xenopus Oocytes. Nini Huynh David Marciano Chisa Suzuki

MOLECULAR BIOLOGY OF DRUG ADDICTION. Sylvane Desrivières, SGDP Centre

(Supplementary information) TRPV1 EXHIBITS DYNAMIC IONIC SELECTIVITY DURING AGONIST STIMULATION. Man-Kyo Chung, Ali D. Güler, and #Michael J.

Basics of Pharmacology

Neurons of the Bed Nucleus of the Stria Terminalis (BNST)

Supporting Online Material for

Comparison of opioid receptor binding in horse, guinea pig, and rat cerebral cortex and cerebellum

RESEARCH PAPER Neutral antagonist activity of naltrexone and 6b-naltrexol in naïve and opioid-dependent C6 cells expressing a m-opioid receptor

Na + /Cl - -dependent Neurotransmitter Transporters. Outline. Synaptic Transmission. How did the neurotransmitter cross the membrane?

Cellular Messengers. Intracellular Communication

Silencing neurotransmission with membrane-tethered toxins

Continued morphine modulation of calcium channel currents in acutely isolated locus coeruleus neurons from morphine-dependent rats

MEK1 Assay Kit 1 Catalog # Lot # 16875

DYNORPHIN-(1-13) SUPPRESSES HEROIN WITHDRAWAL SYMPTOMS IN 6 ADDICTS

Use of a camp BRET Sensor to Characterize a Novel Regulation of camp by the Sphingosine-1-phosphate/G 13 Pathway

2013 W. H. Freeman and Company. 12 Signal Transduction

8-Br-cAMP SQ/DDA NKH477 AC IBMX PDE AMP. camp IP 3 R. Control + ESI-09. Control + H89. peak [Ca 2+ ] c (nm) log [PTH(1-34)] (/M) log [PTH(1-34)] (/M)

Highly Expressed Subtypes With Relatively Low Affinity for [ 3 H]Epibatidine. Michael J. Marks, Paul Whiteaker and Allan C.

THE OPIUM POPPY OPIOID PHARMACOLOGY 2/18/16. PCTH 300/305 Andrew Horne, PhD MEDC 309. Papaver somniferum. Poppy Seeds Opiates

Nervous System Communication. Nervous System Communication. The First Nerve Cells 1/2/11

For Research Use Only

supplementary information

Drug Receptor Interactions and Pharmacodynamics

Communication within a Neuron

STEIN IN-TERM EXAM -- BIOLOGY FEBRUARY 15, PAGE

Myocardial Opiate Receptors

STEIN IN-TERM EXAM -- BIOLOGY FEBRUARY 18, PAGE

Western Immunoblotting Preparation of Samples:

A comparison of e ects measured with isotonic and isometric recording: II. Concentration-e ect curves for physiological antagonists

PAIN & ANALGESIA. often accompanied by clinical depression. fibromyalgia, chronic fatigue, etc. COX 1, COX 2, and COX 3 (a variant of COX 1)

Total Phosphatidic Acid Assay Kit

Overview of Pharmacodynamics. Psyc 472 Pharmacology of Psychoactive Drugs. Pharmacodynamics. Effects on Target Binding Site.

P/Q And N Channels Control Baseline and Spike-Triggered Calcium Levels in Neocortical Axons And Synaptic Boutons

INHIBITION OF THE ACETYLCHOLINE RECEPTOR BY HISTRIONICOTOXIN

Kit for assay of thioredoxin

Arylazido-L-alanine ADP-ribose, a novel irreversible competitive inhibitor of mitochondrial NADH-ubiquinone reductase

Fast Calcium Currents in Cut Skeletal Muscle Fibres of the Frogs Rana temporaria and Xenopus laevis

INTRODUCTION. The 5-HT 2C receptor is a seven transmembrane spanning protein coupled to G proteins.

From opium to analgesic tests: An introduction to the functioning and studying of the opioid system

From opium to analgesic tests: An introduction to the functioning and studying of the opioid system

CONTENTS. Disclosure of Conflict of Interest. None. Is opioid effective in long-term use? Can opioid exacerbate pain? Why are we still using opioids?

Glycine-gated ion channels Converging mechanism and therapeutic potentials

Choline as a tool to evaluate nicotinic receptor function in chromaffin cells.

Heart Matters: Physiology of the Body s Powerhouse 11/28/12. Igor Mitrovic, MD

Electronic Supplementary Information. Table of Contents

PhosFree TM Phosphate Assay Biochem Kit

PHRM20001: Pharmacology - How Drugs Work!

Differentiation of delta and mu opiate receptor localizations by light

Institute of Chemical Physics and *Institute of Biochemistry, University of Tartu, Jakobi 2, EE-2400 Tartu, Estonia

Blockage of 5HT 2C serotonin receptors by fluoxetine (Prozac)

EDWARD F. NEMETH*, MICHAEL E. STEFFEY, LANCE G. HAMMERLAND, BENJAMIN C. P. HUNG, BRADFORD C. VAN WAGENEN, ERIC G. DELMAR, AND MANUEL F.

A Homogeneous Phosphoinositide 3-Kinase Assay on Phospholipid FlashPlate Platforms. Busi Maswoswe, Hao Xie, Pat Kasila and Li-an Yeh

The role of M 2 -muscarinic receptors in mediating contraction of the pig urinary bladder in vitro

Optimization of the GeneBLAzer H2 CRE-bla HEK 293T Cell Line

Agonists at mu opioid receptors spin the wheels to keep the action going.

Cystic Fibrosis. Na+ 2Cl - K+ Na+ Na+

Chromatin IP (Isw2) Fix soln: 11% formaldehyde, 0.1 M NaCl, 1 mm EDTA, 50 mm Hepes-KOH ph 7.6. Freshly prepared. Do not store in glass bottles.

HEK293-GRIK2. Glutamate receptor GluR6. Application Report:

Manual. Precision Red Advanced Protein Assay Reagent. Cat. # ADV02. cytoskeleton.com. Cytoskeleton, Inc.

Calcium alters the sensitivity of intact horizontal cells to

Potentiation of the Inotropic Effect of Isoproterenol by Naloxone in vitro; Non-opiate Receptor Mechanism

BRIEF COMMUNICATION CALCIUM- AND VOLTAGE-ACTIVATED POTASSIUM CHANNELS IN HUMAN MACROPHAGES. frequency of channel opening increased with depolarization

A Membrane Receptor Binding Assay Using MesoScale Electrochemiluminescence Technology

Data Sheet TIGIT / NFAT Reporter - Jurkat Cell Line Catalog #60538

Effect of the endogenous analgesic dipeptide, kyotorphin, on transmitter release in sympathetic

Receptors and Drug Action. Dr. Subasini Pharmacology Department Ishik University, Erbil

Mu and Delta opioid receptors activate the same G proteins in human neuroblastoma SH-SY5Y cells

Supplementary data Inactivation of Human Angiotensin Converting Enzyme by Copper Peptide Complexes Containing ATCUN Motifs.

AMPK Assay. Require: Sigma (1L, $18.30) A4206 Aluminum foil

Utilizing AlphaLISA Technology to Screen for Inhibitors of the CTLA-4 Immune Checkpoint

Pharmacology of Pain Transmission and Modulation

Ghrelin Stimulates Porcine Somatotropes

The ORL 1 Receptor: Molecular Pharmacology and Signalling Mechanisms

Orexin 2 Receptor Reporter Assay Kit

Supplementary Information

TRANSPORT OF AMINO ACIDS IN INTACT 3T3 AND SV3T3 CELLS. Binding Activity for Leucine in Membrane Preparations of Ehrlich Ascites Tumor Cells

Modulation of TRP channels by resolvins in mouse and human

Acute and chronic activation of the µ-opioid receptor with the endogenous ligand endomorphin differentially regulates adenylyl cyclase isozymes

Clinical trial of brain-protective effect of nalmefene hydrochloride in patients with acute traumatic

Naltrexone for the treatment of Crohn s disease

Mária Szücs, Krisztina Boda, and Alan R. Gintzler

2-Deoxyglucose Assay Kit (Colorimetric)

Receptors and Neurotransmitters: It Sounds Greek to Me. Agenda. What We Know About Pain 9/7/2012

Transcription:

FEBS Letters 439 (1998) 152^156 FEBS 21116 Endomorphins fully activate a cloned human mu opioid receptor Jianhua Gong a, Judith A. Strong a, Shengwen Zhang a, Xia Yue a, Robert N. DeHaven c, Je rey D. Daubert c, Joel A. Cassel c, Guangling Yu c, Erik Mansson c, Lei Yu a;b; * a Department of Cell Biology, Neurobiology and Anatomy,University of Cincinnati College of Medicine, 231 Bethesda Avenue, Cincinnati, OH 45267-0521, USA b Department of Psychiatry, University of Cincinnati College of Medicine, Cincinnati, OH 45267-0521, USA c Adolor Corporation, Malvern, PA 19355, USA Received 17 August 1998; received in revised form 6 October 1998 Abstract Endomorphins were recently identified as endogenous ligands with high selectivity for mu opioid receptors. We have characterized the ability of endomorphins to bind to and functionally activate the cloned human mu opioid receptor. Both endomorphin-1 and endomorphin-2 exhibited binding selectivity for the mu opioid receptor over the delta and kappa opioid receptors. Both agonists inhibited forskolin-stimulated increase of camp in a dose-dependent fashion. When the mu opioid receptor was coexpressed in Xenopus oocytes with G proteinactivated K + channels, application of either endomorphin activated an inward K + current. This activation was dosedependent and blocked by naloxone. Both endomorphins acted as full agonists with efficacy similar to that of [D-Ala 2,N-Me- Phe 4,Gly-ol 5 ]enkephalin (DAMGO). These data indicate that endomorphins act as full agonists at the human mu opioid receptor, capable of stimulating the receptor to inhibit the camp/adenylyl cyclase pathway and activate G-protein-activated inwardly rectifying potassium (GIRK) channels. z 1998 Federation of European Biochemical Societies. Key words: Endomorphin; Human mu opioid receptor 1. Introduction Opioids, including both endogenous peptides and exogenous alkaloids, exert their physiological e ects by interacting with membrane-bound opioid receptors. These e ects include regulation of pain perception, reinforcement and reward, neurotransmitter release, hormone secretion, gastrointestinal motility, and respiratory activity [1]. Pharmacological studies have identi ed three major types of opioid receptors in the brain and spinal cord: mu, delta, and kappa, of which the mu opioid receptor plays an important role in opioid analgesia and development of opioid dependence [2,3]. Extensive studies have also led to the identi cation of three major classes of endogenous ligands in the mammalian brain: endorphins, enkephalins, and dynorphins [4]. Endorphins and enkephalins can activate both the mu and delta opioid receptors, while dynorphins are considered to be relatively selective for the kappa opioid receptor. Zadina et al. [5] isolated two endogenous opioid peptides *Corresponding author. Fax: (1) (513) 558-3367. E-mail: lei.yu@uc.edu Abbreviations: DAMGO, [D-Ala 2,N-Me-Phe 4,Gly-ol 5 ]enkephalin; GIRK, G-protein-activated inwardly rectifying potassium channel; hmor, human mu opioid receptor; hdor, human delta opioid receptor; hkor, human kappa opioid receptor; CHO cells, Chinese hamster ovary cells; HK, high potassium; EDM, endomorphin from the mammalian brain, named endomorphin-1 (Tyr-Pro- Trp-Phe-NH 2 ) and endomorphin-2 (Tyr-Pro-Phe-Phe-NH 2 ), that display high a nity and selectivity for the mu opioid receptor. Endomorphin-1 was also demonstrated to have higher binding a nity than the synthetic mu-selective analogue [D-Ala 2,N-Me-Phe 4,Gly-ol 5 ]enkephalin (DAMGO) in vitro, and to produce potent and prolonged analgesia in mice [5]. However, these studies were conducted in brain tissues and in vivo, in which multiple types of opioid receptors are present. The major cellular e ects of activation of all three types of opioid receptors by their agonists are inhibition of adenylyl cyclase and modulation of membrane Ca 2 and K conductances [6,7]. Opioid-mediated increases in K conductance and decrease in Ca 2 conductance both serve to reduce membrane excitability and contribute to the analgesic properties of the opioids. Because camp is an important second messenger in cell signalling, opioid inhibition of adenylyl cyclase implies a more complex pathway for opioid regulation of cellular mechanisms. These two major cellular e ects of mu opioid receptor activation have been studied by using cloned mu opioid receptors since expression of the cloned receptor provides a cellular model with a single de ned receptor type [8^ 10]. In this study, we evaluated the functional e ects, including both e cacy and potency, of endomorphins on the cloned human mu opioid receptor. 2. Materials and methods 2.1. Receptor expression and binding assay Chinese hamster ovary (CHO) cells transfected with receptor cdnas in pcdna3 (Invitrogen) using the calcium phosphate method [11] and expressing the human mu (hmor), delta (hdor), or kappa (hkor) receptor [12] were used in this study. When 90% con uence was reached, the cells were scraped from the plates and were harvested by centrifugation at 1000Ug for 10 min at 4³C. The cell pellets were then washed once and resuspended in ice-cold binding assay bu er containing 50 mm Tris-HCl (ph 7.8), 1 mm EGTA, 5 mm MgCl 2, 10 Wg/ml leupeptin, 10 Wg/ml pepstatin, 200 Wg/ml bacitracin, and 0.5 Wg/ml aprotinin, and homogenized using a Polytron homogenizer (Brinkmann). The homogenates were centrifuged at 48 000Ug for 20 min at 4³C to recover the membrane fractions. For the binding assay, a membrane sample (50^100 Wg protein) was mixed with a radio-labelled ligand ([ 3 H]DAMGO or [ 3 H]diprenorphine, Amersham Life Science) and a test compound in a nal volume of 500 Wl of the assay bu er in 96-well deep-well polystyrene titer plates (Beckman). The nal concentration of [ 3 H]DAMGO or [ 3 H]diprenorphine after mixing was 1 nm. Following incubation at 22³C for 60 min, binding was terminated by vacuum ltration with a Brandel MPXR-96T Harvester through GF/B lters that had been pretreated with a solution of 0.5% (w/v) polyethyleneimine and 0.1% (w/v) bovine serum albumin. The lter-bottom plates were washed four times with 1 ml each of icecold 50 mm Tris-HCl (ph 7.8). Radioactivity on the lters was then 0014-5793/98/$19.00 ß 1998 Federation of European Biochemical Societies. All rights reserved. PII: S0014-5793(98)01362-3

J. Gong et al./febs Letters 439 (1998) 152^156 153 Table 1 Binding a nity values of endomorphins to human mu, delta, and kappa opioid receptors K i (nm) Mu Delta Kappa Endomorphin-1 5.5 þ 0.4 824.3 þ 362.9 s 10 000 Endomorphin-2 8.1 þ 1.7 s 10 000 s 10 000 Summary of the data from three or more separate experiments. The results are presented as mean þ standard error. The K i values of endomorphins for the mu opioid receptor were determined by displacing [ 3 H]DAMGO bound to the receptor, while the K i values for delta and kappa receptors were estimated by displacement of bound [ 3 H]diprenorphine. determined by scintillation spectrometry in a Packard TopCount in Microscint-20 (Packard Instruments, Meriden, CT, USA). Binding data were analyzed by non-linear regression using the GraphPad Prism software program. The K i values were calculated from IC 50 by the Cheng-Pruso equation [13] using the K d values of [ 3 H]DAMGO and [ 3 H]diprenorphine (4.1 nm for DAMGO binding to the mu opioid receptor and 0.33 nm for diprenorphine binding to the delta receptor). Preliminary experiments were performed to show that no speci c binding was lost during the wash of the lters, that binding was at equilibrium by 60 min and remained at equilibrium for at least an additional 60 min, and that the binding was within the linear range with regard to the protein amounts used for the assay. Non-speci c binding, determined in the presence of 10 WM unlabeled naloxone, was approximately 10% of total binding. 2.2. camp assay CHO cells stably transfected with hmor were seeded in 24-well plates at the density of 5U10 4 cells/well. After incubation at 37³C to reach 90% con uence in a F-12 Nutrient Mixture (HAM, Gibco- BRL) supplemented with 1 mm glutamine, 10% (v/v) fetal calf serum, 100 Wg/ml each of penicillin and streptomycin, and 500 Wg/ml geneticin, the cells were incubated in 0.5 ml of F-12 containing 500 WM of isobutylmethylxanthine (IBMX) at 37³C for 15 min prior to the treatment with 500 WM IBMX, 25 WM forskolin, and agonists at appropriate concentrations in 0.5 ml of F-12 at 37³C for 20 min. The reactions were terminated by adding 0.1 ml of luciferase cell culture lysis reagent (Promega) in 0.1 M HCl after removal of F-12. Aliquots were removed and the concentrations of camp were determined using the radioimmunoassay kit (NEN-DuPont) according to the manufacturer's instructions. The data were analyzed by non-linear regression using the GraphPad Prism software program. 2.3. Oocyte expression and electrophysiology Xenopus oocytes were isolated using standard methods [14]. Synthetic mrnas for the human mu opioid receptor [9] and the G-protein-activated inwardly rectifying K channels, GIRK1 [15,16] and GIRK2 [17,18], were transcribed in vitro, as described [8]. Thirtytwo nl of the mrna solution (1 ng of the mu receptor mrna and 0.5 ng each of GIRK1 and GIRK2 mrnas) was injected into each mature (stage V^VI) oocyte using a Drummond automatic microinjector. The injected oocytes were incubated in 50% (v/v) L-15 medium supplemented with 0.8 mm glutamine and 10 Wg/ml gentamycin at 20³C. Two to three days after injection, oocytes were voltage-clamped at 380 mv using a two-microelectrode voltage clamp, the Axoclamp- 2A (Axon Instruments) under the control of pclamp software (Axon Instruments). The electrodes were lled with 3 M KCl and had tip resistance of 0.5^3.0 M6. During recording, oocytes were superfused with either ND96 (96 mm NaCl, 2 mm KCl, 1 mm MgCl 2, 1.8 mm CaCl 2, and 5 mm HEPES, ph 7.5), a high K solution (HK: 96 mm KCl, 2 mm NaCl, 1 mm MgCl 2, 1.8 mm CaCl 2, and 5 mm HEPES, ph 7.5), or HK containing agonists. The data were analyzed with the Microcal Origin software program. 3. Results 3.1. Binding of endomorphins to human opioid receptors Zadina et al. [5] showed that in rodent brain homogenates, endomorphins bind selectively to mu opioid receptors. To determine the binding selectivity of endomorphins for the Fig. 1. Inhibition of forskolin-stimulated cyclic AMP increase by endomorphins and DAMGO. CHO cells stably transfected with the human mu opioid receptor were treated with 25 WM forskolin in the presence or absence of endomorphins or DAMGO at various concentrations from 0.25 nm to 10 WM. The data for each point were from three determinations of a representative experiment.

154 J. Gong et al./febs Letters 439 (1998) 152^156 Fig. 2. Example of endomorphin-evoked K current mediated by the human mu opioid receptor. Two representative endomorphinevoked current traces at a holding potential at 380 mv are shown. Following superfusion with HK, the oocytes were perfused with HK containing endomorphin-1 (100 nm) alone or followed by endomorphin-1 (100 nm) plus naloxone (10 WM), as indicated by the bars above the current traces. cloned human opioid receptors, we used CHO cells transfected with the human mu, delta, and kappa opioid receptor clones for binding studies. Binding a nity values of endomorphins for the human mu, delta, and kappa opioid receptors are shown in Table 1. Both endomorphins exhibited high a nity and selectivity for the human mu opioid receptor, with little or no binding for the delta or kappa opioid receptors. The estimated K i value of endomorphin-1 for the human mu opioid receptor is 5.5 nm, an a nity 150-fold higher than that for the delta opioid receptor and over 1500-fold higher than that for the kappa opioid receptor, indicating its selectivity for the mu opioid receptor. Similarly, endomorphin-2 has a K i value of 8.1 nm for the mu opioid receptor with very little binding to the delta or kappa opioid receptors. 3.2. Inhibition of forskolin-stimulated cyclic AMP increase Opioid receptors, including the mu opioid receptor, have been shown to inhibit adenylyl cyclase activity and thus reduce the intracellular camp content [6]. To examine the functional e ect of endomorphins on the cloned human mu opioid receptor, the potency with which endomorphins inhibit the increase of camp stimulated by forskolin was determined. As shown in Fig. 1, endomorphins 1 and 2 have IC 50 values of 36.2 and 34.5 nm, respectively, and DAMGO has an IC 50 value of 87.1 nm. These data indicate that both endomorphins, like DAMGO, can exert an inhibitory e ect on the camp/adenylyl cyclase pathway via the cloned human mu opioid receptor. The maximum inhibition of the camp increase achieved in our experiments was 58% for endomorphin-1 (95% con dence level 49^66%), 65% for endomorphin-2 (95% con dence level 56^73%), and 55% for DAMGO (95% con dence level 49^61%). There were no signi cant di erences between these values. Thus, comparable levels of maximum camp inhibition between endomorphins and DAMGO indicate that endomorphins are of similar e cacy as DAMGO. 3.3. Activation of the mu opioid receptor coupled to G-protein-activated K + channels The mu opioid receptor has been shown to activate cloned Fig. 3. E cacy of endomorphins and DAMGO in activation of an inwardly rectifying K current mediated by the human mu opioid receptor. Data in panels A and B were from two di erent experiments, presented as mean þ S.E. The concentration of endomorphin- 1 and DAMGO was 50 or 100 nm in panel A, and 50 nm in panel B. A: The K current evoked by endomorphin-1 or DAMGO. Endomorphin-1: 921 þ 49 na, n = 9; DAMGO: 868 þ 43 na, n =9. B: The K current evoked by endomorphin-2 or DAMGO. Endomorphin-2: 745 þ 77 na, n = 5; DAMGO: 742 þ 82 na, n =5. G-protein-activated K channels in Xenopus oocytes upon activation by its agonists, such as DAMGO [8,9,18]. To examine whether endomorphins can activate the mu opioid receptor in this functional assay, hmor, GIRK1 and GIRK2 were coexpressed in oocytes, and the functional coupling of the receptor to the K channels was assessed by two-electrode voltage clamp. When the receptor was activated by superfusing the oocytes with either of the endomorphins, an inward current was observed (Fig. 2). This K current was induced by activation of the expressed receptor, since it was completely blocked by the opioid receptor antagonist naloxone (Fig. 2). Both endomorphins induced maximum K currents comparable to those induced by the full agonist DAMGO (Fig. 3), indicating that both endomorphins are full agonists at the mu opioid receptor in this functional assay. To examine the potency of endomorphins in activating the mu opioid receptor, the dose e ects of endomorphins were determined. Because of the variability of individual oocytes in expressing exogenous proteins, we standardized the receptor-mediated response by taking the ratio of the test agonistevoked current (I test ) at a particular dose to the maximum current induced by DAMGO at a saturating concentration (100 nm), as indicated in Fig. 4A. The activation of the mu opioid receptor by endomorphins is dose dependent within the range from 0.1 nm to 50 nm (Fig. 4B). The EC 50 values calculated from the dose response curves are 4.6 nm for endomorphin-1, 9.7 nm for endomorphin-2, and 14.0 nm for DAMGO. These data demonstrate that both endomorphins can fully induce the functional coupling between the mu opioid receptor and G-protein-activated K channels with similar e cacy and potency to DAMGO. 4. Discussion Activation of the mu opioid receptor by its agonists has two major cellular e ects. One is the inhibitory e ect on the camp/adenylyl cyclase pathway, resulting in reduced levels of intracellular camp [6,19,20], an important second messenger in cell signalling. The other is the modulation of ion channels, such as increasing K conductance or decreasing Ca 2 conductance, to reduce cellular excitability [8,9,21^24]. In this study, we were interested in whether endomorphin-1 and endomorphin-2, two recently discovered endogenous pep-

J. Gong et al./febs Letters 439 (1998) 152^156 155 Fig. 4. Dose response curves for human mu opioid receptor-mediated activation of K currents by endomorphins and DAMGO. A: An example of a current trace showing the experimental protocol and calculation method for the agonist-induced response. Oocytes were clamped at a holding potential of 380 mv and superfused with di erent solutions as indicated. Application of agonists in HK started at the points indicated by the arrows. I k : the basal inward K currents in HK. I max : maximum K currents evoked by DAMGO at a saturating concentration (100 nm). I test :K currents evoked by the test dose of endomorphins or DAMGO. B: Dose response curves of receptor activation. The tested concentrations of agonists ranged from 0.1 nm to 1 WM. The response to a test dose is expressed as the fraction of the maximum activation (I test / I max ). Data are presented as mean þ S.E. (n = 4^5). All oocytes were used only once to avoid desensitization. The EC 50 values calculated from the curves are 4.6 nm for endomorphin-1, 9.7 nm for endomorphin-2, and 14.0 nm for DAMGO. tides with high a nity and selectivity for the mu opioid receptor [5], can activate the human mu opioid receptor to exert these two major cellular e ects. Our binding data demonstrated that both endomorphins bound to the mu opioid receptor with a nity values comparable to that with the high a nity agonist DAMGO, and both endomorphins were highly selective for the human mu opioid receptor over the delta and kappa opioid receptors (Table 1). These observations are consistent with those reported by Zadina et al. [5] in rodent brain tissues which contain several types of opioid receptors. The e ect of agonists on activation of opioid receptors and their potency and e cacy in the activation can be determined in functional assays, as activation of mu opioid receptors by agonists results in reduction in intracellular levels of camp and in activation of co-expressed K channels in Xenopus oocytes to produce an inward current [25]. In this study, both endomorphins were shown to reduce the intracellular level of camp and to evoke an inward K current with high potency by activation of the cloned human mu opioid receptor (Figs. 1, 3 and 4B), supporting the initial observation in functional assays by Zadina et al. [5] using the guinea-pig ileum system. During the preparation of this report, two separate studies published in which endomorphins were compared with DAM- GO using mu opioid receptor-stimulated [ 35 S]GTPQS binding as the assay [26,27]. Based on this assay, the authors reported that endomorphins are partial agonists at the mu opioid receptor. The mu opioid receptor similar to other G proteincoupled receptors, achieves its cellular activity by rst activating G proteins. Thus, receptor-stimulated [ 35 S]GTPQS binding serves as a valuable marker to indicate the ability of an agonist to activate the receptor qualitatively. Whether the extent of [ 35 S]GTPQS binding parallels the e cacy of an agonist quantitatively remains to be established. In this context, it is interesting to speculate on the di erence between the outcome of these two studies and that in this report. We examined the major cellular functions of the mu opioid receptor: inhibition of camp synthesis and activation of K channels. In both functional assays, the endomorphins showed full agonist activity as compared with DAMGO, a well-recognized full agonist at the mu opioid receptor. One explanation for the di erent conclusions from our study and those by Sim et al. [26] and Hosohata et al. [27] is that, while endomorphins may not stimulate [ 35 S]GTPQS binding to the same extent as that by DAMGO, the amount of activated G protein is su cient for a full-scale cellular e ect. In summary, this study showed that endomorphins are selective full agonists for the cloned human mu opioid receptor, capable of conferring high a nity binding and e cacious activation of the human mu opioid receptor. Thus, the discovery of endomorphins by Zadina et al. [5] opens the door to further exploration of the physiological roles of these endogenous peptides. Acknowledgements: This work was supported by National Institutes of Health Grants DA09444 and DA11891. References [1] Pasternak, G.W. (1993) Clin. Neuropharmacol. 16, 1^18. [2] Pasternak, G.W., Childers, S.R. and Snyder, S.H. (1980) Science 208, 514^516. [3] Chaillet, P., Coulaud, A., Zajac, J.M., Fournie-Zaluski, M.C., Costentin, J. and Roques, B.P. (1984) Eur. J. Pharmacol. 101, 83^90. [4] Akil, H., Watson, S.J., Young, E., Lewis, M.E., Khachaturian, H. and Walker, J.M. (1984) Annu. Rev. Neurosci. 7, 223^255. [5] Zadina, J.E., Hackler, L., Ge, L.J. and Kastin, A.J. (1997) Nature 386, 499^502. [6] Childers, S.R. (1991) Life Sci. 48, 1991^2003. [7] North, R.A. (1993) in: Handbook of Experimental Pharmacology, Vol. 104, Opioids I (Herz, A., Ed.) pp. 773^797, Springer, Berlin. [8] Chen, Y. and Yu, L. (1994) J. Biol. Chem. 269, 7839^7842. [9] Mestek, A., Hurley, J.H., Bye, L.S., Campbell, A.D., Chen, Y., Tian, M., Schulman, H. and Yu, L. (1995) J. Neurosci. 15, 2396^ 2406. [10] Kovoor, A., Henry, D.J. and Chavkin, C. (1995) J. Biol. Chem. 270, 589^595. [11] Chen, C. and Okayama, H. (1987) Mol. Cell. Biol. 7, 2745^ 2752.

156 [12] Zhang, S., Tong, Y., Tian, M., DeHaven, R.N., CortesBurgos, L., Mansson, E., Simonin, F., Kie er, B.L. and Yu, L. (1998) J. Pharmacol. Exp. Ther. 286, 136^141. [13] Cheng, Y.-C. and Pruso, W.H. (1973) Biochem. Pharmacol. 22, 3099^3108. [14] Gurdon, J.B. and Wickens, M.P. (1983) Methods Enzymol. 101, 370^386. [15] Dascal, N., Lim, N.F., Schreibmayer, W., Wang, W., Davidson, N. and Lester, H.A. (1993) Proc. Natl. Acad. Sci. USA 90, 6596^ 6600. [16] Kubo, Y., Reuveny, E., Slesinger, P.A., Jan, Y.N. and Jan, L.Y. (1993) Nature 364, 802^806. [17] Patil, N., Cox, D.R., Bhat, D., Faham, M., Myers, R.M. and Peterson, A.S. (1995) Nat. Genet. 11, 126^129. [18] Tong, Y., Wei, J., Zhang, S., Strong, J.A., Dlouhy, S.R., Hodes, M.E., Ghetti, B. and Yu, L. (1996) FEBS Lett. 390, 63^68. J. Gong et al./febs Letters 439 (1998) 152^156 [19] Chen, Y., Mestek, A., Liu, J., Hurley, J.A. and Yu, L. (1993) Mol. Pharmacol. 44, 8^12. [20] Chen, Y., Liu, J. and Yu, L. (1996) Addict. Biol. 1, 49^59. [21] Williams, J.T., Egan, T.M. and North, R.A. (1982) Nature 299, 74^77. [22] North, R.A., Williams, J.T., Surprenant, A. and Christie, M.J. (1987) Proc. Natl. Acad. Sci. USA 84, 5487^5491. [23] Wimpey, T.L. and Chavkin, C. (1991) Neuron 6, 281^289. [24] Twitchell, W.A. and Rane, S.G. (1994) Mol. Pharmacol. 46, 793^ 798. [25] Yu, L. (1996) Addict. Biol. 1, 19^30. [26] Sim, L.J., Liu, Q., Childers, S.R. and Selley, D.E. (1998) J. Neurochem. 70, 1567^1576. [27] Hosohata, K., Burkey, T.H., Alfaro-Lopez, J., Varga, E., Hruby, V.J., Roesk, W.R. and Yamamura, H.I. (1998) Eur. J. Pharmacol. 346, 111^114.