PII: S0014-5793(99)00473-1 Neuronal nAChR stereoselectivity to non-natural epibatidine derivatives Sonia Bertranda, Jo« rg T. Pattb, Jo« rg E. Spangb, Gerrit Westerab, P. August Schubigerb, Daniel Bertranda;* aDëpartement de physiologie, Centre Mëdical Universitaire (Facultë de Mëdecine), 1 rue Michel Servet, CH-1211 Geneva 4, Switzerland bCenter for Radiopharmaceutical Science, Swiss Federal Institute of Technology Zu« rich, Paul Scherrer Institute Villigen and Department of Radiology, Clinic of Nuclear Medicine, University Hospital, Zu« rich, Switzerland Received 17 March 1999 Abstract The frog toxin epibatidine is one of the most powerful ligands of the neuronal nicotinic receptors and derivatives show promising possibilities for labeling in positron emission tomo- graphy studies. In an attempt to reduce epibatidine toxicity, new methyl derivatives were synthesized, tested in positron emission tomography imaging and in electrophysiology. labeling as well as physiological experiments highlighted the differences in sensitiv- ity of the neuronal nicotinic acetylcholine receptors between two methyl enantiomers and the reduction in sensitivity caused by introducing the methyl group. At present, epibatidine derivatives seem the most promising compounds for in vivo labeling of neuronal nicotinic acetylcholine receptors. z 1999 Federation of European Biochemical Societies. Key words : Nicotinic acetylcholine receptor ; Brain ; Epibatidine ; Pharmacophore ; Imaging 1. Introduction With the introduction of tobacco plants on the second voy- age of Christopher Columbus its use and smoking addiction quickly spread amongst a large fraction of the population. Although it was recognized earlier that the alkaloid nicotine is the active compound in tobacco leaves and that it is a potent agonist of the cholinergic system at the neuromuscular and ganglionic synapses, the mechanisms by which it causes addiction remained obscure [1,2]. The identi¢cation of an en- tire family of genes encoding the neuronal nicotinic acetylcho- line receptors (nAChRs) that are speci¢cally expressed at the neuromuscular junction or in nerve cells shone a new light on the mechanisms by which nicotine can exert its action in the central nervous system [3,4]. One of the critical ¢ndings was the observation that neuronal nAChRs display a wide pattern of physiological and pharmacological pro¢les and that a given set of subunits is expressed in a well-de¢ned brain area [5^7]. For instance, it was shown that the major brain neuronal nAChR results from the assembly of the K4 and L2 subunits. Moreover, a high level of expression of this class of receptor was observed on dopaminergic neurons that are known to participate in reward and addictive behaviors [8,9]. With the introduction of in situ hybridization using speci¢c mRNA probes it has become possible to map the pattern of expression of the identi¢ed neuronal nAChR subunits in rat, mouse or human brains [7,10]. Although allowing a high spa- tial resolution the use of this technique cannot be employed for in vivo measurements. The development of new tools is therefore required to establish the relationship between the in situ pattern of neuronal nAChR expression in a de¢ned brain area and physiological behavior. The availability of positron emission tomography (PET) ligands would constitute a ¢rst alternative to postmortem hybridization or biochemical stud- ies and make it possible to correlate nicotine labeling with cognitive or behavioral status. Initial studies performed with radioactive nicotine have re- vealed the inadequacy of this compound in PET studies and that more speci¢c ligands must be identi¢ed to obtain proper spatial and temporal resolutions [11,12]. The identi¢cation of a new frog toxin, epibatidine, which displays a very high af- ¢nity for the neuronal nAChRs, opened new possibilities to design synthetic compounds of potential interest for PET monitoring [13^17]. We produced N-methylated epibatidine for this purpose [13]. Recently this and other methylated epi- batidine analogs were synthesized, with very similar biological properties (e.g. receptor a¤nity and toxicity) [16]. The aim of this work is to examine the properties of the optical isomers of N-methylepibatidine on reconstituted neuronal nAChRs and to determine their potential application for PET investiga- tions. 2. Materials and methods 2.1. Chemistry All chemicals were purchased in analytical quality from Aldrich, Fluka, Merck or Sigma unless otherwise mentioned. The 1H NMR and 13C NMR spectra were recorded on Varian Gemini 200 and Gemini 300 spectrometers. Mass spectra were recorded on a Trio 2000 spectrometer (VG Organic, UK) using the positive ion mode with electrospray (ES+). The separation of the enantiomers was performed by HPLC on a semi-preparative ChirobioticT column (250U10 mm) from Astec and Merck LaChrom equipment (D-7000, L-7100, L-7250, L-7450). Meth- anol was used in preparative HPLC quality from Merck. Optical rotations were measured with a Perkin Elmer 241 polarimeter at 23³C using a sodium lamp (D-line) and are reported as speci¢c rota- tion [K] in degrees. The purity is reported in percent of enantiomeric excess (%ee). The 11C-labeled compounds were produced as described in detail elsewhere [18] by methylation of the desmethyl precursor with [11C]MeI. 2.2. PET studies in the rat Male Sprague-Dawley rats (300^400 g) were anesthetized with Do- mitor and Hypnorm (0.25 ml/kg body weight), according to the pro- cedure required by the veterinary department. Anesthesia was main- tained during the study by subcutaneous injection of Hypnorm/ Domitor (0.12 ml/kg body weight). A catheter was introduced into the tail vein. The rat was placed in the PET scanner (GE-Advance) with the whole body of the rat in the ¢eld of view. Prior to the PET 0014-5793 / 99 / $20.00 ß 1999 Federation of European Biochemical Societies. All rights reserved. PII : S 0 0 1 4 - 5 7 9 3 ( 9 9 ) 0 0 4 7 3 - 1 *Corresponding author. Fax : (41) (22) 702 54 02. E-mail : bertrand@cmu.unige.ch Abbreviations : ACh, acetylcholine ; nAChR, nicotinic receptor ; CNS central nervous system ; PNS, peripheral nervous system FEBS 21938 4-5-99 Cyaan Magenta Geel Zwart FEBS 21938 FEBS Letters 450 (1999) 273^279 scans a transmission scan was performed. An amount of 1^2 MBq of the 11C compound was injected. Data acquisition was started and 22 frames were collected 10U0.5 min, 5U1 min, 2U5 min, 4U10 min and 1U20 min. Regions of interest (ROI) were drawn directly on the `transaxial' PET scans, which are sagittal as a result of the positioning of the rat in the ¢eld of view. The activity in the ROIs was calculated from the absolute count rate by multiplication with area and slice thickness (0.425 cm). The result is displayed as percent injected dose. The amount of activity injected into the rat was calculated from a whole body ROI and the percentage of 11C tracer in the brain was deter- mined as a function of midframe time. The result of the whole body ROI was compared to the amount of activity injected into the rat determined by measuring the syringe before and after injection in a dose calibrator. The values determined by whole body ROI were in agreement with the 11C dose determined with the dose calibrator. 2.3. Electrophysiology Xenopus oocytes were prepared according to the standard proce- dure [19] and nuclear-injected with 10 nl of bu¡er containing equal concentrations (0.1 Wg/Wl) of K and L subunit cDNAs or 0.2 Wg/Wl of K7. Recordings were made using the two-electrode voltage clamp technique. During the experiments, oocytes were continuously super- fused with control solution containing : 82.5 mM NaCl, 2.5 mM KCl, 5 mM HEPES, 2.5 mM CaCl2, 1 mM MgCl2, pH 7.4 adjusted with NaOH. All chemicals were obtained from either Fluka or Sigma (Buchs, Switzerland). Dose-response relationships were adjusted to the empirical Hill equation y� 1 1� EC50 x � �nH �1� Fig. 1. Chemical structure of epibatidine and its methyl derivatives. The upper panel presents the two enantiomers of epibatidine or its methyl derivative. Chemical synthesis of the radiolabeled compound is schematized in the lower panel. Fig. 2. Currents evoked by epibatidine and its derivatives on the major brain K4L2 nAChR. A : Typical currents evoked in a responsive oocyte by short application (2 s) of ACh, (+)-epibatidine and methyl-(+)-epibatidine at saturating concentration. B : Dose-response relationships for (+)-epibatidine and methyl-(+)-epibatidine (mean of four cells). Lines through the data points correspond to the best ¢ts obtained with Eq. 1. C : Currents evoked, in another cell, as in A for ACh, (3)-epibatidine and methyl-(3)-epibatidine. D : Dose-response relationships for (3)-epi- batidine and methyl-(3)-epibatidine (mean of four cells, values in Table 1). Data in B and D were normalized with respect to currents evoked by a saturating ACh concentration. FEBS 21938 4-5-99 Cyaan Magenta Geel Zwart S. Bertrand et al./FEBS Letters 450 (1999) 273^279274 where y = the fraction of activated current, EC50 = concentration of agonist evoking a current of half maximal amplitude, nH = Hill coef- ¢cient, x = agonist concentration. Dose-response curves for (+)- and (3)-epibatidine and their analogs were normalized on an acetylcholine (ACh) saturating concentration. All values indicated throughout the text are given with their respective S.E.M. Cells were held throughout the experiments at 3100 mV. 2.4. Toxicology The acute intravenous toxicity of methylepibatidine was estimated in mice and rats by a specialized outside agency (RCC Research and Consulting Co. Ltd., Itingen, Switzerland). A racemic mixture of methylepibatidine was intravenously administered to groups of ¢ve male and ¢ve female animals. The doses were 0.04, 0.4, 8 and 80 Wg/kg for the HanIbm :WIST (SPF) rats and 0.05, 0.5, 10 and 100 Wg/kg for the HanIbm :NMRI (SPF) mice. Surviving animals were examined four to ¢ve times on day 1 and once daily on days 2^15 for clinical signs. Mortality/viability were recorded together with clin- ical signs at the same time intervals. All animals were necropsied and examined macroscopically. 3. Results and discussion Neuronal nAChRs are integral membrane proteins that are thought to result from the assembly of ¢ve subunits around an axis of pseudosymmetry [20]. Earlier physiological and pharmacological experiments have revealed that ganglionic receptors from the peripheral nervous system (PNS) display patterns of agonists and antagonists clearly distinguishable from those of the muscle receptor [2,20]. Reinforced by many recent studies, these di¡erences can be attributed to a particular expression of neuronal subtypes (reviewed in [20]). Furthermore, it was shown that ganglionic receptors contain at least the K3 and L4 subunits while central nervous system (CNS) receptors are thought to result from the assembly of K4 with L2 [7,21]. A less clear distinction can be made when considering receptors containing the K7 subunit which have been shown to be present in both the CNS and the PNS [6,22^ 24]. To examine the e¡ects of epibatidine and its N-methyl derivatives on the CNS and PNS nAChR types we determined the action of these compounds on neuronal receptors recon- stituted in Xenopus oocytes. In a recent contribution Horti et al. [16] studied N-alkylated epibatidine derivatives with various halogens, £uorine, bro- mine and iodine, at the position of the chlorine on the pyr- idine ring. They found a¤nities to rat brain homogenates, mouse brain distributions and toxicity towards mice which were all very similar. We concentrated on the di¡erences be- tween the enantiomeric N-methyl compounds in their dynamic behavior in the rat brain and in their activation of various nAChR subtypes. A schematic representation of the two enantiomers of epi- batidine and N-methylepibatidine employed throughout this work is given in Fig. 1. 3.1. Sensitivity of the rat K4L2 to epibatidine and methylepibatidine Oocytes were injected with rat cDNA combinations K4L2 and their physiological properties were examined using a dual- Fig. 3. Currents evoked by epibatidine and its derivatives on the ganglionic K3L4 nAChR. A and C : Typical currents evoked in a responsive oocyte by a short application (2 s) of ACh, epibatidine and its derivative at saturating concentration as in Fig. 2A,C. B : Dose-response rela- tionships for (+)-epibatidine and methyl-(+)-epibatidine (mean of four cells). Lines through the data points correspond to the best ¢ts obtained with Eq. 1. D : Dose-response relationships for (3)-epibatidine and methyl-(3)-epibatidine (mean of four cells, values are indicated in Table 1). All procedures were as described in Fig. 2. FEBS 21938 4-5-99 Cyaan Magenta Geel Zwart S. Bertrand et al./FEBS Letters 450 (1999) 273^279 275 electrode voltage clamp. Typical currents evoked, in a respon- sive oocyte, by acetylcholine (ACh), (+)-epibatidine and meth- yl-(+)-epibatidine are illustrated in Fig. 2A. The equal ampli- tude of currents evoked by these saturating test pulses indicates that these three compounds act as full agonists on the CNS receptor. Determination of dose-response curves (Fig. 2B) over a broad range of agonist concentrations re- vealed that the K4L2 receptor is about 15-fold more sensitive to epibatidine than its equivalent methyl derivative. When the same experiments were repeated with the (3) enantiomers a di¡erent picture was observed. Although no di¡erences in amplitude could be observed between the currents evoked by ACh and (3)-epibatidine a small reduction was observed with methyl-(3)-epibatidine (Fig. 2C). On average, however, no signi¢cant di¡erences could be detected between these three agonists. The small reduction observed on these currents can be partly attributed to the fastest desensitization of the receptor to methyl-(3)-epibatidine. Surprisingly, however, dose-response relationships (Fig. 2D) yielded no di¡erences in apparent a¤nity between (3)-epibatidine and methyl-(3)- epibatidine indicating that introduction of the methyl side chain does not alter the receptor sensitivity for the (3) enan- tiomer (data summarized in Table 1). 3.2. Sensitivity of the ganglionic K3L4 epibatidine and its derivatives Analysis of the currents evoked by the two enantiomers of epibatidine and methylepibatidine were carried out as de- scribed above for the CNS nAChR subtype. As shown in Fig. 3A, the (+) enantiomers of epibatidine and methylepiba- tidine both evoked robust currents of comparable amplitude to the natural agonist ACh. Moreover, as seen for the K4L2 nAChR, the K3L4 receptor displays a sensitivity roughly 30 times lower for the (+)-methyl compound than for the native toxin (Fig. 3B). Assessing the properties of the (3) enantiom- ers yielded results comparable to those observed with the K4L2 nAChR. Namely, while both epibatidine and methyl- epibatidine evoked currents of similar amplitude as those re- corded in response to ACh (Fig. 3C), exposure to (3)-epiba- tidine and methyl-(3)-epibatidine yielded approximately similar half activation values (Fig. 3D). Thus, whereas this class of receptor does not distinguish between the (+)- and (3)-epibatidine enantiomers it is about 31-fold more sensitive to the (3) form of the methyl derivative than to its (+) enan- tiomer (see Table 1). 3.3. Sensitivity of the homomeric K7 nAChR It is well documented that the homomeric K7 receptor is strikingly less sensitive to epibatidine than the heteromeric receptors [25]. For instance, whilst the major brain receptor displays a more than two orders of magnitude higher a¤nity for epibatidine than for ACh, the K7 sensitivity remains in the micromolar range. In addition, it is also known that this re- ceptor shows a higher desensitization rate when exposed to epibatidine than when challenged with ACh. Current traces recorded in response to a fast application pulse display the typical pro¢le reported earlier for the homomeric K7 receptor [23,26,27]. Unlike the heteromeric K4L2 or K3L4 receptors, Fig. 4. Sensitivity of the K7 nAChR to epibatidine and its methyl derivatives. Traces recorded in response to (+) enantiomers are shown in A while currents evoked by the (3) enantiomers are shown in C. Note the di¡erences in amplitude of the currents evoked by (+)- and (3)-epiba- tidine. B and D : Ddose-response relationships of the homomeric K7 nAChR (mean of four cells, see Table 1) to epibatidine. Data were ob- tained and processed as in Fig. 2. FEBS 21938 4-5-99 Cyaan Magenta Geel Zwart S. Bertrand et al./FEBS Letters 450 (1999) 273^279276 Fig. 5. PET images of [11C]N-methyl-(+)- and (3)-epibatidine in rats. The region of the liver, urinary bladder and brain showed a high uptake of the tracer. The (+) isomer (A) showed a higher uptake in the region of liver, stomach and spleen compared to the (3) isomer (B). Because of the limited resolution of the PET images, only the total the brain activity curve is shown as % injected dose versus time in Fig. 6. FEBS 21938 4-5-99 Cyaan Magenta Geel Zwart S. Bertrand et al./FEBS Letters 450 (1999) 273^279 277 however, both (+)-epibatidine and methyl-(+)-epibatidine evoked only a fraction of the currents caused by ACh expo- sure (Fig. 4A). Furthermore, as illustrated in Fig. 4B, adjunc- tion of the methyl residue further decreased the apparent af- ¢nity of the K7 receptor to epibatidine by more than one order of magnitude. Data obtained with the (3) enantiomers are roughly comparable to the observation made on the het- eromeric receptors (Fig. 4C). A marked di¡erence is, however, found in the partial agonist mode of action of the (3)-methyl compound (see Fig. 4D and Table 1), which is not observed with the other receptor subtypes. Taken together these data highlight the ¢nding that intro- duction of a methyl side chain causes a decrease in the e¤cacy of the (+) enantiomer and introduces a stereoselectivity that is not observed with the native epibatidine. It is of interest to recall that although the neuronal nAChRs display a poor selectivity in the separation of enantiomers a di¡erence be- tween (3)- and (+)-nicotine has been widely documented. However, another chemical modi¢cation of epibatidine, the removal of the chlorine atom, introduced no stereoselectivity in the molecular recognition by the receptors [28]. Thus, the stereoselectivity caused by the introduction of a methyl sub- stitute onto the nitrogen bridge of the bicyclocyclohexyl ring indicates that this segment of the molecule must be in closer interaction with the ACh binding site than the chlorine atom. 3.4. PET studies with N-[11C]methyl(+)- and (3)-epibatidine To characterize further the possibility of using these epiba- tidine derivatives in PET studies, a ¢rst set of experiments was conducted on rats. Pictures obtained after injection with N- [11C]methyl(+)-epibatidine reveal that the concentration of this compound transiently increased in the brain and then progressively diminished (Fig. 5A). In contrast, when the same experiment was repeated with the (3) enantiomer a di¡erent pattern was revealed. While the signal observed in the liver or kidneys progressively decreased the brain uptake continued to increase (Fig. 5B). Comparison of the PET im- ages and the brain uptake curves showed interesting di¡er- ences in receptor binding properties of the methylated epi- batidine stereoisomers. [11C]N-methyl-(3)-epibatidine binds predominantly to one region in the rat brain. In contrast, two binding sites were seen for [11C]N-methyl-(+)-epibatidine. The additional binding site seen very clearly for the (+) isomer was most likely the eyes of the rat, but because of limited resolution of the PET scan some other brain region such as olfactory bulb or frontal cortex may also show an enrichment of the tracer. Considerable di¡erences were seen when com- paring the time course of brain uptake for the enantiomers of [11C]N-methyl-epibatidine. While [11C]N-methyl-(3)-epibati- dine showed both a fast and slower uptake and an increasing brain activity with a time constant of about 40 s, the (+) enantiomer showed only a transient peak and a slow decay (Fig. 6). Washout of the ligand could not be determined on this fast time scale measurement. A typical washout time con- stant of 3.8 h was measured with single photon emission computed tomography for radioiodinated epibatidine cerebel- lum clearance in baboon [15]. The slowest clearance measured by these authors for this compound was in the thalamus with a clearance time of 11 h. In the toxicity measurements no deaths occurred in the animal population with the two lower doses (0.04, 0.4 Wg/kg for rats and 0.05, 0.5 Wg/kg for mice), while many animals died at the two higher doses (8, 80 Wg/kg for rats and 10, 100 Wg/kg for mice). The LD50 in rats was 20 Wg/kg (95% con¢dence limits 8^180 Wg/kg) and for mice 15 Wg/kg (95% con¢dence limits 3^120 Wg/kg). To break the data down for males and females makes no statistical sense. The clinical signs of toxicity observed in the two high dose groups in- cluded sedation, convulsions, ventral recumbency and dys- pnea indicating an e¡ect on the nerve system. No clinical signs were observed in the low dose groups. No organ abnormal- ities were observed on necroscopy. Thus, it seems that the toxicity of N-methylepibatidine is possibly somewhat less than that of the analogous N-methyl- norchloro£uoroepibatidine [29]. Although adjunction of a methyl side chain to epibatidine reduces the toxic e¡ects somewhat, this is still not su¤cient to ensure the complete Fig. 6. Time course of [11C]N-methyl-(+)- and (3)-epibatidine bind- ing. Quanti¢cation of PET measurements was done as described in Section 2. Squares indicate the values measured with [11C]N-methyl- (+)-epibatidine while data obtained with [11C]N-methyl-(3)-epibati- dine are represented by circles. Lines through data points were drawn to guide the eye. Table 1 Sensitivity of epibatidine and derivatives to rat K4L2, K3L4 and K7 nAChRs cDNA type (+)-Epibatidine Methyl-(+)-Epibatidine (3)-Epibatidine Methyl-(3)-Epibatidine EC50 (WM) nH EC50 (WM) nH EC50 (WM) nH EC50 (WM) nH Rat K7 2.5 þ 016 1.45 þ 0.1 30 þ 0.24 1.60 þ 0.2 2.03 þ 0.5 1.43 þ 0.2 2.5 þ 0.26 2.73 þ 0.2 Rat K4L2 0.021 þ 0.005 1.24 þ 0.1 0.36 þ 0.11 0.45 þ 0.1 0.023 þ 0.007 1.30 þ 0.1 0.024 þ 0.008 1.25 þ 0.1 Rat K3L4 0.036 þ 0.013 1.52 þ 0.4 1.1 þ 0.2 1.60 þ 0.1 0.019 þ 0.006 1.67 þ 0.2 0.035 þ 0.017 1.70 þ 0.1 Half activation values (EC50) and Hill coe¤cients for three receptor subtypes of the four chemical compounds tested are indicated. Values are the means of 3^7 cells with their respective S.E.M. FEBS 21938 4-5-99 Cyaan Magenta Geel Zwart S. Bertrand et al./FEBS Letters 450 (1999) 273^279278 safety required for human administration, which is in agree- ment with the conclusions by Horti et al. [16]. Thus, although indicating the feasibility of the method, new epibatidine de- rivatives await to be designed to accommodate the properties required both for PET and for human safety. These data illustrate, however, the importance of isolating the enantiomers and of physiological testing for their charac- terization. Acknowledgements : We are very grateful to J. Patrick for kindly pro- viding rat cDNAs. This work was supported by grants from the O¤ce Fëdëral de l'Education et des Sciences and the Swiss National Science Foundation, 3100-0371.91 to D.B. and 3200-046939.96 to G.W. References [1] Langley, J.N. (1905) J. Physiol. 33, 374^413. [2] Ascher, P., Large, W.A. and Rang, H.P. (1979) J. Physiol. 295, 139^170. [3] Benowitz, N.L. (1996) Annu. Rev. Pharmacol. Toxicol. 36, 597^ 613. [4] Corrigall, W.A., Coen, K.M. and Adamson, K.L. (1994) Brain Res. 653, 278^284. [5] Cimino, M., Marini, P., Colombo, S., Andena, M., Cattabeni, F., Fornasari, D. and Clementi, F. (1995) J. Neural Transm. Gen. Sect. 100, 77^92. [6] Deltoro, E.D., Juiz, J.M., Smillie, F.I., Lindstrom, J. and Criado, M. (1997) Dev. Brain Res. 98, 125^133. [7] Wada, E., Wada, K., Boulter, J., Deneris, E., Heinemann, S., Patrick, J. and Swanson, L.W. (1989) J. Comp. Neurol. 284, 314^335. [8] Nitta, A., Katono, Y., Itoh, A., Hasegawa, T. and Nabeshima, T. (1994) Pharmacol. Biochem. Behav. 49, 807^812. [9] Balfour, D.J. (1994) Addiction 89, 1419^1423. [10] Zoli, M., Le, N.N., Hill, J.J. and Changeux, J.P. (1995) J. Neuro- sci. 15, 192^1939. [11] Nyback, H., Halldin, C., Ahlin, A., Curvall, M. and Eriksson, L. (1994) Psychopharmacology (Berl.) 115, 31^36. [12] Muric, R.F., Berridge, M.S., Friedland, R.P., Zhu, N. and Nel- son, A.D. (1998) J. Nucl. Med. 39, 2048^2054. [13] Patt, J.T., Westera, G., Buch, A., Fletcher, S.R. and Schubiger, P.A. (1995) J. Lab. Comp. Radiopharm. 17, 355^356. [14] Villemagne, V.L. et al. (1997) J. Nucl. Med. 38, 1737^1741. [15] Musachio, J.L., Villemagne, V.L., Sche¡el, U., Stathis, M., Fin- ley, P., Horti, A., London, E.D. and Dannals, R.F. (1997) Syn- apse 26, 392^399. [16] Horti, A.G. et al. (1998) J. Med. Chem. 41, 4199^4206. [17] Kassiou, M., Sche¡el, U.A s , Ravert, H.T., Mathews,, W.B., Musachio, J.L., London, E.D. and Dannals, R.F. (1998) Life Sci. 63, L13^18. [18] Patt, J.T., Spang, J.E., Westera, G., Buck, A. and Schubiger, P.A. (1999) Nucl. Med. Biol. (in press). [19] Bertrand, D., Cooper, E., Valera, S., Rungger, D. and Ballivet, M. (1991) Methods Neurosci. 4, 174^193. [20] Bertrand, D. and Changeux, J.P. (1995) Semin. Neurosci. 7, 75^ 90. [21] Boyd, R.T., Jacob, M.H., McEachern, A.E., Caron, S. and Berg, D.K. (1991) J. Neurobiol. 22, 1^14. [22] Conroy, W.G. and Berg, D.K. (1995) J. Biol. Chem. 270, 4424^ 4431. [23] Sëguëla, P., Wadiche, J., Dineley-Miller, K., Dani, J.A. and Pat- rick, J.W. (1993) J. Neurosci. 13, 596^604. [24] Alkondon, M. and Albuquerque, E.X. (1995) J. Pharmacol. Exp. Ther. 274, 771^782. [25] Gerzanich, V., Peng, X., Wang, F., Wells, G., Anand, R., Fletch- er, S. and Lindstrom, J. (1995) Mol. Pharmacol. 48, 774^782. [26] Couturier, S. et al. (1990) Neuron 5, 847^856. [27] Gopalakrishnan, M. et al. (1995) Eur. J. Pharmacol. Mol. Phar- macol. 290, 237^246. [28] Sche¡el, U., Taylor, G.F., Kepler, J.A., Carroll, F.I. and Kuhar, M.J. (1995) NeuroReport 6, 2483^2488. [29] Ding, Y.S., Molina, P.E., Fowler, J.S., Logan, J., Volkow, N.D., Kuhar, M.J. and Caroll, F.I. (1999) Nucl. Med. Biol. 26, 139^ 148. FEBS 21938 4-5-99 Cyaan Magenta Geel Zwart S. Bertrand et al./FEBS Letters 450 (1999) 273^279 279