Agonistas Nicotona x Catecolaminas

download Agonistas Nicotona x Catecolaminas

of 6

Transcript of Agonistas Nicotona x Catecolaminas

  • 7/27/2019 Agonistas Nicotona x Catecolaminas

    1/6

    Research report

    Pharmacological characterization of dopamine, norepinephrine and

    serotonin release in the rat prefrontal cortex by neuronal nicotinic

    acetylcholine receptor agonists

    Tadimeti S. Rao*, Lucia D. Correa, Pamala Adams, Emily M. Santori, Aida I. Sacaan1

    Merck Research Laboratories, 3535 General Atomics Court, San Diego, CA 92121, USA

    Accepted 4 August 2003

    Abstract

    Neuronal nicotinic acetylcholine receptors (nAChRs) modulate synaptic transmission by regulating neurotransmitter release, an action

    that involves multiple nAChRs. The effects of four nAChR agonists, nicotine (NIC), 1,1-dimethyl-4-phenylpiperzinium iodide (DMPP),

    cytisine (CYT) and epibatidine (EPI) were investigated on [3H]-norepinephrine (NE), [3H]-dopamine (DA) and [3H]-serotonin (5-HT) release

    from rat prefrontal cortical (PFC) slices. All four agonists evoked [3H]-DA release to a similar magnitude but with a differing rank order of

    potency of EPIHDMPPcNICcCYT. Similarly, all four agonists also increased [3H]-NE release, but with a differing rank order of

    potency of EPIHCYTcDMPP>NIC. NIC-induced [3H]-NE and [3H]-DA release responses were both calcium-dependent and attenuated

    by the sodium channel antagonist, tetrodotoxin (TTX) and by the nAChR antagonists mecamylamine (MEC) and dihydro-h-erythroidine

    (DHhE), but not by D-tubocurare (D-TC). The modulation of [3H]-5-HT release by nAChR agonists was distinct from that seen for

    catecholamines. DMPP produced robust increases with minimal release observed with other agonists. DMPP-induced [ 3H]-5-HT release was

    neither sensitive to known nAChR antagonists nor dependent on external calcium. The differences between nicotinic agonist induced

    catecholamine and serotonin release suggest involvement of distinct nAChRs.

    D 2003 Elsevier B.V. All rights reserved.

    Theme: D-Neurotransmitters, modulators, transporters and receptors

    Topic: Acetylcholine receptors, nicotinic

    Keywords: Nicotine; Neuronal nicotinic acetylcholine receptor; Prefrontal cortex; Catecholamine and serotonin release

    1. Introduction

    Neuronal nicotinic acetylcholine receptors (nAChRs)

    regulate catecholaminergic and cholinergic neurotransmis-

    sion in several brain regions ([4,7,11,13,1719,20,23,27

    29], reviewed in Ref. [37]). The nAChR regulation of

    dopamine (DA) release has been extensively studied in

    the projection areas of the nigrostriatal pathway (e.g.,

    striatum [4,7,11,13,23,28]) and/or in the mesolimbic path-

    way (e.g., nucleus accumbens [27]) as these pathways

    contain nAChRs [3]. The nAChR regulation of NE release

    was also examined in the hippocampus [4,17,28,35]. A

    limited number of studies have evaluated nAChR regulation

    of 5-HT release in the striatum and hippocampus

    [10,12,15,35,36]. Several lines of evidence suggest a dif-

    ferential regulation by nAChRs of DA in the striatum and

    NE release in the hippocampus [4,2830]. In addition, the

    pharmacology of nAChR regulation of hippocampal 5-HT

    appears to be quite different from that of NE release [15].

    These results suggest that multiple subtypes of nAChRs are

    involved in regulating neurotransmission in different brain

    regions and are consistent with the molecular diversity of

    nAChRs [26,31,39]. To date, investigation of nAChR reg-

    ulation of multiple neurotransmitters in a single brain region

    has not been reported and this was the focus of our

    investigation.

    The role of nAChRs in prefrontal cortical (PFC) function

    is of considerable interest as this region receives multiple

    neuronal inputs susceptible to modulation by nAChRs. The

    extensive cholinergic projections play an important role in

    0006-8993/$ - see front matterD 2003 Elsevier B.V. All rights reserved.

    doi:10.1016/S0006-8993(03)03532-7

    * Corresponding author. Kalypsys, Inc., 11099 North Torrey Pines

    Road, La Jolla, CA 92037, USA. Tel.: +1-858-754-3300.

    E-mail address: [email protected] (T.S. Rao).1 Arizeke Pharmaceuticals Inc., 6828 Nancy Ridge Dr. Suite 400, San

    Diego, CA 92121, USA.

    www.elsevier.com/locate/brainres

    Brain Research 990 (2003) 203 208

  • 7/27/2019 Agonistas Nicotona x Catecolaminas

    2/6

    higher brain functions, such as cognition [1,6]. Ligand

    binding and functional studies demonstrate nAChR locali-

    zation in the PFC [16,17,34] and modulation of excitatory

    synaptic transmission [14,34]. Studies in which nAChR

    antagonists are directly injected into the rat PFC indicate a

    critical role of nAChR neurotransmission in information

    processing [8]. In addition to cholinergic projections, thePFC also receives DA projections from the ventral tegmen-

    tal area, NE projections from the locus coeruleus and 5-HT

    projections from the raphe nucleus (dorsal and median

    nuclei) [5,33]. These monoaminergic systems appear to play

    an important role in modulation of memory fields [37]. ACh

    release in the PFC and subsequent activation of nAChRs

    can therefore differentially influence the release of a number

    of neurotransmitters. These activities may be of importance

    in understanding the cognitive enhancing activities of nic-

    otine (NIC) and other nAChR agonists in rodents and

    humans [1,22].

    The results have appeared in abstract form [25,30].

    2. Materials and methods

    1-[7,8-3H] Norepinephrine ([3H]-NE, 40 Ci/mmol) was

    purchased from Amersham (Arlington Heights, IL). 3,4-

    [7-3H] dihydroxyphenylethylamine ([[3H]-DA, 20 Ci/mmol)

    and, [3H]-5-hydroxytryptamine (28 Ci/mmol; 5-HT or Se-

    rotonin) were purchased from NEN (Boston, MA). ( )-

    NIC hydrogen tartrate, mecamylamine (MEC) HCl, D-TC,

    1,1-dimethyl-4-phenylpiperzinium iodide (DMPP), desipra-

    mine HCl, cytisine (CYT), tetrodotoxin (TTX) HCl were

    purchased from Sigma (St. Louis, MO). Dihydro-h-eryth-roidine (DHhE) HCl and (F ) epibatidine (EPI) 2 HCl were

    purchased from Research Biochemical (RBI, Natick, MA).

    All other reagents were of the highest purity commercially

    available.

    2.1. Animals

    Male SpragueDawley rats (250300 g) purchased from

    Harlan (San Diego, CA) were used throughout the study.

    The rats were acclimated to the vivarium (temperature: 22

    24 jC, humidity: 5055%, with 12-h lightdark cycle) for

    35 days before use in experiments. All the experiments

    were conducted as per institutionally approved animal care

    guidelines.

    2.2. Neurotransmitter release assays

    Superfusion release assays in the various brain areas

    were conducted as previously described [28,29]. Briefly,

    rats were decapitated and the brain rapidly dissected on ice.

    The PFC slices were cross chopped (300 Am) in a McIlwain

    tissue chopper and equilibrated in Krebs buffer (in mM:

    sodium chloride, 119.5; potassium chloride, 3.3; calcium

    chloride, 1.3; potassium dihydrogen phosphate, 1.2; mag-

    nesium sulfate, 1.2; EDTA, 0.03 and glucose 11.0; pargy-

    line, 0.01) that was continuously gassed with 955% O2/

    CO2 mixture for 10 min. The PFC slices were loaded with

    one of the following tritiated neurotransmitters (60 nM [3H]-

    DA, 50 nM [3H]-NE and 57 nM of ([3H]-5-HT) for 30 min

    at 37 jC in Krebs buffer. For the [3H]-DA release assay,

    PFC slices were pre-incubated for 5 min at 37j

    C in Krebsbuffer containing desipramine (1 AM) prior to the addition

    of [3H]-DA. Inclusion of 5-HT uptake inhibitor during this

    labeling procedure did not affect the uptake of [3H]-DA to

    any significant extent. Therefore, all subsequent experi-

    ments for [3H]-DA release were conducted only in the

    presence of desipramine. Similar experiments suggested

    limited influence of specific DA uptake inhibitors or 5-HT

    inhibitors on [3H]-NE uptake or DA/NE uptake inhibitors

    on [3H]-5HT uptake, respectively. Therefore, PFC slices

    were loaded with [3H]-NE or [3H]-5HT in the absence of

    any uptake inhibitors. At the end of the loading period,

    tissue was rinsed with fresh warm Krebs buffer, transferred

    to chambers and continuously superfused with oxygenated

    buffer for 60 min. Following collection of superfusates to

    establish basal release, slices were exposed to test com-

    pounds for 3-min interval s. For antagonist sensitivi ty

    experiments, either MEC, DHhE or D-TC was added 3

    Fig. 1. Concentration-related effects of NIC, EPI, DMPP and CYT on [3H]-

    DA release (Panel A) or [3H]-NE release (Panel B) from rat PFC slices.

    Data are normalized to NIC (30 AM) and represent meansFS.E.M. (n = 3

    5 experiments each with two to three replicates).

    T.S. Rao et al. / Brain Research 990 (2003) 203208204

  • 7/27/2019 Agonistas Nicotona x Catecolaminas

    3/6

    min prior to the agonist stimulation and included during the

    agonist stimulation. The fractional efflux of tritium was

    estimated as the amount of radioactivity in the superfusate

    fraction relative to the total amount in the tissue, multiplied

    by 100.

    3. Results

    The nAChR agonists tested increased [3H]-DA and [3H]-

    NE release from rat PFC slices in a concentration-dependent

    manner (Fig. 1).

    In the [3H]-DA release assay, EPI was the most

    potent nAChR agonist among the four agonists examined

    (F [3,15] = 12.4, p < 0.001; Fig. 1, panel A). All four

    agonists evoked [3H]-DA release to a similar magnitude.

    Similarly, all four nAChR agonists also increased [3H]-NE

    release from PFC slices in a concentration-related manner

    with a similar magnitude of increase (Fig. 1, panel B). EPIwas the most potent agonist among the four agonists exam-

    ined while NIC was the least potent agonist (F [3,8] = 25.3,

    p < 0.001).

    NIC-induced [3H]-DA or [3H]-NE release was largely

    calcium-dependent (Fig. 2, panels A and D). NIC-evoked

    Fig. 2. Pharmacological characterization of NIC-induced [3H]-DA or [3H]-NE release from rat PFC slices. Data represent meansFS.E.M. (n = 3 4

    experiments with two to three replicates). (A or E) Effect of presence or absence of 2.4 mM calcium in the superfusion buffer (* p < 0.05 vs. calcium present). (B

    or D) Effect of different nAChR antagonists (*p < 0.05 vs. NIC alone). (C or F) Effect of TTX (*p < 0.05 vs. NIC alone).

    T.S. Rao et al. / Brain Research 990 (2003) 203208 205

  • 7/27/2019 Agonistas Nicotona x Catecolaminas

    4/6

    catecholamine release was sensitive to the nAChR antago-

    nists, MEC and DHhE, but not to D-TC (Fig. 2, panels B

    and E). In addition, NIC-evoked catecholamine release was

    sensitive the sodium channel blocker, TTX (1 AM; Fig. 2,

    panels C and F).

    In contrast to their effects on [3H]-NE and [3H]-DA

    release in PFC, the nAChR agonists showed a differential

    profile on [3H]-5-HT release (Fig. 3). DMPP elicited robust

    increases in [3H]-5-HT release. In contrast, EPI did not

    evoke any noticeable increase in [3H]-5-HT release at

    concentrations as high as 100 AM. DMPP-evoked [3H]-

    5HT release was largely insensitive to the nAChR antago-

    nists, MEC, DHhE or D-TC or to the removal of external

    calcium or TTX (data not shown).

    4. Discussion

    The nAChRs are proposed to play an important role in

    modulating synaptic transmission by their effects on neuro-

    transmitter release [38]. The nAChRs have a potentially

    enormous structural and functional diversity due to their

    pentameric structure with multiple genes encoding the alpha

    (a2a9) and beta (h2h4) subunits involved in the

    formation of heteromeric or homooligomeric receptors

    [26,31,39]. Neurotransmitter release studies in rodent brain

    slices or synaptosomes not only provide a useful means of

    defining function of native nAChRs, but also provide some

    insight into the composition of the receptors. If release is

    regulated by different receptors, this should reflect in

    differential agonist rank order of potency and efficacy, as

    well as differential antagonist sensitivity. It appears that

    different nAChRs regulate the release of a given neurotrans-

    mitter in different brain regions [4,23,38]. The results from

    this study demonstrate differential regulation of catechol-

    amine and 5-HT release in the rat PFC by nAChR agonists.

    EPI was the most potent in evoking catecholamine

    release, yet it was least effective at evoking [3H]-5HT

    release. The potency of EPI on PFC catecholamine release

    is consistent with reports that is the most potent nAChR

    agonist in the release of catecholamines from striatal or

    hippocampal preparations [23,2830,38] as well as in in

    vivo assays [24]. The nAChR agonists did not show marked

    potency and efficacy differences in eliciting catecholamine

    release. Puttafarken et al. [23] reported the rank order ofpotency of EPI>CYT < NIC>DMPP in evoking [3H]-DA

    release from PFC. These differences may be related to

    methodological differences such as the inclusion of NE

    uptake inhibitor during the loading of PFC slices with

    [3H]-DA in the present investigation.

    Since both a and h subunits have been shown to

    contribute to agonist pharmacology [16], it is conceivable

    that subtle differences in the heteromeric assembly of

    nAChR subunits, and relative distribution of these recep-

    tors may contribute to differences in agonist rank order of

    potency and/or antagonist sensitivity. In situ hybridization

    studies indicated that DA and NE cell bodies have distinct

    nAChR subunit mRNA distribution (reviewed in Refs.

    [25,39]), and therefore, the respective projections are also

    likely to possess distinct nAChRs. This reflected in re-

    gional differences in nAChR-evoked catecholamine release

    [4,28,38].

    The nAChR pharmacology of 3H]-DA and [3H]-NE

    release in PFC slices provides some evidence as to which

    nAChR subunits may be involved. Thus, ath2-containing

    heteromeric nAChRs, CYT functions as a partial agonist.

    The competitive antagonist, DHhE, is relatively more potent

    at inhibiting heteromeric h2-containing nAChR responses

    relative to those from h4-containing nAChRs [2,9]. In slice

    superfusion assays, DHhE significantly attenuates nAChR-mediated striatal DA release without significant effects on

    nAChR-mediated hippocampal NE release [4,2830]. On

    the other hand, another nAChR antagonist, D-tubocurare (D-

    TC) is more effective at attenuating NIC-induced hippo-

    campal NE release than NIC-induced striatal DA release.

    These data suggest that D-TC may have an opposite nAChR

    subunit selectivity to that of DHhE. The use of highly

    subtype selective antagonists such as alpha-conotoxin AuIB

    and alpha-conotoxin MII further support the notion that

    striatal DA release and hippocampal NE are differentially

    regulated by nAChRs [11,13,17]. The pharmacological

    sensitivity of nicotine-induced [3H]-DA and [3H]-NE re-

    lease in PFC to DHhE, but not to D-TC, suggests the likely

    involvement of h2-containing nAChRs. However, the full

    agonist activity of cytisine in [3H]-DA and [3H]-NE release

    from the rat PFC slices implies that the h2 subunit combi-

    nations in the PFC are different from those in the striatum.

    Similarly, the lack of D-TC sensitivity of [3H]-DA and [3H]-

    NE in PFC suggest that nAChR combinations in the PFC

    are also different from those involved in the hippocampal

    NE release.

    The pharmacology of nAChR agonist-induced [3H]-5-

    HT release in the PFC is distinctly different from [3H]-NE

    or [3H]-DA release in the same region. Surprisingly, EPI

    Fig. 3. Concentration-related effects of NIC, EPI, DMPP and CYT on [3H]-

    5-HT release from rat PFC slices. Data are normalized to DMPP (30 AM)

    and represent meansFS.E.M. (n = 3 experiments with two to three

    replicates in each experiment).

    T.S. Rao et al. / Brain Research 990 (2003) 203208206

  • 7/27/2019 Agonistas Nicotona x Catecolaminas

    5/6

    was the least effective in [3H]-5-HT release with DMPP

    being the most efficacious and most potent of the four

    ligands examined. This profile does not match pharmacol-

    ogy of any known recombinant nAChRs. In addition,

    DMPP-induced [3H]-5-HT release appeared to have atypi-

    cal nAChR pharmacology in that the release was insensitive

    to putative nAChR antagonists and was independent onextracellular calcium. In this respect, nAChR agonist-in-

    duced [3H]-5-HT release from PFC shows considerable

    similarities to that seen the hippocampal slices. Thus,

    Lendvai et al. [15] observed a lack of effect of EPI, NIC

    or CYT on electrically evoked hippocampal [3H]-5-HT

    release while DMPP evoked a robust release. DMPP-

    evoked [3H]-5-HT release in the hippocampus was also

    calcium-insensitive and showed only modest attenuation by

    MEC at concentrations as high as 10 and 20 AM [15].

    Kenny et al. [12] also reported a complex influence of

    nAChRs on hippocampal 5-HT release. Although consid-

    erable evidence exists for presynaptic localization of

    nAChRs on the cortical catecholamine terminals [3], defin-

    itive evidence for the localization of nAChRs on seroto-

    nergic terminals exists only for the striatum [21] and

    hypothalamus [32]. Thus, the destruction of 5-HT neurons

    markedly decreased [3H]-ACh binding to nAChRs in the

    striatum and hypothalamus, without significant reductions

    in the thalamus or cortex [32]. [3H]-5-HT release in this

    context argues that mechanisms other than receptor-medi-

    ated exocytosis are operative.

    In summary, the present investigation demonstrates a

    differential regulation of catecholamine and 5-HT release

    in rat PFC by nAChRs. Pharmacological manipulations that

    either directly activate nAChRs in PFC or indirectly activatethrough increases in ACh are likely to influence DA and NE

    neurotransmission and, to a lesser extent, 5-HT neurotrans-

    mission in the PFC. These activities, combined with in-

    creased glutaminergic neurotransmission observed with

    nAChR activation [34] strongly suggest that nAChRs in

    the PFC subserve the various components of excitatory

    functions such as focus/attention, abstract thinking and

    decision processes [34,37]. The results from this investiga-

    tion implies that nicotinic agonists can differentially affect

    neurotransmitter release in a given brain region and that the

    magnitude of such responses will largely be determined by

    the subtype selectivity of the agonist.

    Acknowledgements

    The authors acknowledge support and critical input from

    Dr. G. Kenneth Lloyd during the course of the investigation.

    References

    [1] B. Bontempi, K.T. Whelan, V.B. Risbrough, T.S. Rao, J.J. Bucca-

    fusco, G.K. Lloyd, F. Menzaghi, SIB-1553A, ((F )-4-[2-(1-methyl-

    2-pyrrolidinyl)ethyl]thiophenol hydrochloride, a subtype selective li-

    gand for nicotinic acetylcholine receptors with putative cognitive-en-

    hancing properties: effects on working and reference memory

    performances in aged rodents and non human primates, J. Pharmacol.

    Exp. Ther. 299 (2001) 297 306.

    [2] L.E. Chavez-Noriega, J.H. Crona, M.S. Washburn, A. Urrutia, K.E.

    Elliott, E.C. Johnson, Pharmacological characterization of recombi-

    nant human neuronal nicotinic acetylcholine receptors ha2h2, ha2h4,

    ha3h2, ha4h2, ha4h4 and ha7 expressed in xenopus oocytes, J.

    Pharmacol. Exp. Ther. 280 (1997) 346356.

    [3] P.B.S. Clarke, A. Pert, Autoradiographic evidence for nicotine recep-

    tors on nigrostriatal and mesolimbic dopaminergic neurons, Brain

    Res. 348 (1985) 355358.

    [4] P.B.S. Clarke, M. Reuben, Release of [3H]-noradrenaline from rat

    hippocampal synaptosomes by NIC: mediation by different nicotinic

    receptor subtypes from striatal [3H]-dopamine release, Br. J. Pharma-

    col. 117 (1996) 595606.

    [5] J.R. Cooper, F.E. Bloom, R.H. Roth (Eds.), The Biochemical Basis of

    Neuropharmcology, Oxford Univ. Press, New York, NY, 1996.

    [6] S.B. Dunnett, B.J. Everitt, T.W. Robins, The basal forebrain cortical

    cholinergic system: interpreting the functional consequences of exci-

    totoxic lesions, Trends Neurosci. 14 (1991) 494500.

    [7] S. Grady, M.J. Marks, S. Wonnacott, A.C. Collins, Characterization

    of nicotine-receptor mediated [3H]-dopamine release from synapto-

    somes prepared from mouse striatum, J. Neurochem. 59 (1992)

    848856.

    [8] S. Grannon, B. Poucet, C. Thomas-Blanc, J.-P. Changeux, C. Vidal,

    Nicotinic and muscarinic receptors in the rat prefrontal cortex: differ-

    ential roles in working memory, response selection and effortful pro-

    cessing, Psychopharmacology 119 (1995) 139 144.

    [9] S.C. Harvey, C.W. Leutje, Determinants of competitive antagonist

    effects of nicotine on neuronal nicotinic receptorh subunits, J. Neuro-

    sci. 16 (1996) 37983806.

    [10] F. Hery, S. Bourgoin, N. Hamon, J.P. Ternaux, J. Glowinski, Control

    of the release of newly synthesized [3H]-5-hydroxytryptamine by

    nicotinic and muscarinic receptors in the rat hypothalamic slices,

    Arch. Int. Pharmacodyn. Ther. 296 (1997) 9197.

    [11] S.A. Kaiserr, L. Soliakov, S.C. Harvey, C.W. Leutje, S. Wonnacott,

    Differential inhibition by alpha-conotoxin-MII of the nicotinic stim-ulation of [3H]-dopamine release from rat striatal synaptosomes and

    slices, J. Neurochem. 70 (1998) 1069 1076.

    [12] P.J. Kenny, S.J. File, M.J. Neal, Evidence for a complex influence of

    nicotinic acetylcholine receptors on hippocampal serotonin release,

    J. Neurochem. 75 (2000) 24092414.

    [13] J.M. Kulak, T.A. Nguyen, B.M. Olivera, J.M. McIntosh, a-Conotoxin

    MII blocks nicotine-stimulated dopamine release in rat striatal synap-

    tosomes, J. Neurosci. 17 (1997) 5263 5270.

    [14] E.K. Lambe, M.R. Picciotto, G.K. Aghajanian, Nicotine induces glu-

    tamate release from thalamocortical terminals in prefrontal cortex,

    Neuropsychopharmacology 28 (2003) 216 225.

    [15] B. Lendvai, H. Sershan, A. Lajtha, E. Santha, M. Barnyi, E.S. Vizi,

    Differential mechanisms involved in the effect of nicotinic agonists

    DMPP and lobeline to release [3H]-5-HT from rat hippocampal slices,

    Neuropharmacology 35 (1996) 1769 1777.[16] C.W. Luetje, J. Patrick, Both a- and h-subunits contribute to the

    agonist sensitivity of neuronal nicotinic acetylcholine receptors,

    J. Neurosci. 11 (1991) 837845.

    [17] S. Luo, J.M. Kulak, G.D. Cartier, R.B. Jacobsen, D. Yoshikami, B.M.

    Olivera, J.M. McIntosh, a-Conotoxin AuIB selectively blocks a3h4

    nicotinic acetylcholine receptors and nicotine-evoked norepinephrine

    release, J. Neurosci. 18 (1998) 85718579.

    [18] M. Marks, J.A. Stitzel, E. Romm, J.M. Wehner, A.C. Collins, Nic-

    otinic binding sites in rat and mouse brain: comparison of acetylcho-

    line, nicotine and a-bungarotoxin, J. Pharmacol. Exp. Ther. 30 (1986)

    427436.

    [19] D. Marshall, L. Soliakov, P. Redfern, S. Wonnacott, Tetrodotoxin-

    sensitivity of nicotine-evoked dopamine release from rat striatum,

    Neuropharmacology 35 (1996) 1531 1536.

    T.S. Rao et al. / Brain Research 990 (2003) 203208 207

  • 7/27/2019 Agonistas Nicotona x Catecolaminas

    6/6

    [20] S.N. Mitchell, M.P. Brazell, M. Joseph, M.S. Alavijeh, J.A. Gray,

    Regionally specific effects of acute and chronic nicotine on rates of

    catecholamine and 5-hydroxytryptamine synthesis in rat brain, Eur. J.

    Pharmacol. 167 (1989) 311312.

    [21] S.V. Nayak, P. Ronde, A.D. Spier, S.C. Lummis, R.A. Nichols, Nic-

    otinic receptors co-localize with 5-HT3 serotonin receptors on striatal

    nerve terminals, Neuropharmacology 39 (2000) 2681 2690.

    [22] P. Newhouse, A. Potter, E.D. Levin, Nicotinic system involvement in

    Alzheimers disease and Parkinsons disease: implications for thera-

    peutics, Drugs Aging 11 (1997) 206 288.

    [23] P.S. Puttafarken, I. Jacobs, I.C.R. Falynek, Characterization of

    nicotinic acetylcholine receptor-mediated [3H]-dopamine release

    from rat cortex and striatum, Neuropharmacology 39 (2000)

    26732680.

    [24] T.S. Rao, L.D. Correa, R.T. Reid, G.K. Lloyd, Evaluation of anti-

    nociceptive effects of neuronal nicotinic acetylcholine receptor

    (nAChR) ligands in the rat tail-flick assay, Neuropharmacology 35

    (1996) 393 405.

    [25] T.S. Rao, R.T. Reid, A.I. Saccan, G.K. Lloyd, Pharmacological char-

    acterization of neuronal nicotinic acetylcholine receptor-mediated

    neurotransmitter release in vitro and in vivo, Proc. Winter Conf. Brain

    Res., 1998, Snow Bird, UT, #73.

    [26] L. Role, D.K. Berg, Nicotinic receptors in the development and mod-

    ulation of CNS synapses, Neuron 16 (1996) 10771085.

    [27] P. Rowell, L.A. Carr, A.C. Garner, Stimulation of [3H]-DA release

    by nicotine in rat nucleus accumbens, J. Neurochem. 49 (1987)

    14491454.

    [28] A.I. Sacaan, J.L. Dunlop, G.K. Lloyd, Pharmacological characteriza-

    tion of neuronal acetylcholine-gated ion channel receptor-mediated

    hippocampal norepinephrine and striatal dopamine release from rat

    brain slices, J. Pharmacol. Exp. Ther. 274 (1995) 224 230.

    [29] A.I. Sacaan, F.M. Menzaghi, J.L. Dunlop, L.D. Correa, K.T. Whelan,

    G.K. Lloyd, Epibatidine: a nicotinic acetylcholine-receptor agonist

    releases monoaminergic neurotransmitters. In vitro and in vivo evi-

    dence in rats, J. Pharmacol. Exp. Ther. 276 (1996) 509515.

    [30] A.I. Sacaan, E.M. Santori, M. Keegan, G.K. Lloyd, T.S. Rao, Dop-

    amine and norepinephrine release from rat prefrontal cortex is regu-

    lated by different nicotinic acetylcholine receptor subtypes, Soc.

    Neurosci. Abs. 23 (1997) 477.

    [31] P.B. Sargent, The diversity of neuronal nicotinic acetylcholine recep-

    tors, Annu. Rev. Neurosci. 16 (1993) 403443.

    [32] R.D. Schwartz, J. Lehmann, K.J. Kellar, Presynaptic nicotinic chol-

    inergic receptors labeled by [3H]-acetylcholine on catecholamine and

    serotonin axons in brain, J. Neurochem. 42 (1984) 14951498.

    [33] U. Ungerstedt, Stereotaxic mapping of the monoamine pathways in

    the rat brain, Acta Physiol. Scand., Suppl. 367 (1971) 120.

    [34] C. Vidal, J.-P. Changeux, Nicotinic and muscarinic modulations of

    excitatory synaptic transmission in the rat prefrontal cortex in vitro,

    Neuroscience 56 (1993) 23 32.

    [35] E.S. Vizi, H. Sershan, H. Balla, A. Mike, K. Windish, Z. Juranyi, A.

    Lajtha, Neurochemical evidence of heterogeneity of presynaptic and

    somatodendritic nAChRs, Ann. N.Y. Acad. Sci. 757 (1995) 8499.

    [36] T.C. Westfall, H. Grant, H. Perry, Release of dopamine and 5-hydroxy-

    tryptamine from rat striatal slices following activation of nicotinic

    cholinergic receptor, Gen. Pharmacol. 14 (1983) 321 325.

    [37] G.V. Williams, P. Goldman-Rakic, Modulation of memory fields by

    dopamine D1 receptors in prefrontal cortex, Nature 376 (1995)

    572575.

    [38] S. Wonnacott, Presynaptic nicotinic ACh receptors, Trends Neurosci.

    20 (1997) 9298.

    [39] M. Zoli, C. Lena, M.R. Picciotto, J.-P. Changeux, Identification of

    four classes of brain nicotinic receptors using h2 mutant mice,

    J. Neurosci. 18 (1988) 4461 4472.

    T.S. Rao et al. / Brain Research 990 (2003) 203208208