REVIEW ARTICLE
Muscle and neuronal nicotinic acetylcholine receptors
Structure, function and pathogenicity
Dimitra Kalamida1, Konstantinos Poulas1,2, Vassiliki Avramopoulou2, Efrosini Fostieri2,
George Lagoumintzis1, Konstantinos Lazaridis2, Anastasia Sideri2, Marios Zouridakis1,2 and
Socrates J. Tzartos1,2
1 Department of Pharmacy, University of Patras, Greece
2 Department of Biochemistry, Hellenic Pasteur Institute, Athens, Greece
Keywords
acetylcholine receptor; ACh binding protein;
extracellular domain; ligand-gated ion
channels; Myasthenia gravis; neuromuscular
junction; nicotinic receptors; protein
function; protein structure; transmembrane
proteins
Correspondence
S. J. Tzartos, Department of Pharmacy,
University of Patras, GR26500, Rio Patras,
Greece
Tel: +30 261 096 9955
E-mail: tzartos@upatras.gr
or
Department of Biochemistry, Hellenic
Pasteur Institute, GR11521 Athens, Greece
Fax: +30 210 647 8842
Tel: +30 210 647 8844
E-mail: tzartos@pasteur.gr
(Received 19 January 2007, revised 30 April
2007, accepted 11 June 2007)
doi:10.1111/j.1742-4658.2007.05935.x
Nicotinic acetylcholine receptors (nAChRs) are integral membrane proteins
and prototypic members of the ligand-gated ion-channel superfamily, which
has precursors in the prokaryotic world. They are formed by the assembly
of five transmembrane subunits, selected from a pool of 17 homologous
polypeptides (a1–10, b1–4, c, d, and e). There are many nAChR subtypes,
each consisting of a specific combination of subunits, which mediate
diverse physiological functions. They are widely expressed in the central
nervous system, while, in the periphery, they mediate synaptic transmission
at the neuromuscular junction and ganglia. nAChRs are also found in
non-neuronal ⁄ nonmuscle cells (keratinocytes, epithelia, macrophages, etc.).
Extensive research has determined the specific function of several nAChR
subtypes. nAChRs are now important therapeutic targets for various diseases, including myasthenia gravis, Alzheimer’s and Parkinson’s diseases,
and schizophrenia, as well as for the cessation of smoking. However,
knowledge is still incomplete, largely because of a lack of high-resolution
X-ray structures for these molecules. Nevertheless, electron microscopy
studies on 2D crystals of nAChR from fish electric organs and the determination of the high-resolution X-ray structure of the acetylcholine binding
protein (AChBP) from snails, a homolog of the extracellular domain of the
nAChR, have been major steps forward and the data obtained have
important implications for the design of subtype-specific drugs. Here, we
review some of the latest advances in our understanding of nAChRs and
their involvement in physiology and pathology.
Abbreviations
A-AChBP, Aplysia californica AChBP; AAN, autoimmune autonomic neuropathy; ACh, acetylcholine; AChBP, acetylcholine-binding protein;
AChR, acetylcholine receptor; AD, Alzheimer’s disease; ADHD, attention deficit hyperactivity disorder; B-AChBP, Bulinus truncatus AChBP;
a-Bgtx, a-bungarotoxin; a-Cbtx, a-cobratoxin; CICR, calcium-induced calcium release; CMS, congenital myasthenic syndrome; CNS, central
nervous system; a-Ctx, a-conotoxin; DA, dopamine; EAAN, experimental autoimmune autonomic neuropathy; EAMG, experimental
autoimmune Myasthenia gravis; ECD, extracellular domain; ER, endoplasmic reticulum; ERK ⁄ MAPK, extracellular signal-regulated
mitogen-activated protein kinase; GST, glutathione S-transferase; 5-HT, 5-hydroxytryptamine; IFN-c, interferon-c; IL, interleukin; L-AChBP,
Lymnaea stagnalis AChBP; LBD, ligand-binding domain; LGIC, ligand-gated ion channels; M1–4, transmembrane fragments 1–4; mAb,
monoclonal antibody; mAChR, muscarinic AChR; MAPK, mitogen-activated protein kinase; MG, myasthenia gravis; MIR, main immunogenic
region; MuSK, muscle-specific receptor tyrosine kinase; nAChR, nicotinic acetylcholine receptor; NF-jB, nuclear factor-jB; NMDA,
N-methyl-D-aspartate; NMJ, neuromuscular junction; PD, Parkinson’s disease; PNS, peripheral nervous system; TNF, tumor necrosis factor;
VOCCs, voltage-operated Ca2+ channels; VTA, ventral tegmental area.
FEBS Journal 274 (2007) 3799–3845 ª 2007 The Authors Journal compilation ª 2007 FEBS
3799
Nicotinic acetylcholine receptors
D. Kalamida et al.
Ion channels
The cell membrane is the main barrier for ion movement, so specific proteins, such as ion channels, have
evolved to transport ions across it. Ion channels are
gated pores that permit the passive flow of ions down
their electrochemical gradients. Because of their
important functional roles, their membrane location
and structural heterogeneity and the restricted tissue
expression of some channel types, ion channels are
ideal drug targets. They share a common structural
pattern: the central water-filled pore, through which
the ions move, is usually formed by four or five transmembrane a helices (tetrameric or pentameric channels,
respectively), arranged like the staves of a barrel.
These different pore-forming helices can be parts of
different subunits.
Access to the pore is governed by a ‘gate’, which
can be opened or closed by electrical, chemical or
mechanical signals, depending on the type of channel.
Ligand-gated ion channels (LGICs) form a group of
ion channels in which ‘gate’ opening is controlled by
the binding of a chemical messenger. They share several common physiological and structural features and,
because of their oligomeric structure, have a number
of ligand-binding sites. The ligand-binding sites and
the ‘gate’ lie at a considerable distance from each
other, the ligand-binding sites quite commonly being
located at the interface between two adjacent subunits,
which is energetically more favorable given that
domain interfaces undergo larger conformational changes upon ligand binding.
Nicotinic acetylcholine receptors
Acetylcholine receptors (AChRs) are integral membrane proteins that respond to the binding of acetylcholine (ACh), which is synthesized, stored and finally
released by cholinergic neurons. Like other transmembrane receptors, AChRs have been classified according
to either their pharmacological properties or their relative affinities for various molecules, and can therefore
be further divided into: (a) nicotinic AChRs (nAChRs,
the ‘ionotropic’ AChRs), which are particularly
responsive to nicotine [1,2]. nAChRs are the prototype
members of the LGICs superfamily; and (b) muscarinic AChRs (mAChRs, the ‘metabotropic’ AChRs),
which are particularly responsive to muscarine [3,4].
mAChRs are members of the membrane-bound G-protein-coupled receptor superfamily.
In the N-terminal extracellular domain (ECD) of
each LGIC subunit there is a conserved sequence of 13
residues flanked by covalently bonded cysteines, form3800
ing a loop located between the ligand-binding domain
(LBD) and ion-channel domain. Because of this unique
common characteristic feature, LGICs are also called
the Cys-loop receptor superfamily. nAChRs are composed of five homologous subunits organized around a
central pore and are further divided into two groups:
(a) the muscle type, found in vertebrate skeletal muscles, where they mediate neuromuscular transmission
at the neuromuscular junction (NMJ), as well as in fish
electric organs; and (b) the neuronal type, found
mainly throughout the peripheral nervous system
(PNS) and central nervous system (CNS), but also in
non-neuronal tissues.
cDNAs for 17 types of nAChR subunits have been
cloned from several species. These consist of a subunits
(a1–10), which carry the main parts of the ligand-binding sites, b (b1–4), c, d, and e subunits. Based on their
different ligand-binding properties, the diverse group
of the nAChRs has been divided into two main classes:
(a) the a-bungarotoxin (a-Bgtx)-binding nAChRs,
which can be either homopentamers of a7, a8 or a9
subunits or heteropentamers (e.g. a2be(c)d); and
(b) nAChRs which do not bind a-Bgtx, contain the
a2–a6 and b2–b4 subunits, exist only as heteropentamers and bind agonists with high affinity [2].
Most of the early studies on nAChR subunit composition and structure were performed on molecules
isolated from the electric organs of the electric fish,
Torpedo and Electrophorus, a tissue extremely rich in
nAChRs (many milligrams of nAChR per kilogram
of Torpedo electric organ). Torpedo nAChR, which
shows a high degree of homology with vertebrate
skeletal muscle nAChR [5], has been studied extensively; it has two ACh-binding sites located at interfaces between subunits, a transmembrane ion
channel, and all the structural elements required for
activation and desensitization processes [6,7]. Using
recombinant DNA techniques, close homologies
between nAChR subunit sequences from fish electric
organs and skeletal muscle from higher vertebrates,
including humans, have been revealed [8]. Therefore, all these nAChRs are often named muscle-type
nAChRs.
Early in the study of the nAChR, investigators
attempted to characterize the receptor using a number
of ligands, but the great advance occurred when small
polypeptides from the venom of poisonous snakes were
tested and found to block neuromuscular transmission
in vertebrates. a-Bgtx from Bungarus multicinctus
venom was historically the first toxin used to characterize nAChRs and the demonstration that it causes
almost irreversible block of Torpedo [9] and Electrophorus [10] nAChRs was a great step forward, as it
FEBS Journal 274 (2007) 3799–3845 ª 2007 The Authors Journal compilation ª 2007 FEBS
Nicotinic acetylcholine receptors
D. Kalamida et al.
made possible the isolation and characterization of the
molecule.
Extensive studies on nAChRs from various species
have demonstrated that each nAChR subunit consists
of: (a) the N-terminal ECD of each subunit which is
210–220 amino acids long and bears the LBD for
agonists and competitive antagonists [11,12]; (b) four
small (15–20 amino acids long) hydrophobic transmembrane segments (M1–M4) and two small hydrophilic loops, linking segments M1–M2 and M2–M3;
(c) a larger loop varying in size (100–150 residues)
and sequence between subunits, which lies between
M3 and M4, and bears phosphorylation sites [13]; and
(d) the C-terminal end of each subunit which bears a
small (4–28 amino acids) hydrophilic extracellular segment.
Several post-translational modifications contribute
to the structure and function of the molecule. An unusual disulfide bond, common to all a subunits, is
formed between two adjacent Cys residues that correspond to amino acids 192 and 193 of the Torpedo
a1 subunit. This particular bond contributes to the
agonist-binding site. Glycosylation sites, the number
and location of which differ between subunits, are
located in the ECD. As mentioned above, phosphorylation sites have also been identified in the intracellular
domain between the M3 and M4 transmembrane segments.
A breakthrough in our understanding of the structure and function of nAChRs came from solving the
crystal structure of the acetylcholine-binding protein
(AChBP) [14,15]. This protein is a naturally occurring
homolog of the ECD of nAChR which can be produced in large quantities and its water solubility facilitates crystallization and X-ray analysis. Subsequent
solution of the X-ray structures of the AChBP complexes with agonists and antagonists [16–18] provided
excellent models for the LBDs of LGICs and their
complexes with putative drugs through homology
modeling. Recently, prokaryotic homologs to LGICs
have been shown to exist [19] which can also be
obtained in large amounts. The cloning and expression
of the Gloeobacter violaceus channel was achieved and
homology modeling in parallel with electrophysiological experiments revealed key residues for its
function [20].
nAChR 3D structure
Structural information on nAChRs has been derived
from electron microscopy studies on 2D arrays of the
Torpedo nAChR [7,21,22]. These earlier studies
revealed the dimensions and shape of the molecule,
defined the arrangement of the subunits and the
boundaries between them and provided an insight into
the location of the binding sites and the organization
of the ion channel. Subsequent 4.6 and 4 Å resolution
electron microscopy studies provided an insight into
the structure of the extracellular and transmembrane
subunit domains [23,24]. Extracellularly, the AChbinding pockets were shown to be surrounded by
seven-stranded b sheets forming a larger b-sandwich
structure. These data confirmed CD studies [25–30],
which indicated that the LBD consists predominantly
of b sheets. It was clearly shown that the four membrane-spanning segments of each subunit (M1–M4) are
a helices [24].
Detailed information on the atomic structure of an
ACh-binding domain first became available following
elucidation of the crystal structure at 2.7 Å resolution
of the AChBP from the glial cells of the mollusc,
Lymnaea stagnalis [14] (Fig. 1). This protein, a soluble
homopentamer of a 210 amino acid subunit, is a structural and functional homolog of the ECD of Cys loop
receptor subunits, showing up to 24% sequence identity with human nAChR ECDs and 15–18% with
ECDs of other LGICs, and is used as a model for the
LBD of nAChRs. The resolved dimensions of the
AChBP (a cylinder, 62 Å high, with a diameter of
80 Å and a central hole 18 Å in diameter) are in good
agreement with those estimated by electron microscopy
for the Torpedo nAChR [23]. When viewed along the
fivefold axis, the AChBP homopentamer resembles a
toy windmill with blade-like protomers (Fig. 1A). Each
protomer consists of an a helix close to the N-terminal,
two short 310 helices and 10 b strands (b1–b10), connected through equal number of loops (Fig. 1B). The
b strands are arranged in two sets joined through a
cysteine disulfide bridge, the Cys loop, forming a
b-sandwich hydrophobic core.
To date, no 3D X-ray structure of any nAChR or
any other LGIC is available. Although very small,
nondiffracting 3D crystals of Torpedo nAChR were
generated as early as in 1988 [31,32], several attempts
to obtain high-quality crystals of this large membrane
protein or even of its smaller (mainly extracellular)
fractions have met with little success.
Based on the crystal structure of AChBP, the atomic
model of Torpedo muscle-type nAChR at 4 Å resolution electron microscopy studies (Fig. 2), allowed a
detailed description of the whole receptor in its closedchannel form at a chemical level [33]. More recently,
crystal structures of various molluscan AChBPs with
known cholinergic ligands and toxins have provided
detailed information on the conformational changes
induced upon ligand binding.
FEBS Journal 274 (2007) 3799–3845 ª 2007 The Authors Journal compilation ª 2007 FEBS
3801
Nicotinic acetylcholine receptors
D. Kalamida et al.
Fig. 1. Crystal structure of Lymnaea stagnalis AChBP, which is homologous to nAChR ECD at 2.7 Å resolution. (A) Top view. Each subunit
of the homopentamer is shown in a different color and symbolized with a different letter (A–E). The five ligand-binding sites between the
subunits are shown in ball-and-stick representation. (B) Side view of AChBP protomer from outside the pentameric ring. The side of the protomer, bearing the conserved Cys loop is called the principal (plus) side. Also shown, is the AChBP region corresponding to the MIR epitope
of the a subunit of the muscle nAChR. Reproduced from Brejc et al. [14], with permission.
Fig. 2. Electron microscopy structure
of Torpedo nAChR at 4 Å resolution.
(A) Ribbon diagrams of the whole nAChR
from Torpedo electric organ, as viewed from
the synaptic cleft. Also shown are the
locations of aTrp149 (gold) and the MIR
epitope. (B) Side view of the a subunit. The
location of the functionally important A, B,
C, b1–b2 and Cys loops is shown. Also,
the MIR epitope and the membrane
(E, extracellular; I, intracellular) are
presented. Reproduced from Unwin [33],
with permission.
nAChR overview
In the 4 Å model, the Torpedo nAChR has a total
length of 160 Å normal to the membrane plane and
3802
is divided into three domains: an N-terminal extracellular LBD, a membrane-spanning pore and an intracellular domain (Fig. 2B). The receptor subunits in the
LBD are each organized around a curled b-sandwich
FEBS Journal 274 (2007) 3799–3845 ª 2007 The Authors Journal compilation ª 2007 FEBS
Nicotinic acetylcholine receptors
D. Kalamida et al.
hydrophobic core, consisting of 10 b strands which are
joined through the Cys loop and contain one N-terminal a helix, like the protomers of the closely related
AChBP [14]. The hydrophobic core of this receptor
domain consists of conserved residues, equivalent to
those of the AChBP, with the exception of the aLeu6
near the end of the a helix. This domain also contains
several loop regions, i.e. loops A, B, C, Cys loop and
b1–b2 loop, which are critical for receptor function,
identified as components of the LBD (Fig. 2B).
Early biochemical studies involving site-directed
mutagenesis and affinity labeling indicated that two
separate parts of the nAChR-LBD are involved in the
formation of the agonist ⁄ competitive antagonist-binding site [34–36]. One is called the ‘principal component’
of the binding site and resides on the a subunit,
whereas the other is called the ‘complementary compo-
nent’ and resides on the adjacent non-a subunit. The
ACh-binding pocket of the nAChR is formed between
loops A, B and C on the a subunit and strands b5 and
b6 of the b-sandwich core of the adjacent c or d subunit and lies 40 Å above the membrane surface and
on opposite sides of the channel pore [33]. The key residues of the loops implicated in the formation of the
ACh binding site are Tyr190, Cys192 and Tyr198 of
the C loop, which is incorporated in the b9–b10 hairpin and Trp149 of the B loop (Fig. 3A).
a1-ECDs also contain the main immunogenic region
(MIR) [37–40] (Fig. 2), a region of overlapping epitopes (including amino acids 67–76 of the a1 subunit),
against which a large fraction of autoantibodies against
nAChR is directed in the autoimmune disease myasthenia gravis (MG). The critical segment of the MIR that
serves as the epitope of autoantibodies is localized to
Fig. 3. (A) ACh-binding region of the closed channel of Torpedo nAChR at the interface between a and c subunits. Loops B and C of the a
subunit and the adjacent b5 and b6 strands of c subunit are shown. (B) (a) simplified Ca traces of the ligand-binding region of the a subunit
in the closed channel; (b) equivalent region of L-AChBP complexed with carbamylcholine [16]; (c) superimposition of the two regions, revealing the closure of B and C loops around the bound agonist. Reproduced from Unwin [33], with permission.
FEBS Journal 274 (2007) 3799–3845 ª 2007 The Authors Journal compilation ª 2007 FEBS
3803
Nicotinic acetylcholine receptors
D. Kalamida et al.
residues aTrp67–aAsp71, with aAsn68 and aAsp71
being the most important. All five residues appear to
be exposed to the solvent, because of the wide separation between the loop they form (part of the b2–b3
loop) and the N-terminal a helix of the LBD. The
equivalent loops of the other subunits (non-a) seem to
have the same fold, but these are closer to the respective helices and this results in the partial burial of the
residues aligning with aTrp67–aAsp71. There are considerable differences in the amino acid sequence of the
67–76 segment between a1 and the neuronal a subunits.
Smaller differences occur between a1 and a3 ⁄ a5 and
this is in agreement with observations that some antiMIR mAbs bind, in addition to a1, to a3 and a5 but
not to other neuronal a subunits [41,42] (S. J. Tzartos
et al., unpublished data). Although the corresponding
region in the AChBP (residues 65–72) has no sequence
or functional similarity to the MIR on the a1 subunit,
the crystal structure of the AChBP shows that this is
located in a highly accessible area at the ‘top’ of the
pentamer (Fig. 1B) [14], as would normally be expected
for a very antigenic region.
In the membrane-spanning domain, the a helices of
each subunit (M1–M4) are arranged symmetrically,
forming the channel pore [24]. The five M2 helical segments of all subunits line the pore, forming an inner
ring, whereas the other 15 helical segments in M1, M3
and M4 from all five subunits coil around each other,
forming an outer ring, which shields the inner ring of
M2 domains from the membrane lipids. In the closed
channel, the M2 helices come together near the middle
of the membrane, forming a hydrophobic girdle, which
is considered to be the gate of the channel, functioning
as an energetic barrier to ion permeation [33]. Apart
from the a-helical regions M1–M4, the small loops
M1–M2 (intracellular) and M2–M3 (extracellular) are
also considered to be part of the membrane-spanning
region (Fig. 2B). These loops are functionally important, as the transmembrane domain interacts through
the M2–M3 loop with the b1–b2 loop and the Cys
loop of the ECD. Moreover, the membrane-spanning
domain is joined covalently to the LBD through an
extension of the b10 strand into M1 helix.
The intracellular region consists of the M3–M4 loop,
which contains a curved a helix (MA) immediately
before the M4 transmembrane region (Fig. 2B). Most
of the loop immediately after the M3 transmembrane
region (the M3–MA loop) seems to be rather disordered and it is not resolved in the electron microscopy
structure. Each subunit contributes one MA a helix, to
shape the wall of the vestibule. These five helices form
an inverted pentagonal cone which has five intervening
spaces (‘windows’) of a width < 8 Å (thus comparable
3804
with the diameter of a hydrated sodium or a potassium
ion), surrounded by negatively charged side chains.
These windows represent obligatory ion pathways,
because no alternative routes exist for transport
inwards or outwards the intracellular vestibule. Therefore, they constitute a charge and size ‘selectivity filter’,
which facilitates cation transport, but prevents anions
and large ions from going through. Both the extracellular and intracellular vestibules of the channel are
lined mainly by negatively charged side-chain groups,
which form an electrostatic environment that stabilizes
and thus favors the influx of cations (Na+, K+ and
Ca2+) [33].
In the LBD, the Torpedo subunits were shown to
interact mainly through polar side chains [43], similar
to the interactions seen in the AChBP. Charged side
chains on both a1 subunits form ion pairs with side
chains on neighboring subunits. The subunit–subunit
interactions of the membrane-spanning domain are
mainly attributed to hydrophobic side chains projecting from the helices M1, M2 and M3, implicating relatively few residues on M1 and M3. Intracellularly, the
subunit–subunit interactions involve contacts between
the M1–M2 loops as well as between the M3–M4 and
MA regions of neighboring subunits. Because of the
incomplete resolution of the electron microscopy structure in this region, no detail of these interactions is
available.
Atomic structure of the ligand-binding site
In the crystal structure of AChBP [14] each ligandbinding site is found in a cavity at each interface
between the five subunits, lined by aromatic and
hydrophobic residues, previously shown to be involved
in ligand-binding in nAChRs by mutational analyses
and site-directed labeling [11,36,44–46]. The ligandbinding site is formed by the contribution of highly
conserved, through the LGIC family, residues from
loops A (Tyr89), B (Trp143, Trp145) and C (Tyr185,
the double cysteine 187–188 and Tyr192) of the principal (plus) side of one subunit and by less conserved
residues from loops D (Trp53, Gln55), E (Arg104,
Val116, Leu112 and Met114) and F (Tyr164) of the
complementary (minus) side of the adjacent subunit
(Fig. 4A). When the protomer is viewed by the side
and perpendicular to the fivefold axis, the plus side of
each protomer is the one that bears the Cys loop
(Fig. 1B).
The structure of AChBP also revealed the interactions between residues of each protomer in the dimer
interface. The subunit interface consists entirely of
loop regions on the plus side and mainly of secondary
FEBS Journal 274 (2007) 3799–3845 ª 2007 The Authors Journal compilation ª 2007 FEBS
Nicotinic acetylcholine receptors
D. Kalamida et al.
K143
Fig. 4. Representation of the ligand-binding site and of the interactions upon agonist binding in (A) Lymnaea stagnalis AChBP (Hepes bound)
[14], (B) Lymnaea stagnalis AChBP (nicotine bound) [16], (C) Lymnaea stagnalis AChBP (carbamylcholine bound) [16], (D) Aplysia californica
AChBP (lobeline bound) [55], and (E) Aplysia californica AChBP (epibatidine bound) [55]. Reproduced from Brejc et al. [14], Celie et al. [16]
and Hansen et al. [55], with permission.
FEBS Journal 274 (2007) 3799–3845 ª 2007 The Authors Journal compilation ª 2007 FEBS
3805
Nicotinic acetylcholine receptors
D. Kalamida et al.
structure elements (a1, b1–3, b5–6 and L9) on the
minus side. Its buried nature leads to a preference of
hydrophobic residues, including only a bifurcated salt
bridge formed between Glu149 on the plus side of one
subunit and Arg3 and Arg104 on the minus side of the
adjacent subunit. In contrast to the well-conserved residues stabilizing the protomer structure by the formation of hydrophobic cores, the interface residues
important for the pentameric formation and some for
ligand binding are not well conserved between the
members of the LGICs, thus creating specificity for
different ligands. The only highly conserved interface
residues are those forming the plus side of the ligandbinding site.
Differences in the subunit interfaces of AChBPs from
different species, lead to distinct ligand-binding properties. This is more stressed by the comparison of the subsequent crystal structure of Bulinus truncatus AChBP
(B-AChBP) [47] with that of Lymnaea stagnalis AChBP
(L-AChBP). Although their structures were found to be
very similar, the residues of the subunit interface have
not been conserved in B-AChBP except for the ligandbinding residues. These differences in subunit interface
contacts between B-AChBP and L-AChBP have a
major effect on the stability and ligand-binding properties of these proteins. L-AChBP is more stable than
B-AChBP as shown by thermal stability studies using
CD [47], probably because L-AChBP has on average
one extra hydrogen bond and two additional salt bridges in each interface. Apparently, the mode of pentamer
formation is poorly conserved. Analyzing the AChBP
structures for interface regions that could improve the
formation of stable pentamers in nicotinic structures
can prove beneficial, because expression of properly
assembled nAChR LBDs has proven remarkably difficult, and in many cases a mixture of monomers, dimers,
and multimers was obtained [26,48–50]. In addition,
B-AChBP has a 5–10-fold higher affinity for the
binding of cholinergic ligands [nicotine, acetylcholine,
carbamylcholine, and (+)-tubocurarine], compared
with L-AChBP, but interestingly does not bind significantly to a-Bgtx [47]. The differences in ligand-binding
were attributed to three nonconserved residues on the
complementary side of the ligand-binding of the two
AChBPs (L-AChBP Arg104, Leu112 and Met114 changed to B-AChBP Val103, Ile111 and Val113, respectively)
and probably to alternative residues which may be part
of the subunit–subunit interfaces. Mutation of these
three L-AChBP residues to the B-AChBP corresponding ones led to a triple mutant L-AChBP, which showed
a clear gain-of-affinity for binding both nicotine and
(+)-tubocurarine, equivalent to that of the B-AChBP
[47]. However, the remaining differences in ACh binding
3806
and in entropic and enthalpic contribution showed that
regions outside the binding site (and probably in the
interface) also contribute to the affinity [47]. Interestingly, B-AChBP contains a lysine residue at position
183 (adjacent to the conserved Tyr184 of loop C),
which seems to be responsible for abolishing a-Bgtx
binding, similar to the human a2, a3 and a4 subunits
that do not bind the toxin. NMR and mutagenesis studies have shown that nAChRs that bind to a-Bgtx have
a Tyr or Phe residue at this position [51–53] and
replacement of this Tyr by Lys completely prevents
a-Bgtx binding [53,54].
Although the initial crystal structure of L-AChBP
was obtained in the absence of any cholinergic ligand,
the Hepes molecule used in the crystallization buffer,
mimics known cholinergic ligands, as it contains a positively charged quaternary ammonium group. Indeed,
this molecule was detected well-placed in each ligandbinding site of L-AChBP, stacked on the highly conserved Trp143 by cation-p interations, as expected for
nicotinic agonists. This revealed for the first time the
critical role of Trp143 in ligand binding.
More recently, cocrystallization of various molluscan
AChBPs with several agonists and antagonists
[16–18,55,56] revealed details of the atomic interactions
between ligands and specific residues in the ligandbinding site. The precise identification of the molecular
determinants in these crystal complexes and the understanding of the interactions with ligands are expected
to contribute to the development of highly selective
(subtype-specific) nAChR modulators and to the
design of specific drugs for several nAChR-related
disorders, such as Alzheimer’s disease (AD) and Parkinson’s disease (PD), as well as nicotine addiction. In
addition to the Ach-binding site, other binding sites on
nAChRs have also been identified. There is a binding
site for positive allosteric modulators around and
including Lys125 of the N-terminal extracellular region
of the a1 subunit [57], a steroid-binding site at an allosteric site distinct from both the ACh-binding site
and the ion channel [58] and two binding sites for noncompetitive blockers or negative allosteric modulators
[59]; the first site is located on the M2 transmembrane
segments of the nAChR within the pore and the
second site appears to lie at the interface between the
nAChR protein and membrane lipids. Interestingly, a
recently performed molecular docking of three known
allosteric modulators of nAChRs (galanthamine,
codeine and eserine) with AChBP and with models of
human a7, a3b4 and a4b2, based on the X-ray structure of AChBP, identified three distinct binding sites,
located in the channel pore [60]. The first site seems to
be located between the L1 and L4 loops (a ⁄ + subunit)
FEBS Journal 274 (2007) 3799–3845 ª 2007 The Authors Journal compilation ª 2007 FEBS
Nicotinic acetylcholine receptors
D. Kalamida et al.
and the b3 and b5 strands (b ⁄ – subunit), situated
exactly opposite the agonist-binding site, at a distance
< 12 Å. The second site appeared to be situated
between loops L4 (a ⁄ + subunit) and L5 (b ⁄ – subunit)
and finally the third site was shown to be close to L5
loop and b7 and b2 strands (b ⁄ – subunit). Considering
the position of the third site at the bottom of the
LBD, this might be implicated in the modulation of
the motions for opening or closing the gate of the
channel.
Agonist binding
Crystal structures of L-AChBP with bound nicotine or
carbamylcholine [16] and more recently of Aplysia californica AChBP (A-AChBP) with bound lobeline or
epibatidine [55] revealed agonist binding to a pentameric LGIC. The architecture of the ligand-binding
pocket, with the aromatic nest made of Tyr93, Trp147,
Tyr188 and Tyr195 from the plus face and Tyr55 from
the minus face of the interface (A-AChBP numbering),
is highly conserved in the AChBPs (Fig. 4B–E). In all
agonist-bound AChBP structures, the agonist is fully
enveloped by the protein through hydrogen bonds,
p-cation, dipole-cation and van der Waals forces. A
highly conserved Trp143 (L-AChBP numbering) from
the principal face of the subunit at the center of each
complex makes strong aromatic p-cation interactions
with the agonists (Fig. 4B). The highly conserved
Asp85 (L-AChBP numbering) also has a major structural role. It is situated behind the central Trp143 and
polarizes its main-chain carbonyl group, further stabilizing the ligand positively charged group. The vicinal
disulfide is in contact with all four agonists, mostly
through Cys187 with carbamylcholine and through
Cys188 with nicotine (L-AChBP numbering). In all
cases, ligand-binding is further supported by interactions with residues of the less-conserved minus face,
creating ligand-binding specificity. Both lobeline and
epibatidine exhibit additional interactions with AAChBP Trp147 (L-AChBP Trp143). The central piperidine ring of lobeline exhibits a stacking interaction
with A-AChBP Trp147, favoring a hydrogen bond
between the tertiary amine and the Trp carbonyl oxygen, whereas the pyridine amine of epibatidine binds
to A-AChBP Trp147 and Ile118 (Leu112 in L-AChBP)
through a solvent molecule. Tyr185 in L-AChBP
(Tyr188 in A-AChBP) makes aromatic contacts with
carbamylcholine and epibatidine (Fig. 4B), but not
with nicotine, and forms van der Waals interactions
with the lobeline methyl group. Tyr192 in L-AChBP
(Tyr195 in A-AChBP) forms aromatic contacts with
all agonists and L-AChBP Tyr89 (A-AChBP Tyr93)
contacts the ligands with its methyl group. This Tyr93
is further involved in the formation of a second hydrogen bond between the bridge ring of epibatidine and
A-AChBP (Fig. 4E). Leu112 and Met114 in L-AChBP
(Met116 and Ile118, in A-AChBP, respectively) make
hydrophobic contacts with nicotine, carbamylcholine
and lobeline, whereas Ile118 is involved in the first
hydrogen bond between epibatidine and A-AChBP.
The L-AChBP Arg104 (A-AChBP Val118) makes
hydrophobic contacts with carbamylcholine and polar
contacts with epibatidine. Interestingly, L-AChBP
Leu102 and Met114 residues contribute significantly to
nicotine binding, as they form a hydrogen bond with
pyridine N1 through a water molecule. Nicotine binding is further enhanced by a second hydrogen bond
between the pyrollidine N2 and the carbonyl group of
L-AChBP Trp143.
Antagonist binding
Binding sites for competitive antagonists, such as
snake neurotoxins and cone snail conotoxins are also
located at a subunit interfaces [35,61]. Using synthetic
peptides, a-neurotoxin-binding sites have been identified on both neuronal and muscle a subunits [62–66].
Specifically, we found that the main a-Bgtx-binding
segment corresponds to residues 189–195 in the
Torpedo [63] and to 186–197 in the human a7 subunit
[66]. The a-Bgtx-binding segment in human a1 subunit
corresponds to residues 185–196 [62].
Photoaffinity-labeling, site-directed mutagenesis and
competition studies have shown that the a-Bgtx-binding sites on the neuronal a7 and muscle-type nAChRs
have a similar conformation and interact with identical
or overlapping sites on the a-Bgtx molecule. An aromatic amino acid in loop C (Phe187 in human a7) is
necessary for a-Bgtx binding, judging by the reduced
a-Bgtx binding affinity of human a1 and the absence
of a-Bgtx binding to neuronal a3 and a4 receptors,
which have nonaromatic residues at the homologous
position [63,66–68]. Negatively charged amino acids in
loop C are also important in maintaining an electrostatic interaction with positively charged amino acids
in a-Bgtx [63,66,69]. Although the same amino acid
sequences contribute to the agonist- and competitive
antagonist-binding sites, difference in antagonist-binding affinity can be attributed to species-dependent
amino acids. Glycosylation also affects a-Bgtx binding
to some, but not all, nAChRs [26,50,70–74]. Deglycosylation of the ECD of the human a1 subunit
(expressed in Pichia pastoris) abolishes a-Bgtx binding
[75], but has no effect on recombinant ECDs of Torpedo a1 and human a7 expressed in Escherichia coli
FEBS Journal 274 (2007) 3799–3845 ª 2007 The Authors Journal compilation ª 2007 FEBS
3807
Nicotinic acetylcholine receptors
D. Kalamida et al.
and P. pastoris [26,50,70], reflecting a differential role
of glycosylation in protein folding.
The crystal structures of L-AChBP with a-cobratoxin (a-Cbtx) [56], and of A-AChBP with a-conotoxins
(a-Ctxs) [17,18,55] and the alkaloid antagonist methyllycaconitine (MLA) [55], revealed the specific residues
that are involved in toxin ⁄ antagonist-binding and the
conformational changes induced in nAChR-LBD upon
antagonist binding.
a-Cbtx is a long a-neurotoxin, with a characteristic
three-fingered fold; four disulfide bonds form a
b-structure hydrophobic core from which three loops
(I, II and III) emerge. The a-helical loop II bears the
fifth disulfide bond, essential for recognition of the
respective neuronal nAChRs [76]. In the crystal structure of the complex L-AChBP–a-Cbtx, Cbtx loop II
was inserted deeply into each of the five ligand-binding
pockets with an angle 45 relative to the median axis
of the AChBP ring [56], through essentially hydrophobic and aromatic contacts. Phe29 and Arg33 on the tip
of Cbtx loop II interact with the highly conserved
Trp53, Tyr185, Tyr192 and Trp143 of the principal
side of the AChBP ligand-binding pocket (L-AChBP
numbering) (Fig. 5A). Additional contacts include
Cbtx residues surrounding the tip of loop II with
Glu163, Glu55, Leu112, Met114 and Tyr164 on the
complementary side. In nAChR models, amino acids
involved in a-neurotoxin binding have been mapped to
loop C in the principal component and to loop F in
the complementary component [77]. Binding of a-Cbtx
to AChBP induces a significant displacement of loops
C and F by 10 Å, uncapping the pocket and stabilizing C loop in an ‘open’ conformation similar to that
of the ligand-free AChBP structures [55].
a-Ctxs are short-peptide toxins in which a helical
region, braced by two conserved disulfide bonds, defines
a characteristic two-loop framework [78]. Because of
their rigid structure and amino acid diversity, they bind
distinct nAChR subtypes with unique selectivity. a-Ctx
PnIA, a competitive inhibitor of neuronal nAChRs,
shows higher affinity for a3b2 nAChRs than for the
a7 subtype [61,78]. However, Ala10Leu substitution
shifts its selectivity from the a3b2 to the a7 subtype
[79]. The PnIA double mutant (Ala10Leu ⁄ Asp14Lys)
[17] further increases the efficacy to chicken a7 nAChR
compared with the PnIA mutant (Ala10Leu) [80].
a-Ctx-ImI, a specific and high-affinity inhibitor of the
a7 nAChR [81], was recently reported to display even
higher affinity for the human a3b2 nAChR [82]. In the
AChBP–Ctx complexes, both PnIA double mutant and
ImI bind deeply into each AChBP ligand-binding
pocket, burying 60 and 75% of their solvent-accessible
surface area, respectively. Upon binding of these
3808
a-Ctxs, loop C of A-AChBP is stabilized in an ‘open’
conformation similar to the A-AChBP–Cbtx complex
[56]. In contrast, loop F does not undergo significant
movement, probably because a-Ctxs are much smaller
than a-Cbtx. The disulfide bond Cys2–Cys8 (loop I) of
both a-Ctxs interacts with the highly conserved disulfide
bond (in loop C) of AChBP (Fig. 5B,C). However,
PnIA double mutant and ImI use different interactions
to create a high-affinity and ⁄ or selective binding. The
PnIA double mutant makes mainly hydrophobic contacts in the ACh pocket using Pro7, Ala9 and Leu10 as
anchoring points, whereas ImI uses the tripeptide
Asp5–Pro6–Arg7 in loop I as a binding determinant
forming a salt bridge and more hydrogen bonds. The
Leu10 side chain of PnIA, responsible for high affinity
for a7 nAChR is positioned in a hydrophobic pocket
formed by Val146 (principal side), Val106, Met114 and
Ile116 (complementary side), whereas Lys14 of this
double mutant is exposed to the solvent and does not
affect binding to A-AChBP. The selective binding of
ImI to nAChR subtypes is reflected in the specific inhibition of A-AChBP compared with L-AChBP [17,83].
The lack of affinity of ImI toward L-AChBP could, at
least in part, be explained by a substitution of Asp75
(A-AChBP), which forms a unique interaction with ImI
Trp10, by Gln in L-AChBP. In addition, Ile116 in
A-AChBP is similar to the corresponding Leu119 of the
a3b2 nAChR, recently identified as a key residue for
a-Ctxs binding [84]. In L-AChBP, this residue corresponds to the larger Met114 which may interfere with
Ctx binding. Finally, the substitution of Met114
(A-AChBP) by Leu112 in L-AChBP could further
contribute to loss of affinity of ImI to L-AChBP.
The stabilization of the loop C conformation of
A-AChBP in an ‘open’ state upon binding of a-Cbtx
and a-Ctxs, was also observed in the MLA–AChBP
complex (Fig. 5D) [55]. It therefore appears to be
essential for antagonist activity.
Conformational changes induced by ligand
binding
The structures of the agonist-bound AChBPs when
compared with the Hepes-bound or even better with
the apo-form (in the absence of any ligand or ligandlike molecule) A-AChBP crystal structure [55] revealed
conformational changes induced by agonist binding
which may trigger channel opening in intact nAChRs.
Analysis of the main-chain differences indicated that
loop C makes significant backbone movements, capping the entrance of the binding cavity and trapping
the bound agonists. In the uncapped conformation of
loop C, the conserved Tyr185 (equivalent to aTyr190
FEBS Journal 274 (2007) 3799–3845 ª 2007 The Authors Journal compilation ª 2007 FEBS
Nicotinic acetylcholine receptors
D. Kalamida et al.
Fig. 5. Representation of the ligand-binding site and of the interactions upon antagonist binding in (A) Lymnaea stagnalis AChBP (a-cobratoxin bound) [56], (B) Aplysia californica AChBP (a-conotoxin PnIA (Ala10Leu D14Lys) bound) [17], (C) Aplysia californica AChBP (a-conotoxin
ImI-bound) [55], and (D) Aplysia californica AChBP (MLA-bound) [55]. Reproduced from Celie et al. [17], Hansen et al. [55] and Bourne et al.
[56], with permission.
in the receptor) is 8 Å from the conserved Lys139
(equivalent to aLys145) in b7 strand, which forms a
salt bridge with the conserved Asp194 (equivalent to
aAsp200) in b10 strand. However, in the agonist-
bound crystal structures of AChBP, C loop tilts
inwards, with Tyr185 within 2–3 Å of Lys139 and
Asp194 displaced from the Tyr ⁄ Lys pair (L-AChBP
numbering). In the receptor, mutation of any of these
FEBS Journal 274 (2007) 3799–3845 ª 2007 The Authors Journal compilation ª 2007 FEBS
3809
Nicotinic acetylcholine receptors
D. Kalamida et al.
three residues markedly impairs channel gating and all
three are interdependent in contributing to gating [85].
So, when agonist binds, capping of loop C draws a
conserved Tyr into register with a conserved Lys,
forming a hydrogen bond which stabilizes the loop C
conformation in its binding conformation.
Apparently, all AChBP crystal structures can be
classified into two categories with respect to the loop
C conformation: ‘open’ or ‘closed’. The first category
includes the structures of the apo-form of A-AChBP
as well as all the antagonist-bound AChBPs (a-Cbtx-,
-a-Ctx–PnIA double mutant, a-Ctx–ImI- and MLA–
A-AChBP), representing the resting state of nAChRs.
The second category includes the AChBP structures
with bound agonists (nicotine–L-AChBP, carbamylcholine–L-AChBP, lobeline–A-AChBP and epibatidine–A-AChBP) or buffer molecules which mimic a
cationic agonist (Hepes–L-AChBP, Hepes–A-AChBP),
representing the activated state of the nAChRs. Upon
agonist binding, loop C clearly makes rigid-body
motions and swings as much as 11 Å between the two
extreme positions observed in the ImI and epibatidine
complexes. Superposition of all structures also indicates rigid-body movements of the b1–b2 loop and the
Cys loop, upon agonist binding.
Channel opening
Electron microscopy experiments on helical tubes from
Torpedo postsynaptic membranes (generated using a
specific technique to mimic the release of ACh and
thus allowing the study of the channel in the open
state) [7] first provided an insight into the gating mechanism of the nAChR channel. Ligand-binding triggers
a tertiary conformation change in the protein, resulting
in opening of the ion channel. Later studies of the
whole conformational change [86], the transmembrane
structure [24] and the closed channel [33] of the Torpedo nAChR, explained in more detail the mechanism
of channel opening. The subunits of the closed-channel
form have two alternative conformations: one characteristic of a subunits and the other characteristic of the
three non-a subunits. In both a subunits, the inner
b sheets of the LBD are rotated 10 relative to the
non-a subunits about a normal to the membrane plane
axis. The orientation of the outer b sheets in the a subunits is different compared with that in the non-a subunits. This special conformation of the a subunits is
called ‘distorted’, because these subunits convert to a
similar conformation to that of the non-a- and of the
agonist-bound AChBP. This distorted conformation is
stabilized by several interactions between the interfaces
of both sides of a subunits and the adjacent subunits,
3810
implicating residues on loop B and on the inner
b sheets.
By comparing the a-ECDs of the receptor in its
closed-channel form with the structure of the agonistbound AChBP [16,55], insight can be gained into the
rearrangements that should take place during ACh
binding, allowing the distorted a subunits to relax. In
the closed-channel form of the receptor, loop C projects away from the body of the a subunit, in contrast
with the ligand-bound AChBP, where loop C is closer
to the A and B loop residues implicated in ACh binding (e.g. the Ca distance between Cys192 of loop C
and Trp149 of loop B is 6 Å longer in the a subunits
of the closed-channel form of the receptor than in ligand-bound AChBP). B loops of the a subunits come
close to the b5 and b6 strands of the adjacent subunit
(c or d) and are possibly stabilized in this tense
arrangement by interactions across the subunit interface (e.g. salt bridge between aAsp152 and cArg78 or
dArg81). All these imply that these loops must
undergo quite large rearrangements to allow coordination of the binding residues to ACh. Both B and C
loops would surround the ACh molecule so as to
enable coordination of the relevant side chains. Loop
B would rotate clockwise and loop C would both twist
and rotate anticlockwise (Fig. 3B). Loop B plays an
interesting role in this local rearrangement as it joins
the outer and inner b sheets and therefore must participate directly in driving their relative displacements,
leading to the opening of the channel.
In principle, several components of the nAChR-LBD
could be involved in transmitting the conformational
changes upon ACh-binding, to the membrane-spanning
domain, where the channel gate is located. As shown in
the Torpedo nAChR structure only the Cys loop, the
b1–b2 loop and the covalently connected b10 strand of
the LBD make direct contacts with the membranespanning domain (Fig. 2B). Both loops interact with
amino acid residues of the M2–M3 segments; Cys loop
near the N-terminus of M3 and b1–b2 loop near the
C-terminus of M2 (Fig. 6). In the a subunits these loops
are 2–3 Å closer to the end of M2, along the M2–M3
linker than in the non-a subunits. The main amino acid
residues of the Cys loop that interact with residues Ile,
Tyr and Phe (aligning with aIle274, aTyr277 and
aPhe280) of M2–M3 linker are the consecutive PhePro-Phe (common to all five subunits). In the b1–b2
loop, two residues seem to be important for interaction
with the M2–M3; the residue aligning with aVal46
(Val44 in AChBP) and the adjacent highly conserved
Gln (aligns with aGlu45). The side chains of these residues together make an arc embracing the M2–M3
backbone. In the non-a subunits, the side chains
FEBS Journal 274 (2007) 3799–3845 ª 2007 The Authors Journal compilation ª 2007 FEBS
Nicotinic acetylcholine receptors
D. Kalamida et al.
sequence identity with human a7 nAChR and molecular modeling studies have predicted a very similar conformation for Glvi compared with AChBPs and
Torpedo nAChR [20]. A striking feature of Glvi protein is that in addition to the absence of the N-terminal a helix and the large cytoplasmic loop, it also
lacks the disulfide bridge forming the conserved Cys
loop. Therefore, it is very interesting to understand the
mechanism of channel opening in this protein, which
seems to function as a proton-gated channel, and trace
any contribution of the equivalent to the nAChR Cysloop domain of this protein. Also very interesting
would be to understand how this protein compensates
for channel selectivity, due to the lack of the cytoplasmic loop (lack of selectivity ‘windows’), compared with
Torpedo nAChR. Of course, all these issues remain to
be addressed by the elucidation of the structure of the
Glvi protein at high resolution, something that would
be of great value, as this would reveal the core structure of a complete LGIC for the first time.
Fig. 6. Representation of interactions between the Cys loop and
b1–b2 loop of the LBD with M2–M3 linker of the transmembrane
domain of Torpedo nAChR. The Cys loop, the b1–b2 loop, the
extension of M1 into b10-strand, and components of the membrane-spanning domain are in red, blue, green and gray, respectively. Reproduced from Unwin [33], with permission.
equivalent to Val46 seem slightly displaced from the
ends of M2 helices and do not make equivalent
contacts.
In conclusion, the opening of the channel gate upon
binding of ACh (or other agonist) to the LBD can be
explained in two ways. One possibility is that, following the relaxing rearrangements of the structural elements of the a subunits, both displaced b1–b2 and Cys
loops of the a subunits rotate back toward their relaxed
non-a locations and therefore stop interacting with the
relevant residues of the M2–M3 segment. Therefore,
M2–M3 is no longer ‘locked’ to one place and is free
to move due to its flexibility conferred by the conserved
aGly275 at the end of the M3, allowing gating motions
to occur (Fig. 6). Another possibility is that during the
displacements of b1–b2 and Cys loops following ACh
binding, the particular interaction involving aVal46 of
the b1–b2 loop is maintained and thus loop movement
drags the end of M2 away from the axis of the channel,
destabilizing the weak hydrophobic interactions holding the M2 helices of the gate together, so that they
break apart and the channel opens.
Interestingly, the recently characterized pentameric
Gloeobacter violaceus (Glvi) protein, considered as the
ancestor of eukaryotic pentameric LGICs, shares 20%
Muscle-type nAChRs
General features of muscle-type nAChRs:
fetal and adult types
In fetal muscle prior to innervation or after denervation and in fish electric organs, the nAChR subunit
stoichiometry is (a1)2b1cd [87], whereas, in adult
muscle, the c subunit is replaced by e to give the
(a1)2b1ed stoichiometry [88,89]. The c ⁄ e and d subunits are involved, together with the a1 subunits, in
shaping the ligand-binding sites and maintaining
cooperative interactions between the a1 subunits [90].
The presence of different non-a subunits confers different affinities to the two binding sites [91]. The site
in ac (the a1 subunit next to the c subunit) is biochemically distinguishable from that in ad due to the
fact that ac has a higher affinity for the competitive
antagonist, (+)-tubocurarine [23,92]. Binding of ACh
to the ac and ad sites induces conformational changes, predominantly in the a1 subunits, which are communicated to the transmembrane region, causing
channel opening [86]. The b1 subunit is important for
nAChR clustering, as shown by studies on hybrid
muscle nAChRs, in which the b1 subunit was
replaced by its neuronal counterpart [93]. Phosphorylation of unassembled c subunits and glycosylation of
the d subunit are required for increased nAChR
assembly efficiency [94,95].
The transition from the c-type to the e-type nAChR
occurs synchronously at all endplates within a fast
muscle, suggesting that neural activity causes suppres-
FEBS Journal 274 (2007) 3799–3845 ª 2007 The Authors Journal compilation ª 2007 FEBS
3811
Nicotinic acetylcholine receptors
D. Kalamida et al.
sion of the c subunit gene through transcriptional activation of the e subunit gene [96,97]. The fetal-type
nAChR continues to be expressed in the thymus and
in some extraocular muscles of the adult [96]. Transcriptional regulatory elements in the c subunit direct
muscle-specific, developmentally regulated, activitydependent synapse-specific transcription in vivo [98].
Humans, but no other species, express equal
amounts of mRNA for the normal a1 subunit and for
the a1* subunit, which contains 25 additional amino
acids between amino acids 58 and 59 [99], generated
by alternative splicing of the primary RNA transcript.
The a1* subunits do not undergo conformational maturation and do not form functional channels either
alone or with other subunits [100]. The significance of
this isoform is unknown.
Several mutations in genes encoding muscle-type
nAChR subunits induce premature termination of
translation or affect residues essential for nAChR
assembly. Such mutations in the e subunit are usually
compensated by expression of the fetal c subunit.
Re-expression of the fetal-type nAChR occurs in many
chronic neurogenic and some inflammatory myogenic
disorders and is surprisingly restricted to type I muscle
fibers (slow twitch or red) [101]. Type I fibers contain
more mitochondria and myoglobin and rely on aerobic
oxidation for energy [102]. Expression of fetal-type
nAChR is therefore crucial for maintaining neuromuscular transmission in deficiencies caused by nAChR
e subunit mutations and is associated with upregulation
of c subunit mRNA [101]. No significant upregulation
of a1 subunit mRNA has been seen in patients with
neurogenic disorders, in contrast to animal studies
[101,103,104]. This implies that the a1 and c subunits
are independently regulated. Alternatively, the a1 and
other subunits might be present in excess [101].
In addition to their critical physiological role, muscle-type nAChRs are an important autoantigen
involved in MG. MG is caused by failure of neuromuscular transmission as a result of the binding of
autoantibodies to muscle nAChR, which causes loss of
functional nAChRs, leading to defective signaling at
the NMJ [105]. (see section: Acetylcholine receptorassociated diseases).
nAChR gene expression and regulation at the
NMJ
During myogenesis, nAChR subunits are initially
expressed at low levels in myoblasts, then their expression increases and they are assembled and inserted into
the plasma membrane at a concentration of 103 lm)2
[106]. In mature muscles, nAChRs are present at high
3812
concentrations (104 lm)2) in the postsynaptic membrane at the NMJ, but are almost absent from the
remaining 99% of the muscle fiber surface [107]. Both
transcriptional
and
post-translational
processes
contribute to this redistribution in mature muscle:
(a) upon innervation, the diffusely distributed nAChRs
migrate to synaptic areas, where they become anchored
to the subsynaptic cytoskeleton; (b) nerve-derived factors cause transcriptional activation of the nAChR
genes in those nuclei that directly underlie the synapse;
and (c) muscle activity suppresses nAChR subunit gene
expression in nonsynaptic nuclei and promotes expression by synapse-associated nuclei [96]. Synapse-specific
nAChR transcription requires an enhancer sequence,
the N-box motif, which is present in the promoter
and ⁄ or intronic regions of nAChR subunit genes [108].
Expression of the e subunit is therefore controlled by
neurotrophic factors, whereas c-subunit expression is
regulated by both muscle electrical activity and neurotrophic factor(s) from the nerve, so that the c subunit
is expressed by extrasynaptic nuclei in developing, denervated and paralysed muscles and e-subunit expression is confined to subsynaptic nuclei under all
circumstances [89,96].
The multidomain proteoglycan, agrin, is a key factor
in the clustering of pre-existing nAChRs and in local
gene expression of postsynaptic proteins. It is synthesized by the nerve and released from motor nerve
terminals in the synaptic cleft, where it activates the
muscle-specific receptor tyrosine kinase (MuSK). As a
result of alternate splicing, agrin exists in several
isoforms that differ in their ability to induce nAChR
clustering [109]. Agrin-deficient knockout mice have
normal nAChR levels, but few postsynaptic clusters
[106]. Following activation by agrin, MuSK undergoes
autophosphorylation on several cytoplasmic tyrosine
residues, which then act as docking sites for signaltransducing molecules and promote nAChR clustering
through rapsyn [108–110]. Tyrosine phosphorylation of
the nAChR b1 subunit links nAChR to the cytoskeleton and contributes to efficient clustering. Rapsyn
interacts with the intracellular portion of nAChR subunits. Other effector molecules, such as neuregelin-1
and nitric oxide, have been implicated in promoting
clustering and gene expression at the NMJ, but all are
dependent on the agrin ⁄ MuSK signaling pathway
[109,111,112]. Kukhtina et al. [113] demonstrated that
the intracellular loop of the d subunit is unfolded
and predicted the presence of 12 functional binding
motifs involved in protein–protein interactions. Thus,
it is likely that other binding partners involved in
nAChR assembly, trafficking and clustering will be
discovered.
FEBS Journal 274 (2007) 3799–3845 ª 2007 The Authors Journal compilation ª 2007 FEBS
Nicotinic acetylcholine receptors
D. Kalamida et al.
Electrophysiological studies have shown that the
switch in the composition of nAChRs from fetal- to
adult-type is a key process in the maturation of the
NMJ, because it is accompanied by changes in the
functional properties of endplate channels, which are
the sites of neuromuscular contact [88]. Channel mean
open time decreases from 5 to 1.5 ms during the first
two postnatal weeks and mean channel conductance
increases by 50% [114–117]. Fetal- and adult-type
receptors have 39 and 59 pS single-channel conductance with increased Ca2+ influx and 10.4 and 5.3 ms
open time, respectively [88]. Extracellular Mg2+ and
Zn2+ affect differentially synaptic transmission of
fetal- and adult-type receptors in response to ACh by
reducing single-channel conductance of the former less
potently than the latter [118]. Studies on recombinant
T. californica nAChRs and chimeric fetal and adult
mouse nAChRs, in which M1–M4 of the c subunit
were exchanged for those of the e subunit and vice
versa, suggested that the difference in conductance can
be attributed to amino acid charge differences in
segments M1–M4, especially in M2 of the c and e subunits [119]. By contrast, the difference in channel open
time was attributed to M4 segment and the amphipathic helix between segments M3 and M4 [119,120].
This is in agreement with structural studies suggesting
that cations (Mg2+ and Zn2+) interact strongly with
the additional negative residues that line the
e-nAChR’s cytoplasmic and extracellular rings close
to the membrane-spanning M2 segment [121]. The
response of fetal and adult-type nAChRs to antagonists can also differ. For example, mouse fetal-type
nAChRs are more sensitive to 5-hydroxytryptamine
(5-HT), an open-channel blocker of muscle nAChRs,
than adult-type nAChRs [122]. Subunit composition
also affects the metabolic stability of nAChRs; the
half-life of adult-type nAChRs at the adult MNJ is
10 days compared with 24 h for fetal-type nAChRs
[123].
The c- to e-subunit switch coincides with other postnatal events in the maturation of the NMJ, such as
transition from multiple to single axon innervation of
the muscle fiber, elaboration of branched morphology
by the endplates, formation of junctional folds, as well
as localization and interaction with synaptic antigens,
raising the possibility that these events might be related [97,117]. The functional significance of the switch
has been investigated by studies on knockout mice for
the e-subunit gene [88,115,117,124,125]. Expression of
fetal-type receptors partially compensated for the lack
of adult-type receptors in these animals, which showed
severe muscle weakness and died prematurely. Adulttype nAChRs are therefore essential for the mainten-
ance of the highly organized structure of neuromuscular synapses in adult muscle [115,125]. Furthermore,
the shorter mean open time of adult nAChRs contributes to the stabilization of the initial nerve–muscle
contacts and synapse maturation in differentiated muscles [97,115]. Mutations in the pore-forming M2 region
of the e subunit causing high Ca2+ permeability by
prolonging channel mean open time also demonstrated
that the e subunit is a key determinant of Ca2+ permeability of adult muscle and protects the synaptic
region from the harmful effects of excessive calcium
influx [116,117,126,127]. Mutant mice in which the
fetal-type c subunit was replaced with a chimeric one
with e-subunit-like functional properties had altered
muscle innervation pattern, suggesting that fetal-type
nAChRs ensure an orderly innervation pattern for
skeletal muscle [115]. It has also been suggested that
the long duration currents mediated by fetal-type
nAChRs are necessary for developing spontaneous
muscle contractile activity and stabilizing the clustering
of nAChRs at the endplates [115,128].
Expression studies and mutational analysis have
provided insights into the regulation of nAChR assembly. Processing and assembly of nAChR subunits is a
slow process, taking 2 h to complete [129]. Torpedo
nAChR assembly, in particular, is temperature sensitive and increases as the temperature decreases from
37 to 20 C [130,131]. Two models have been proposed
for nAChR assembly.
The ‘sequential model’ suggests that newly synthesized subunits either assemble rapidly into a1b1c trimers or remain unassembled and are rapidly degraded
[130,131]. The chaperone protein, calnexin, associates
with 50% of newly synthesized subunits during the
early events of nAChR assembly, but not after maturation [132], promoting stabilization of unassembled
subunits into a1b1c trimers and, by slowly dissociating
from these, controls the rate of nAChR folding and
oligomerization. A rate-limiting step in nAChR assembly is the formation of a-Bgtx-binding sites on
a1b1c trimers. Such trimers capable of binding a-Bgtx
are short-lived intermediates and rapidly assemble into
a1b1cd tetramers, which can then bind ACh [133]. The
addition of the a1 subunit to the a1b1cd tetramers to
form the second ACh binding site is another rate-limiting step. The ‘sequential model’ therefore suggests that
subunits assemble at the interface between the b1 and
c subunits and that the second a1 subunit assembles at
the interface between the c and d subunits. N-Linked
glycosylation at residue 141 and the formation of
disulfide bridges between cysteines 128–142 and 192–
193 are also required for the correct maturation of the
Torpedo a1 subunit and the subsequent formation of
FEBS Journal 274 (2007) 3799–3845 ª 2007 The Authors Journal compilation ª 2007 FEBS
3813
Nicotinic acetylcholine receptors
D. Kalamida et al.
the a-Bgtx binding site, and are potential sites of regulation for nAChR assembly [70,134].
In the ‘heterodimer model’ [135–137], the a1 subunits
assemble with the c or d subunit, generating, respectively, a1c or a1d complexes and the ACh-binding sites
are formed at the interface of the heterodimers.
Following ACh-binding-site formation, the heterodimers assemble with b1 subunits into (a1)2b1cd
heteropentamers. As in the ‘sequential model’, interactions with other subunits or chaperones are required
to prevent misfolding. Evidence supporting this model
came from experiments in which the a1 subunits were
either expressed alone or in combination with one of
the other subunits (b1, c or d) [133,138,139]. However,
the ‘heterodimer model’ cannot account for the two
distinguishable ACh-binding sites being formed at
different times and on different subunit complexes
during assembly [133].
Both models propose that discrete assembly intermediates are formed and confined in the endoplasmic
reticulum (ER), whereas unassembled subunits or
incorrectly processed and assembled complexes are
recognized by the cellular machinery and degraded
through proteasomes prior to reaching the Golgi apparatus, leading to the accumulation of only correctly
assembled complexes [133]. The ubiquitin–proteosome
system through ER-associated degradation is thought
to be involved in the control of this process [140].
Expression of recombinant muscle-type nAChRs
The expression of recombinant polypeptides corresponding to functional domains of muscle nAChRs is
essential for their detailed physiological and structural
analysis. Because of the high yield of protein, prokaryotic expression systems were initially used. However,
because of the lack of essential post-translational modifications, they are considered to be problematic,
because aggregation of nAChR subunits occurs and
denaturating conditions are therefore required to solubilize the protein. Eukaryotic expression systems,
despite the usually lower yield, offer the advantage of
post-translational modifications, such as glycosylation,
so the expressed proteins may have near-native conformational features.
Full-length, truncated and mutant T. californica,
mouse, rat and human muscle-type nAChR subunits
have been expressed in E. coli, Saccharomyces cerevisiae, P. pastoris, baculovirus-infected insect cells, Xenopus oocytes, mouse fibroblasts, HEK 293 cells and
COS cells. A far from exhaustive overview of some of
these studies, which provided insights into the function
of the individual subunits, is presented below.
3814
Torpedo nAChR
The ECD of the T. marmorata a1 subunit (residues 1–
209) was expressed in inclusion bodies in E. coli [26].
Following renaturation, it exhibited high a-Bgtx binding affinity, suggesting that it had a near-native conformation. The same fragment of the T. californica a1
subunit ECD was also expressed in E. coli in inclusion
bodies and was in vitro refolded [27]. The renatured
protein bound a-Bgtx and mAbs raised against the
intact T. californica nAChR [27]. Two fragments of
the T. californica a1 subunit, consisting of part of the
ECD with and without the first transmembrane segment (residues 143–210 and 143–238, respectively) were
expressed in E. coli and had a significant affinity for
a-Bgtx [28]. This study was important in identifying
the ECD determinants necessary for a-Bgtx binding
[141].
Mouse nAChR
The ECD of the mouse nAChR a1 subunit (residues
1–210) fused with a glycosylphosphatidylinositol
anchorage sequence has been expressed in mammalian
cells (CHO cells) and Xenopus oocytes [25]. The purified protein bound a-Bgtx and had the conformationspecific binding sites expected for the correctly folded
subunit. Furthermore, a similar construct (residues
1–211) has been expressed in the yeast, P. pastoris, as
a soluble protein with a high affinity for a-Bgtx and
conformation-dependent mAbs [29].
Human nAChR
We expressed the ECD of the human nAChR a1 subunit (residues 1–207) in E. coli and reconstituted it
using the ‘artificial chaperone’ approach [142]. The
reconstituted protein bound conformation-dependent
anti-nAChR mAbs, but not a-Bgtx, apparently due to
lack of N-glycosylation which seems to be necessary
for high-affinity binding of a-Bgtx to mammalian muscle nAChR [75]. The slightly longer form of residues
1–210 has also been overexpressed, in our laboratory,
as a water-soluble molecule in P. pastoris [75]. This
glycosylated molecule bound a-Bgtx, conformationdependent anti-nAChR mAbs and myasthenic
patients’ antibodies against nAChR [143]. A fusion
between the ECD of human nAChR a1 subunit (residues 1–205) and maltose-binding protein was constructed and expressed in E. coli as a soluble protein [144].
The nAChR a1 fragment was recovered following
protease cleavage. Both the fusion protein and the
nAChR a1 fragment seemed to have a near-native
FEBS Journal 274 (2007) 3799–3845 ª 2007 The Authors Journal compilation ª 2007 FEBS
Nicotinic acetylcholine receptors
D. Kalamida et al.
conformation judging by the binding of conformationdependent mAbs.
The ECDs of the human muscle non-a subunits, b1
(residues 1–221), c (residues 1–218) and e (residues
1–219), have also been expressed, by our group, in
P. pastoris as soluble glycosylated proteins [48]. The
proteins seem to have native-like conformational characteristics, as evidenced by the high binding affinity for
conformation-dependent anti-nAChR mAbs and autoantibodies from the sera of MG patients [145]. Variants of these proteins with mutations and ⁄ or the
addition of hydrophilic epitopes have also been constructed in our laboratory to increase their solubility
and expression yield for structural trials (unpublished
data).
Coexpressions
All four full-length T. californica subunits have been
expressed in S. cerevisiae and had the antigenic properties of authentic Torpedo subunits [146]. Combinations
of all four full-length T. californica subunits (a1, b1, c
and d) have been stably expressed in Xenopus oocytes
and mammalian cell lines and have provided insights
into nAChR assembly and regulation [135,139,147–
150]. These studies demonstrated that incorporation of
the b1 subunit into the a1cd complex is a rate-limiting
step in nAChR assembly, that association of the a1
subunit with either the c or the d subunit is necessary
for agonist binding, and nAChRs lacking the b1, c or
d subunit show weak channel activity. Furthermore,
coexpression of T. californica nAChR subunits carrying site-directed mutations at conserved sites (N-glycosylation and disulfide bond) and normal subunits in
Xenopus oocytes led to intracellular retention of most
of the assembled nAChR complexes and allowed the
identification of regulation sites for nAChR assembly
[71,72,151]. The ECDs of all four T. californica subunits have been coexpressed in baculovirus-infected
insect cells and assembled into pseudo-pentameric
native-like nAChR ECD complexes [49]. The intracellular loop of the Torpedo d subunit has also been
expressed in E. coli and, following urea treatment and
efforts at refolding, was found to be unfolded by
NMR analysis complemented by protein structure prediction algorithms, suggesting that an interaction with
as yet unknown protein partners is required for it to
assume an ordered conformation [113].
Hybrid bovine–T. californica and mouse–T. californica nAChRs have also been expressed in Xenopus
oocytes as functional receptors [152–155]. These
hybrids were useful for probing channel-gating determinants within the primary sequence of the individual
subunits and assessing species differences in channel
properties. Hybrid cat–T. californica nAChRs, expressed in Xenopus oocytes, have shown that species
differences in the c subunit primary sequence account
for the observed differences in the properties of the
receptors [156]. Torpedo and hybrid rat–Torpedo
chimeras expressed in mammalian muscle cell lines
provided insights into the temperature sensitivity of
Torpedo nAChR assembly [157]. All muscle-type
a1 subunits are encoded by a single gene, except in
Xenopus, in which two genes have been identified; each
of these was coexpressed with T. californica b1, c, and
d subunits and the two nAChRs were found to have
strikingly different affinities for a-Bgtx ( 1000-fold
difference) [158]. Hybrid expression has also proved
useful in structural studies. Expression of hybrid
bovine–T. californica nAChRs in Xenopus oocytes
showed that M2 and the bend between M2 and M3 of
the d subunit are important in determining the rate
of ion transport through the open channel [159].
Hybrid T. californica a1–mouse b1, c, d nAChRs and
human a1–T. californica b1, c, d nAChRs expressed in
mammalian muscle cells (fibroblasts and TE671 rhabdomyosarcoma cells) revealed the immunodominance
of antia1 subunit sera in anti-MG sera [160,161].
Expression of combinations of mammalian subunits
in Xenopus oocytes and muscle cell lines has allowed
the measurement of the kinetics of subunit assembly,
as well as the study of the electrophysiological and
biochemical properties of the resulting complexes
[100,136,137,162–168]. Coexpression of all four mammalian receptor subunits as functional receptors in
muscle cell lines has also been carried out
[100,116,168–170]. Furthermore, all four full-length
mammalian receptor subunits have been expressed as
fully assembled functional receptors in Xenopus
oocytes and nonmuscle cell lines to determine the
properties of the receptors in the absence of interactions with other muscle proteins [95,168,171–173].
The resulting receptors had the pharmacological,
electrophysiological and metabolic properties of nAChRs normally found in myotubes and have proved
valuable in studying nAChR synthesis, assembly and
transport to the cell surface. Human a1 nAChR subunit exists in two isoforms (a1 and a1*), which differ
in length, as mentioned previously. Each isoform has
been coexpressed with the other three full-length fetal
and adult subunits in Xenopus oocytes and the properties of the corresponding fetal and adult channels
investigated [100]. These studies, complemented by
site-directed mutagenesis of the two a1 isoforms,
demonstrated that these isoforms differ in the porelining region and that only the a1 subunit, expressed
FEBS Journal 274 (2007) 3799–3845 ª 2007 The Authors Journal compilation ª 2007 FEBS
3815
Nicotinic acetylcholine receptors
D. Kalamida et al.
in combination with the other subunits, is integrated
into functional receptors. Differences in the electrophysiological properties of the corresponding fetal and
adult receptors were also identified. Truncated fragments of the mouse nAChR a1 subunit containing the
ECD and all, part or none of the first transmembrane
segment and the d subunit were coexpressed in COS
cells, showing that the M1 segment is necessary for
heterodimer formation [174].
Animal studies
Chimeric subunits consisting of the N-terminal of
the c subunit and the C-terminal of the e subunit
have been constructed by homologous recombination
and expressed in mice [119]. Mice homozygous for
the chimera were healthy and fertile and expressed
nAChRs with the properties of the adult-type endplate throughout their lifetime; the authors concluded
that one function of the fetal-type nAChR is to
ensure the orderly innervation of the skeletal muscle
[115].
Neuronal-type nAChRs
General features of neuronal-type nAChRs
Neuronal nAChRs are widely expressed in the nervous
system in peripheral ganglia and certain areas of the
brain, and in nonexcitable cells, such as epithelial cells
and cells of the immune system. To date, nine a (a2–
a10) and three b (b2–b4) subunit genes have been
cloned. The a7–a10 subunits are found either as homopentamers (of five a7, a8 or a9 subunits) or as heteropentamers (of a7 ⁄ a8 and a9 ⁄ a10) [175]. By contrast,
the a2–a6 and b2–b4 subunits form heteropentameric
receptors, usually with a (ax)2(by)3 stoichiometry. The
a5 and b3 subunits cannot form functional receptors
when expressed alone or in paired combinations with b
or a subunits, respectively. They only form operational
channels when coexpressed with other functional subunit combinations, and are thus commonly termed
‘orphan’ or ‘auxiliary’ subunits. Although these subunits were initially thought to have a structural role,
emerging evidence showed that a5 incorporation into
a6b4b3 receptors was required for high-affinity nicotinic ligand binding, suggesting that they play a critical
part in the assembly and pharmacological properties of
nAChRs and their role in ligand binding may have to
be readdressed [176].
Earlier classification of the nAChRs was based
upon their pharmacological properties. As a result,
neuronal-type nAChRs were divided into two classes:
3816
(a) the high-affinity agonist-binding class (with nm
affinities), which do not bind a-Bgtx, later found to be
the heteropentameric nAChRs formed by a2–a6 and
b2–b4 subunits; and (b) a second class that binds
agonists with lower (lm) affinities and binds a-Bgtx
with nm affinities (sometimes referred to as a-Bgtx–
nAChRs), later shown to be usually homopentameric
molecules formed by a7–a9 subunits [105]. The homopentameric nAChRs are thought to have five identical
ACh-binding sites per molecule (one at each a subunit
interface), whereas the heteropentameric nAChRs have
two ACh-binding sites, located at the interface between
an a and a b subunit.
nAChR subunit composition has been shown to be
important in regulating the response to agonists and
the subcellular localization of assembled channels.
Ectopic expression of the b2 and b4 subunits with
combinations of a2–a4 subunits in HEK 293 cells
revealed that b2-containing nAChRs have a higher
affinity for most ligands than b4-containing nAChRs
[177]. Recently, Lukas’s group [178] showed that
a4 nAChRs containing either b2 or b4 subunits have
distinct pharmacological and physiological properties,
with a4b4 nAChRs having higher current amplitude
and stronger responses to agonists than a4b2 nAChRs.
Incorporation of an auxiliary subunit into an
ab nAChR alters its properties [179]; for instance, the
a4a5b2 nAChR has a higher Ca2+ permeability and
desensitization rate and higher EC50 values than the
a4b2 nAChR [180]. Moreover, when a5 is coexpressed
with a3 and b2 subunits, the receptor formed shows
increased ACh sensitivity compared with the a3b2
receptor [41]. Interestingly, this effect is not observed if
b4 is used instead of b2, whereas an increase in desensitization rate and Ca2+ permeability is seen with both
the b2- and b4-containing channels. Furthermore, Fischer and colleagues [181] have shown that deletion of
a5 in mice has different effects depending on the localization of the nAChR. Somatic nAChRs from neurons
of the superior cervical ganglion had different agonist
potencies depending on the presence of a5, but the
magnitude of the response was not affected, whereas,
in the case of presynaptic receptors, the response was
significantly higher in neurons not expressing a5.
Incorporation of the a5 subunit has also been shown
to affect the receptor affinity for agonists and antagonists in neurons of the autonomic nervous system
[182].
Given the intricate role of each nAChR subtype at
various positions in the cell and its contribution to
signal transmission, nAChR targeting at specific sites
is crucial. For instance, in the chick ciliary ganglion
a3, a5 and b4 containing receptors are located at
FEBS Journal 274 (2007) 3799–3845 ª 2007 The Authors Journal compilation ª 2007 FEBS
Nicotinic acetylcholine receptors
D. Kalamida et al.
the interneuronal synapse, whereas a7 receptors are
excluded from the synapse and are scattered perisynaptically. The cytoplasmic loop of the a3 subunit appears
to regulate receptor targeting in this case, as exchanging the loop of a7 with that from a3, but not a5,
causes a7 to aggregate in the synaptic region [183].
Studies in cultured hippocampal neurons showed that
a7 nAChRs were distributed in the somatodendritic
area, while a4b2 nAChRs were found both in the
dendrites and axon. Further analysis using fusion
proteins of CD4 or interleukin (IL)-2 receptor,
normally universally distributed, with domains of the
nAChRs intracellular loops reveiled that there is a 48
amino acid stretch of the a7 subunit loop conferring
dendritic distribution and a 25 amino acid region in
the a4 subunit loop responsible for axonal targeting
[184].
Neuronal-type nAChR-mediated Ca2+ signaling
Neuronal-type nAChRs show a significant, subtypedependent permeability to Ca2+ and a strong inward
rectification (voltage dependency) [185,186]. The high
Ca2+ selectivity of neuronal-type nAChRs is of pivotal
physiological importance, because intracellular Ca2+
signals are involved in the rescue ⁄ demise of developing
neurons and the modulation of their activity.
Although, Ca2+ influx is often preceded by Na+ membrane depolarizing ion currents, within this part of the
review, only the neuronal-type nAChR-mediated Ca2+
signaling are discussed.
The neuronal-type nAChR-mediated increase in the
intracellular Ca2+ concentration results primarily from
direct permeation of the nAChR pore, due to its high
Ca2+ permeability. The Ca2+ influx causes membrane
depolarization and subsequent activation of voltageoperated Ca2+ channels (VOCCs) [187,188] or,
alternatively, triggers further Ca2+ release from ryanodine-dependent intracellular stores (calcium-induced
calcium release, CICR), generating prolonged Ca2+
signals [188–191]. It seems that different neuronal-type
nAChR subtypes are coupled with different Ca2+
pathways. The most Ca2+-permeable homomeric
a7 nAChRs, albeit capable of activating VOCCs
[192,193], mainly induce Ca2+ currents, which subsequently trigger the CICR machinery [188,194,195]. By
contrast, neuronal nAChRs containing a3- and ⁄ or
b2 subunits in the brain and ganglionic neurons operate primarily through Ca2+ signals coupled with the
opening of VOCCs [188,190,194]. Thus, second messenger Ca2+ signals act as converters of the acute
neuronal nAChR stimulation into sustained downstream effector functions, such as neurotransmitter
release, gene expression and metabolism, which shape
neuronal activity. Interestingly, a reciprocal relationship exists, with intracellular Ca2+ levels affecting
neuronal-type nAChR activity [196,197].
Neuronal-type nAChR expression
Expression of neuronal-type nAChR subunits varies in
different cell types and neurons located in different
parts of the nervous system. nAChR a4, a5, a6, b2
and b4 subunits, expressed in mesostriatal dopaminergic neurons assemble in different nAChRs combinations that play a role in modulating the release of
striatal dopamine (DA) [198,199]. a7 receptors, which
account for the majority of the a-Bgtx-binding sites in
the nervous system, are widely distributed and are present at a high concentration in the hippocampus, especially in GABAergic neurons [200–202]. a8-containing
receptors have been found only in the chick nervous
system as homopentamers or in combination with a7,
while a9 receptors are mainly expressed in the cochlea
and sensory ganglia of the nervous system [203].
Finally, a10 appears to serve as a structural subunit in
the formation of functional receptors with a9 in
cochlear mechanosensory hair cells; these receptors
mediate transmission to efferent olivocochlear fibers
[175,204].
Because different subunit combinations can lead to
different pharmacological properties, the array of
subunits expressed by any given cell affects its
responses to stimuli. Dissection of the promoter of the
a3 and a5 genes has revealed many different regulatory
elements required to meet the needs of specific cells
[205–208].
Once synthesized, nAChR subunits must assemble
into pentameric channels and be transported from the
ER through the Golgi to the cell surface. nAChRs
that have not been properly assembled are targeted to
the proteasome for degradation. Similar to muscle
nAChRs, ubiquilin-1, a member of the ubiquitin protein family, is thought to be involved in controlling
this process [209]. In the case of a7, it has been
shown that, in COS cells, sequences within its M1
segment prevent its surface localization [210], whereas,
in a4 and b2, a stretch of conserved hydrophobic
amino acids in the cytoplasmic domain between M3
and M4 is required for efficient transport to the cell
surface [211]. RIC-3, an ER ⁄ Golgi-localized protein,
appears to have a dual effect on nAChR trafficking,
retaining mature a4b2 nAChRs intracellularly and
enhancing surface transport of functional a7 nAChRs
[212,213]. a7 nAChRs form characteristic clusters in
the somatic spines of ciliary neurons. Lipid rafts are
FEBS Journal 274 (2007) 3799–3845 ª 2007 The Authors Journal compilation ª 2007 FEBS
3817
Nicotinic acetylcholine receptors
D. Kalamida et al.
essential for maintaining these clusters, as they promote the interaction between a7 nAChRs and rapsyn
that is required for nAChR clustering [214,215].
Upon stimulation, a7 receptors are internalized and
new receptors are targeted to the surface from intracellular pools, both processes being controlled by the
soluble N-ethylmaleimide-sensitive factor attachment
protein receptor (SNARE) [216]. Although this does
not alter the number of surface nAChRs, it is necessary for maintaining the ability of the nAChR to
respond to subsequent stimuli and propagate downstream signaling.
Exposure to nicotine results in an increase in the
number of nicotinic ACh-binding sites. Early results
showed that a4b2 nAChRs are upregulated in rat
brains after chronic exposure to nicotine [217,218], and
further investigation revealed that this upregulation is
dependent on the nAChR subtype. More specifically,
nicotine upregulates mostly high-affinity nAChRs such
as a3b4 and a3b2 [218–221]. This upregulation is not
dependent upon mRNA transcription or de novo protein synthesis, because there is no detectable increase
in levels of subunit mRNA or total protein (surface
plus intracellular), and protein synthesis inhibitors do
not block the nicotine-induced upregulation. Furthermore, it appears that the flow of ions through the
channel is not required either, because the channel
blocker, mecamylamine, has an additive effect to nicotine [222]. Indeed, upregulation is due to an increase in
the proportion of the nAChR that is expressed on the
cell surface and this is thought to occur due to a nicotine-driven increase in the assembly of intracellular
nAChR subunits and a decrease in the rate of their
internalization [223,224]. Despite the increase in surface nAChRs, function is not enhanced and the upregulated channels have a decreased capacity for ion flow
[219,221]. Upregulation is also observed in cells ectopically expressing nAChRs, indicating that this is not an
adaptive mechanism to compensate for desensitized
nAChRs [222,225].
attention to the role of the neuronal-type nAChRs in
the periphery.
Neuronal nAChRs in the CNS
Modulation of neuronal transmission
Neuronal nAChRs are widely distributed in the brain.
In terms of subcellular localization, they are found in
pre-, peri- and postsynaptic areas [231,232] (Fig. 7).
Presynaptic and perisynaptic nAChRs act as autoreceptors or heteroreceptors regulating the release of
several important neurotransmitters, mainly ACh,
DA, norepinephrine, glutamate, 5-HT and 4-aminobutyrate throughout the CNS. It is noteworthy that the
synaptic release of a particular neurotransmitter can
be regulated by different neuronal-type nAChR subtypes in different CNS regions. For instance, DA
release from striatal and thalamic DAergic neurons is
controlled, respectively, by the a4b2 subtype or both
the a4b2 and a6b2b3 subtypes [233,234]. Interestingly,
glutamatergic neurotransmission seems to be exempt
from this ‘pluralistic’ neurotransmitter control
network, being ubiquitously regulated by a7 nAChRs
[233].
Presynaptic neuronal-type nAChRs modulate neurotransmitter release by promoting exocytotic mechanisms either through activation of VOCCs following
membrane depolarization or by direct alteration of the
intracellular Ca2+ concentration due to the intrinsic
Ca2+ permeability of the neuronal-type nAChR pore.
There is evidence suggesting that neurotransmitter
release is affected by both mechanisms. DA release
from striatal synaptosomes is mediated by nona7 neuronal nAChR subtypes functionally coupled to
Roles of neuronal-type nAChRs
Although neuronal-type nAChRs are major players in
rapid synaptic transmission in the PNS [226,227], the
relatively few examples of neuronal nAChR-mediated
fast-signal propagation in the mammalian CNS
[228–230] suggest that, in the brain, neuronal-type
nAChRs act in a more sophisticated way, mainly as
synaptic modulators [5]. Recently, the identification of
several neuronal-type subtypes in non-neuronal cells
and, most importantly, the discovery of the antiinflammatory cholinergic pathway have attracted much
3818
Fig. 7. Neuronal nAChRs can be found at perisynaptic, presynaptic
and postsynaptic areas. Depending on their distribution, they can
exert a broad variety of functions modulating neuronal signaling at
both the pre- and postsynaptic levels.
FEBS Journal 274 (2007) 3799–3845 ª 2007 The Authors Journal compilation ª 2007 FEBS
Nicotinic acetylcholine receptors
D. Kalamida et al.
VOCCs [235–237], whereas, at rat hippocampal glutamatergic terminals, Ca2+ entry through a7 nAChRs
initiates CICR from presynaptic stores, inducing glutamate release and eventually eliciting bursts of miniature excitatory postsynaptic currents [238]. In addition
to Ca2+-dependent exocytosis, neuronal-type nAChRs
modulate neurotransmitter release through secondary
messenger pathways, allowing them to exert more
subtle regulatory actions. Ca2+-dependent protein kinase C has been implicated in striatal DA release by
neuronal nAChRs [239] and extracellular signal-regulated mitogen-activated protein kinase (ERK ⁄ MAPK)
stimulation by protein kinase C has been related to
nicotine-evoked catecholamine release by adrenal medullary cells [240].
Postsynaptic nAChRs probably mediate a small
proportion of the fast excitatory transmission in the
CNS and are generally considered of less physiological
importance than the presynaptic and perisynaptic
nAChRs. Although it has been quite difficult to locate
sites in the brain where ACh release produces fast
postsynaptic nicotinic responses, the presence of
postsynaptic a7, a4b2 and a3b4 subtypes has been
demonstrated in several brain regions [233,241]. In
particular, when the nicotinic postsynaptic excitation
coincides with presynaptic glutamate release, postsynaptic nAChR activity has been shown to enhance the
induction of synaptic potentiation in the hippocampus
[242].
Regulation of gene expression by nAChRs
The expression of various neuronal-type nAChR subtypes early during embryogenesis [243–246] and the
existence of a primary machinery enabling both ACh
synthesis and the response to ACh [247] suggest a role
for cholinergic signaling during early neuronal
development. Although the importance of neuronaltype nAChR expression before neuronal differentiation
remains obscure, the involvement of neuronal-type
nAChRs in the regulation of early gene expression
was originally suggested by the Ca2+-dependent regulation of c-fos gene transcripts after nAChR activation
in PC12 cells [248]. Nicotine-induced upregulation of
c-fos and junB gene expression has now been demonstrated in various brain regions [249–251]. Because
these immediate-early genes, generally involved in
mechanisms of abuse, function as transcription factors,
their nicotine-mediated upregulation suggests that
nicotine may modulate the expression of additional
genes. In fact, exposure of SH-SY5Y cells to nicotine
has been shown to alter the expression of a diverse set
of proteins, including transcription factors, protein
processing factors, RNA-binding proteins and plasma
membrane-associated proteins, and there is evidence
implicating activation of neuronal-type nAChRs in
this gene profile alteration [252].
In addition, neuronal-type nAChR-mediated gene
regulation provides a means for modulating neurotransmitter release per se, as demonstrated by the
example of tyrosine hydroxylase [253,254]. This
enzyme catalyzes the crucial rate-limiting step in catecholamine biosynthesis and is subject to various
control mechanisms. Prolonged nicotine treatment upregulates tyrosine hydroxylase expression both in vivo
and in cell culture [253], and stimulation of a7
nAChRs is implicated in the nicotine-triggered increase
in tyrosine hydroxylase mRNA levels [254]. This effect
is Ca2+ dependent and requires a sustained elevation
of the intracellular Ca2+ concentration due to release
from intracellular stores and downstream activation of
protein kinase A or MAPK, which, in turn, phosphorylate the cAMP-response element-binding protein
(CREB) [255–257].
Contribution to plasticity and memory functions
Neuronal-type nAChRs have been implicated in cognition, mainly because of the respective beneficial or
adverse effect of neuronal-type nAChR agonists and
antagonists on learning and memory. The hippocampus, generally thought to be the key brain area for the
encoding and retrieval of memory, has attracted particular interest with respect to neuronal-type nAChR
activity. In the hippocampus, at least three distinct
functional neuronal-type nAChR subtypes (a7, a4b2
and a3b4) can be detected [258]. Synaptic function in
the hippocampus seems to result from GABAergic
(inhibitory) and glutamatergic (excitatory) inputs to
the hippocampal interneurons, which are modulated
by different nAChR subtypes exhibiting different
desensitization rates upon agonist stimulation
[242,259,260].
Recent significant progress in the field of the
molecular pathways underlying human cognition has
implicated the ERK ⁄ MAPK signaling cascades in
various cognitive mechanisms [261]. Neuronal-type
nAChRs mediate the Ca2+-dependent activation of
ERK ⁄ MAPK and CREB in several neuronal cell lines
[262–264]. In the hippocampus in particular, nicotineevoked neuronal-type nAChR stimulation causes
store-mediated Ca2+-influx, promoting activation
of Ca2+-calmodulin-dependent protein kinase and
ERK ⁄ MAPK and CREB phosphorylation, as shown
in vitro [265,266].
Ca2+ signaling and the ERK ⁄ MAPK cascades are
also involved in the neuronal-type nAChR-mediated
neuroprotection of hippocampal neurons in vitro
FEBS Journal 274 (2007) 3799–3845 ª 2007 The Authors Journal compilation ª 2007 FEBS
3819
Nicotinic acetylcholine receptors
D. Kalamida et al.
[267,268]. Neuronal-type nAChRs are generally
involved in neuroprotection ⁄ neurodegeneration. This
has been demonstrated both in animal models, in
which increased neurodegeneration is observed in aged
b2-subunit knockout mice [269], and in humans
suffering from AD, in whom a4b2 nAChRs are
markedly reduced in the brain [270] and a7 nAChRs
seem to interact with the amyloid plaque-component,
b-amyloid peptide [271], as discussed below. Interestingly, a recent study proposed an alternative
mechanism in which b-amyloid peptide induces the
pathology of AD by triggering a postsynaptic a7-mediated increase in N-methyl-d-aspartate receptor endocytosis [272]. Additional studies will be required to
unravel this novel mechanism possibly leading to
dementia.
Cholinergic mechanisms of reward and addiction
The ventral tegmental area (VTA) has been implicated
in the rewarding motivational effects of a wide variety
of addictive drugs, including nicotine [273,274]. There
are two major DAergic routes extending from the
VTA to the nucleus accumbens (mesolimbic pathway)
or the prefrontal cortex (mesocortical pathway).
Within the VTA, both DAergic and GABAergic neurons are implicated in signaling pathways of reward
[274], the latter providing inhibitory input to DAergic
neurons [275]. The early acute effects of nicotine
predominantly affect GABAergic neurons, where
4-aminobutyrate release is modulated by rapidly desensitizing neuronal-type nAChR subtypes [198,276]. In
fact only the a4 and b2 subunits are found on GABAergic neurons [277]. Hence, the stimulation and fast
subsequent desensitization of a4b2 nAChRs produce
prolonged stimulation of DAergic neurons by removal
of the inhibitory 4-aminobutyrate input (Fig. 8). Nicotine also acts on presynaptic neuronal-type nAChRs
located on VTA glutamatergic terminals which show a
lower rate of desensitization after nicotine exposure,
eliciting an excitatory glutamatergic input to the
DAergic neurons (Fig. 8) [276,278]. Indeed, several
studies have suggested that a7-containing nAChRs
mediate the presynaptic actions of nicotine in the
CNS, especially glutamate release [279–281]. Finally,
DAergic neurons express nAChR a4, a3, a5, a6, b2
and b3 subunits [277] (Fig. 8). The somatodendritic
nAChRs, in the form of various neuronal nAChR subunit combinations, on DAergic neurons of the VTA
can directly excite these neurons by receiving cholinergic signals ascending from outside the VTA [282].
This process eventually results in transient DA
responses that are terminated by nAChR desensitization [283–285].
3820
Fig. 8. Modulation of DA release in the VTA by nAChRs. Native
nAChRs modulate DA release in DAergic neurons of the mesolimbic and nigrostriatal pathways both directly and indirectly. nAChR
agonists exert direct modulation of DA release through presynaptic
and preterminal nAChRs. Alternatively, activation of a7 nAChRs on
glutamatergic terminals trigger release of Glu, which in turn stimulates ionotropic Glu receptors on DAergic terminals, finally inducing
DA release. Additionally, desensitization of the a4b2 nAChR on
GABAergic interneurons can remove the inhibitory 4-aminobutyrate
(GABA) input on DAergic neurons, thereby indirectly eliciting DA
release. Modified from Jensen et al. [234], with permission.
The role of neuronal-type nAChRs in non-neuronal
tissues and cells
Apart from the importance of neuronal-type nAChRs
in the nervous system, their expression has been demonstrated in ‘nonexcitable’ cells, namely lymphocytes,
monocytes, macrophages, dendritic cells, adipocytes,
keratinocytes, endothelial cells and epithelial cells of
the intestine and lung [286–289]. The complex role of
neuronal-type nAChRs in the periphery is reflected by
their ability to attenuate or enhance the progression of
several pathologies, e.g. attenuate ulcerative colitis and
enhance Crohn’s disease. In addition, different nAChR
types are responsible for different functions. For example, it has recently been shown in the human lung that,
although stationary cells express mainly a7 nAChRs
[290], migrating bronchial epithelial cells express
a3a5b2 receptors, indicating that they may be involved
in the wound-repair process [291], whereas in keratinocytes, both a3- and a7-containing receptors are present and are responsible, respectively, for mediating
keratinocyte chemokinesis and chemotaxis [292].
Most importantly, the key position of neuronal-type
nAChRs as the intermediate link between the involuntary nervous system and inflammation has stimulated
interest in the field of anti-inflammatory therapeutics.
The role of neuronal-type nAChRs in inflammation
and angiogenesis is discussed in detail below.
FEBS Journal 274 (2007) 3799–3845 ª 2007 The Authors Journal compilation ª 2007 FEBS
Nicotinic acetylcholine receptors
D. Kalamida et al.
The cholinergic anti-inflammatory pathway
The vagus nerve is a ‘parasympathetic’ nerve that
emanates from the cranium and innervates all major
organs, creating a neuronal network of peripheral surveillance. It branches into both sensory (input) and
motor (output) fibers, thus establishing a bidirectional
connection between the brain and the immune system.
Thus, once the sensory fibers of the vagus nerve are
activated by stimuli resulting from persistent inflammation, the brain reacts to this information by activating
the efferent fibers of the vagus nerve, which deliver
‘immuno-suppressive’ messages to the periphery.
Various findings suggested the existence of a cholinergic anti-inflammatory mechanism: upon stimulation, the vagus nerve reflexively releases ACh, and
ACh significantly reduces the release of pro-inflammatory cytokines in lipopolysaccharide-stimulated human
macrophage cultures through a-Bgtx-sensitive nAChRs
[293]. However, the identity of the specific nAChR
subtype on macrophages remained elusive until
recently. It is now well-documented that physiological
(vagus nerve-secreted ACh) or pharmacological (exposure to agonists, such as nicotine) stimulation of the
homopentameric a7 nAChR, present on the surface of
tissue macrophages, blocks the expression of the proinflammatory cytokines, tumor necrosis factor (TNF),
IL-1, IL-6 and IL-18, as well as the secretion of the
high-mobility group box 1 (HMGB1) protein [294–
296] (Fig. 9). Interestingly, the a7 nAChR controls
cytokine production at the post-transcriptional level,
without affecting levels of mRNAs for TNF, IL-1,
IL-6 and IL-18 [295]. The situation is different for
HMGB1, the constitutive intracellular expression of
which is crucial for the survival and normal transcriptional regulation of macrophages [297]. a7 nAChR
blocks the secretion, rather than the translation of
HMGB1, probably by inhibiting its translocation from
the nucleus to the cytoplasm [295]. In terms of the subcellular mechanisms underlying the cholinergic antiinflammatory machinery, several studies have provided
evidence for the a7-mediated inhibitory action of nicotine on the nuclear factor-jB (NF-jB) pathway, which
is crucial for macrophage activation and pro-inflammatory cytokine secretion [295,298,299]. The Jak2-STAT3
pathway is also involved, as shown both in vivo and
in vitro, because a7 nAChR stimulation after nicotine
binding induces phosphorylation of Jak2, which in
turn activates STAT3 [300].
Implication in angiogenesis
Nicotine induces the proliferation of endothelial cells
in vitro [301] and endothelial cells express functional
neuronal-type nAChRs [302], which are mainly
a7 nAChRs [290,303], but also a3-, a4-, b2- or b4-containing nAChRs [303]. The involvement of neuronaltype nAChR-mediated cholinergic pathways (mainly
a7) in endothelial cell growth and angiogenesis, including tumor angiogenesis, has been demonstrated
[303,304]. Formation of the cholinergically induced
endothelial network is partially dependent on vascular
endothelial growth factor and completely dependent
on the phosphatidylinositol 3-kinase and MAPK pathways, eventually resulting in NF-jB activation [303].
According to a recent study, nicotine abrogated the
apoptotic effect of several chemotherapeutic agents in
nonsmall cell lung carcinoma cell lines. Its action is
exerted through an a3-containing nAChR subtype and
requires the activation of an Akt pathway [305,306].
Expression of recombinant neuronal-type
nAChRs
The large number of nAChR subunits and the fact
that many subunits are usually expressed in a given
cell type complicate the study of their properties and
functions. A strategy to avoid this problem is the ectopic expression of recombinant receptor subunits or
combinations. Cells commonly used for the heterologous expression of neuronal-type nAChRs include
Xenopus oocytes, epithelial cells and neural crestderived cell lines. Expression of full-length a7 in the
human epithelial cell line SH-EP1, which are null for
nAChRs, showed that it retained the same pharmacological and functional properties with the native receptor, as evidenced by ligand binding and current
responses and can therefore be used for functional
studies [307,308]. However, some studies indicate that
not all cells can form functional a7 nAChR channels.
Studies on the ectopic expression of a7 in neural-crestderived rat cells (PC12 and GH3), mouse fibroblasts
(NIH 3T3) and insect ovary cells (Sf9) showed that
only the cell lines of neural-crest origin were able to
produce assembled receptors as indicated by 125I-labeled a-Bgtx binding, although all transfected cells
expressed the transgene as transcribed mRNA when
transfected [309]. In a similar study, a7 was expressed
in SH-EP1 and GH4C1 (rat pituitary) cells in varying
amounts but not in CHO, CV1 (monkey fibroblasts)
or SN56 (fusion of mouse septal neurons and a neuroblastoma line) cells, whereas a4 and b2 receptors
were produced in all five cell lines [310]. These findings
suggest that there are cell-specific factors regulating
receptor formation and maturation at the post-translational rather than the transcriptional level and these
should be taken into account when trying to ectopically express a nAChR.
FEBS Journal 274 (2007) 3799–3845 ª 2007 The Authors Journal compilation ª 2007 FEBS
3821
Nicotinic acetylcholine receptors
D. Kalamida et al.
Fig. 9. The ‘nicotinic anti-inflammatory pathway’. The vagus nerve can regulate inflammation through an a7 nAChR-mediated physiological
pathway. Homomeric a7 nAChRs are present on the surface of tissue macrophages. Upon physiological (vagus nerve-secreted ACh) or pharmacological (via exposure to agonist, such as nicotine) stimulation, the production of the pro-inflammatory cytokines TNF-a, IL-1, IL-6 and
IL-18 is inhibited and the inflammatory crisis controlled. Reproduced from Ulloa [296], with permission.
Xenopus oocytes have been used for the production
of truncated forms of a7, a4 and b2, consisting of their
ECDs with or without the M1 segment. a4 and b2
ECD coexpression resulted in detectable epibatidine
binding only in the presence of the M1, although both
forms of the proteins were expressed and glycosylated
[311]. By contrast, the a7 subunit ECDs had ligandbinding affinities and sedimentation velocities indicative of native-like receptor formation irrespective of the
presence of the M1 [312]. However, the M1 was
required for a production yield comparable to that of
the full-length receptor, probably due to limitations in
folding and assembly. This evidence suggests that, at
least for some neuronal-type subunits, expression of
the ECD together with the M1 segment may be
required to allow efficient protein formation.
The a4 and b2 subunits have also been expressed in
Xenopus oocytes as concatamers, in which the C-terminus of a4 was linked by a 18–36 amino acid linker
peptide to the N-terminus of b2, and vice versa [313].
3822
The resulting receptors were more efficient than
those formed by unlinked subunits. Taking this a
step further, Groot-Kormelink et al. [314] expressed
a3b4 receptors as pentameric constructs connected by
a flexible linker, resulting in the formation of receptors
with a predetermined stoichiometry and subunit
arrangement. This approach could prove valuable in
dissecting variations between receptors with minor
differences in subunit composition and organization.
Such systems are useful for the study of pharmacological and functional properties of nAChRs, but are
not suitable for the production of large quantities of
receptor protein, which would be useful in the elucidation of receptor structure. As previously mentioned,
most attempts at large-scale production have been
targeted at the muscle-type nAChR subunits, but
some have been carried out on neuronal-type nAChR
subunits. nAChRs are large integral membrane proteins and are therefore difficult to produce in large
quantities. In addition, their transmembrane hydro-
FEBS Journal 274 (2007) 3799–3845 ª 2007 The Authors Journal compilation ª 2007 FEBS
Nicotinic acetylcholine receptors
D. Kalamida et al.
phobic character makes structural studies very difficult. However, it may be possible to circumvent this
by expressing only the ECD of the receptor subunits,
because this contains all the elements of the ligandbinding site and folds independently of the rest of the
molecule, as shown by chimeric nAChR a7-5HT3
receptors [315]. The a7 ECD has been expressed in
E. coli as a fusion protein with maltose-binding
protein and glutathione S-transferase (GST) [316,317].
A downside was that the protein was found in insoluble inclusion bodies, which need to be solubilized
with urea or guanidine, but most of the resulting
refolded protein was in the form of high molecular
mass aggregates. However, when 0.1% SDS was
added during the refolding procedure aggregates were
greatly decreased [30]. In the same study, a mutant
a7 ECD is described with a C116S mutation, which
appeared to further decrease aggregate formation and
showed a slightly improved a-Bgtx binding affinity.
More recently, a double mutant form of the a7 ECD
was expressed in P. pastoris by our group. In this
mutant, in addition to the C116S, the a7 Cys loop
was exchanged for the Cys loop of the soluble acetylcholine-binding protein (AChBP) [50]. The resulting
protein had a greatly improved solubility as well as a
higher affinity for a-Bgtx compared with the wildtype a7 ECD expressed in P. pastoris or E. coli.
Further work has shown that mutation of additional
hydrophobic residues exposed on the surface of the
molecule, as identified from a model of the human a7
ECD we have constructed based on the recently
solved structures of the L. stagnalis AChBP and the
Torpedo muscle nAChR, offers extra solubility and
enhanced a-Bgtx binding affinity (M. Zouridakis,
P. Zisimopoulou, E. Eliopoulos, K. Poulas and S. J.
Tzartos, in preparation). Electron microscopy studies
(by N. Unwin, MRC, Cambridge, UK) showed that
the recombinant protein forms particles similar to the
expected ECD of the whole nAChR (Fig. 10). Currently, efforts are also being made by our group to
produce receptor subunits in higher eukaryotic systems, insect and mammalian expression systems, aiming towards a more physiological glycocylation and
maturation of the proteins.
nAChR-associated diseases
Muscle-type nAChR-associated diseases
The muscle-type nAChR is known to be the target in
several inherited and acquired diseases, most of which
lead to impaired neuromuscular transmission and muscle weakness. The acquired autoimmune disease MG,
Fig. 10. Electron microscopy image of individual human nAChR a7
ECD oligomers expressed in P. pastoris. Negative-stain electron
microscopic image of glycosylated human a7 ECD novel mutant
(M. Zouridakis, P. Zisimopoulou, E. Eliopoulos, K. Poulas and
S. J. Tzartos, manuscript in preparation). Arrowheads indicate an
end-on view of the ECD oligomers. Images were kindly taken by
N. Unwin.
is the most common and best studied of these and is
usually caused by autoantibodies to muscle nAChRs
[2,105,318,319]. The rather rare inherited conditions,
called congenital myasthenic syndromes (CMS), are
associated with several abnormalities affecting ACh
release, acetylcholinesterase activity, nAChR function
and ⁄ or nAChR number [320].
Myasthenia gravis
MG is a remarkably heterogeneous noninherited autoimmune disease, usually characterized by the presence
of circulating autoantibodies that bind to and destroy
muscle nAChRs. Its hallmarks are muscular weakness
and fatigability. It can either remain localized to a single muscle group (e.g. ocular MG) or can spread to
several skeletal muscles (generalized MG). Autoantibodies reduce the number of available nAChRs, causing a defect in neuromuscular transmission and muscle
weakness [319]. This weakness is prominent in muscles
that are used frequently and repetitively, for example,
the ocular, bulbar and facial muscles (resulting in ptosis and double vision), while other commonly affected
muscles include those of the limbs, neck and shoulders.
Although about 85% of MG patients have antibodies
against nAChR, 15% of patients with generalized
MG do not (seronegative MG) [321]. About 20–40%
FEBS Journal 274 (2007) 3799–3845 ª 2007 The Authors Journal compilation ª 2007 FEBS
3823
Nicotinic acetylcholine receptors
D. Kalamida et al.
of seronegative MG patients have serum antibodies
against the postsynaptic protein, MuSK [322]. Other
autoantibodies against striated muscle tissue proteins
(mainly titin and ryanodine receptors) have been found
in subgroups of MG patients [323]. Current medications control MG by improving neuromuscular transmission or down-modulating the immune system. They
include acetylcholinesterase inhibitors and steroidal or
nonsteroidal immunosuppressants, and, in severely
affected patients, administration of intravenous immunoglobulins or plasmapheresis. Surgical treatment consists of thymectomy. MG is the most common disease
affecting the NMJ, with a prevalence reported to be
higher than 70 per million [324].
The autoantibodies can reduce the number of functional nAChRs at NMJs by at least three mechanisms.
First, being bivalent molecules, antibodies can crosslink nAChRs in the muscle postsynaptic membrane,
thus stimulating internalization and degradation, leading to an overall loss of nAChRs, a process called
antigenic modulation. A second, and probably the
most important, mechanism is the complement-mediated focal lysis of the postsynaptic membrane. Antibodies bind to nAChR and trigger the complement
cascade, resulting in the focal destruction of the
postsynaptic membrane by the membrane attack complex. Third, antibodies against the nAChR-binding site
can directly inhibit receptor function [40,325].
The autoimmune character of MG was first demonstrated when myasthenic symptoms were observed in
rabbits immunized with nAChR [326] and, since that
time, the pathogenic role of nAChR antibodies in MG
patients has been established by several approaches
(e.g. detection of circulating antibodies against
nAChR, passive transfer of the disease from humans
to animals, localization of immune complexes, IgG
and complement on the postsynaptic membrane and
the beneficial effects of plasmapheresis). Novel therapeutic approaches to MG are now being explored in
animal models. Active immunization of animals with
purified nAChR from different species induces acute
or chronic experimental autoimmune MG (EAMG),
with symptoms starting about 10 days after immunization. This EAMG model is characterized by a high
antibody titer against nAChR, loss of more than half
of the nAChRs in muscles and the presence of antibodies bound to the remaining receptors in the postsynaptic membrane. The process is thought to be
mediated by both antibody and complement-mediated
focal lysis of the postsynaptic membrane. The pathological mechanisms that impair neuromuscular transmission in MG and chronic EAMG are similar [327].
Chronic EAMG provides a good model for testing
3824
new therapeutic approaches. However, this model does
not share the as yet unknown molecular and ⁄ or cellular mechanisms that sustain the autoimmune response
to nAChRs in humans with MG. Alternatively,
EAMG can be passively transferred to animals by
injection of serum from MG patients or animals with
EAMG or of anti-MIR mAbs [328,329].
More than half of the antibodies against nAChR in
MG patients and animals with EAMG are directed
against the MIR [40]. The MIR was originally defined
by the ability of mAbs against the nAChR to inhibit
the binding to the nAChR of other antibodies or
mAbs against nAChR. It is now established that the
MIR is not a single epitope, but rather a cluster of
overlapping conformation-dependent epitopes, with
residues 67–76 of the a1 subunit forming the core of
the region. This segment is on the extreme synaptic
end of each of the two a1 subunits. These extreme
ends of the a subunits are the most accessible parts of
the nAChR (Fig. 2) and this accessibility may be relevant to the high immunogenicity of the MIR [14,22].
mAbs against the MIR exhibit almost all of the pathological properties of serum autoantibodies in MG [40].
The mechanisms that induce and sustain the antibody-mediated autoimmune response to the muscle
nAChR are not clear and may differ in various forms
of the disease. MG onset requires a genetically favorable background combined with a number of environmental stimuli. In most MG patients, the
immunogen is likely to be native nAChR, because the
spectrum of autoantibody specificities is very similar
to that seen in animals immunized with native muscle
nAChR and different from that of animals immunized with denatured muscle nAChR [105]. The fetal
form may be the immunogen in some MG cases,
because MG sera often react with epitopes unique to
this subtype [330].
Because of the frequent thymic abnormalities found
in MG patients and the probable clinical improvement
after thymectomy, it is believed that the thymus is
implicated to the onset and ⁄ or maintenance of MG. In
the thymus, both single nAChR subunits and whole
nAChR molecules are normally expressed by thymic
epithelial cells and myoid cells [331]. It has been proposed that an inflammatory response in the hyperplastic thymus modulates the expression of nAChR by
thymic epithelial cells and myoid cells [332] and that
this nAChR expression in the hyperplastic thymic
environment, combined with a genetic predisposition,
could be sufficient to prime nAChR-responsive CD4+
T cells, leading to autoimmunity [333]. Supporting
evidence was provided by Poea-Guyon et al. [334],
who demonstrated that a large number of IFN-c- and
FEBS Journal 274 (2007) 3799–3845 ª 2007 The Authors Journal compilation ª 2007 FEBS
Nicotinic acetylcholine receptors
D. Kalamida et al.
TNF-a-regulated genes are highly expressed in the
myasthenic thymus. Moreover, a recently proposed
hypothesis by Bernasconi and colleagues [335] tries to
correlate innate immunity and MG as a result of the
increased levels of Toll-like receptor 4 (TLR4) mRNA
seen in patients with thymitis, compared with patients
with thymomas.
Thymomas are thymic epithelial tumors frequently
associated with paraneoplastic autoimmunity. Although
no whole nAChR molecules are found in thymomas,
single nAChR subunits have been detected. However, it
is uncertain whether thymomas actively sensitize, or
simply fail to induce tolerance in, newly produced
thymocytes autoreactive with the nAChR [336–338].
The understanding of the mechanisms underlying
MG has led to the design of novel therapeutic strategies. Tolerance induction (oral or nasal) [339], T-cell
receptor vaccination [340], protective antibody fragments against nAChR [340–344] and phosphodiesterase
inhibitors [345] are some of the experimental approaches under investigation. Our group, having successfully
expressed the ECDs of the human a1, b1, c and e subunits in P. pastoris in water-soluble form, is developing
a specific antibody against subunit apheresis method
[48,75]. The immobilization of each ECD on an insoluble carrier (CNBr–Sepharose) provides ‘immunoadsorbent’ columns that can be used for the ex vivo
elimination of patients’ antibodies against nAChR. In
addition, ECD-immunoadsorbents have allowed the
isolation of subunit-specific autoantibodies from the
sera of MG patients and the study of their in vitro (cell
cultures) and in vivo (EAMG) pathogenicity. To date,
autoantibodies to the a1 and b1 subunits have been isolated and their potency has been compared with those
of the untreated and antibody-depleted sera (unpublished data). The anti-a1 autoantibodies were found to
be much more potent than the anti-b1 autoantibodies
in terms of their modulating and pathogenic activity,
and the depleted sera showed a dramatically reduced
activity compared with the untreated sera (unpublished
data), strongly suggesting that the antibody against
nAChR is the main ⁄ sole pathogenic factor in the MG
sera. The elucidation of the role of autoantibodies to
each single nAChR subunit will shed light on the pathogenic mechanisms of MG and should lead to novel
therapeutic approaches.
Congenital myasthenic syndromes
Congenital myasthenic syndromes (CMSs) are a highly
heterogeneous group of inherited disorders, characterized by defective neuromuscular transmission, resulting
in muscle fatigue. Following the identification of muta-
tions in nAChR subunits, other genes encoding presynaptic, synaptic or postsynaptic proteins were also
identified as candidate genes for CMSs [320,346–348].
Engel [348,349] initially classified these syndromes
according to the lesion site (postsynaptic, synaptic or
presynaptic) and pathophysiology. The classification of
CMSs is still tentative, as further studies, including
mutation analysis and chromosome studies, are likely
to provide further information. To date, the majority
of these disorders present abnormalities in postsynaptic
function at the NMJ.
The first myasthenic symptoms of CMSs occur early
in life, usually in the first two years after birth. In
some rare cases, onset occurs in the second to third
decade of life. The severity and course of CMSs are
highly variable, ranging from minor symptoms to progressive disabling weakness and even death. Clinical
diagnosis of CMSs is often possible on the basis of
myasthenic symptoms involving fatigable weakness of
the ocular, bulbar or limb muscles present since birth
or early childhood [350]. On electromyography, a
decremental compound action potential on repetitive
low-frequency stimulation of the motor nerve and a
negative test for antibodies against nAChR and calcium channel can help distinguish CMSs from MG
and Lambert–Eaton syndrome.
To date, 60 mutations in genes encoding the different nAChR subunits (a1, b1, e and d) have been
reported, and affect the ECDs, transmembrane segments M1–M3, and the cytoplasmic domain between
M3 and M4 [350]. Despite the diversity of these mutations, we can group them into two major categories:
kinetic and low-expressor mutations. Kinetic mutations
fall into two subclasses according to whether they
increase the response to ACh (as in slow-channel syndromes) or decrease the response to ACh (as in fastchannel syndromes).
Slow- and fast-channel syndromes
The main characteristics of these syndromes are kinetic
abnormalities of nAChR function. The term ‘slowchannel syndrome’ originates from the abnormally
slow decay of the synaptic currents caused by abnormally prolonged opening of the nAChR channel. As a
consequence, the postsynaptic region is overloaded
with cations, which causes endplate myopathy, loss of
nAChR from the folds, widening of the synaptic space,
vacuolar change and apoptosis. In most cases, slowchannel syndromes appear early in life and cause
severe disability by the end of the first decade [351].
Slow-channel syndromes are caused by dominant
gain-of-function mutations. At least 18 slow-channel
mutations have been reported [351–353]. Croxen and
FEBS Journal 274 (2007) 3799–3845 ª 2007 The Authors Journal compilation ª 2007 FEBS
3825
Nicotinic acetylcholine receptors
D. Kalamida et al.
colleagues [354] also recently described a recessive
loss-of-function mutation resulting in a slow-channel
syndrome. Although the majority of slow-channel syndromes result from mutations in M2 of the a1, b1, d
and e subunits, some are caused by mutations in other
functional domains of the subunits. Of particular interest are the aG153S mutation (near the ACh-binding
site) and the aN217K mutation (in the N-terminal
region of M1), which act mainly by increasing the
affinity for ACh [353].
The term ‘fast-channel syndrome’ originates from
the abnormally fast decay of the synaptic response,
caused by abnormally brief channel opening due to
decreased affinity for ACh, decreased gating efficiency or a decreased number of openings of the
channel upon acetylcholine binding ⁄ occupancy [355].
The majority of fast-channel syndromes are caused by
recessive loss-of-function mutations; however, a single
missense mutation in the nAChR a1 subunit gene,
causing replacement of Phe256 in M2 with leucine, has
a dominant-negative effect [356]. The mutations are
located in different functional domains of the a1, b1
and d subunits. Mutations in the ECDs decrease the
affinity for ACh, those in transmembrane sections
impair gating efficiency and those in the long cytoplasmic loop of the e subunit destabilize channel kinetics.
Usually, the mutated allele causing the kinetic abnormality is accompanied by a null mutation in the second allele. At least 13 fast-channel mutations have
been identified [320,347].
The clinical features of fast-channel syndromes
resemble those of MG, but tend to be milder when they
affect gating efficiency [357], moderately severe when
channel kinetics are impaired [358] and severe when the
affinity for ACh is affected [359,360]. In most fast-channel syndromes, therapy relies on combined treatment
with 3,4-diaminopyridine and cholinesterase inhibitors.
nAChR deficiency with or without kinetic abnormalities
In patients with CMS, several homozygous or heterozygous recessive mutations in the nAChR subunits are
found that result in a reduced number of functional
nAChRs at the postsynaptic membrane. These lowexpressor and null mutations have been reported in all
subunits of the adult nAChR, but are more frequent
in the e subunit, particularly its long cytoplasmic
M3–M4 linker. Patients with mutations in the
e subunit have milder symptoms than those with mutations in other subunits.
More than 50 e subunit mutations have been reported. Some of these cause premature termination of
translation by producing a nonsense or splice site [361]
or frame-shift mutations [352,362,363]. In addition,
3826
some missense mutations alter residues essential for
assembly (glycosylation sites, cysteine loop) or occur in
the signal peptide, resulting in reduced gene expression.
Point mutations of regulatory elements (N-box) or the
promoter region of the e subunit gene also result in low
e subunit gene expression [364,365]. Of particular interest is the 1369delG mutation, which results in the loss
of the C-terminal cysteine, Cys470, crucial to both the
maturation and surface expression of the adult receptor
[366]. In addition, the frame-shifting e1267delG mutation, occurring in Romany populations, results in
nAChR deficiency at the endplate [362,363,367]. The
prevalence of this mutation appears to be high due to a
founder effect in the Romany population [368].
In nAChR deficiency, most patients respond quite
well to anticholinesterase drugs, while others derive
additional benefit from 3, 4-diaminopyridine [369].
nAChR deficiency caused by rapsyn mutations
Rapsyn is a 43 kDa postsynaptic protein that, together
with agrin and MuSK, plays an essential role in
nAChR clustering at the postsynaptic membrane.
Mutations in rapsyn were discovered in patients diagnosed with CMS who showed endplate deficiency without any mutation in any nAChR subunit [370].
Endplate studies in these patients revealed decreased
staining for rapsyn and nAChR, as well as impaired
postsynaptic development.
Twenty-one rapsyn mutations have been identified
[320,347] in the coding (missense, frame-shift, stop and
splice site mutations) and promoter regions. The missense mutation, N88K, has been identified in all
patients with mutation in the RAPSN gene and can be
either homozygous or heterozygous. Other mutations
in the coding region result in mutations in different
domains of the protein. Patients carrying rapsyn mutations can have mild or severe symptoms. Most respond
moderately well to anticholinesterase drugs, and some
derive additional benefit from 3,4-diaminopyridine.
Mutations in the MuSK gene
MuSK plays a crucial role in the agrin–MuSK–rapsyn
pathway by organizing the postsynaptic scaffold and
nAChR aggregation. Recently, two heteroallelic mutations in the MuSK gene, a frame-shift mutation
(c.220insC) and a missense mutation (V790M), were
identified in a single individual with a CMS phenotype.
The frame-shift mutation resulted in absence of MuSK
expression. The missense mutation did not affect
MuSK catalytic activity, but reduced the expression
and stability of MuSK, leading to decreased agrindependent nAChR aggregation [371]. As described
above (in the MG section), MuSK also plays an
FEBS Journal 274 (2007) 3799–3845 ª 2007 The Authors Journal compilation ª 2007 FEBS
Nicotinic acetylcholine receptors
D. Kalamida et al.
important role in autoimmune MG, in which 20–40%
of anti-nAChR-seronegative MG patients have antibodies against MuSK, which are believed to be the
pathogenic factor in these patients [322].
Neuronal-type nAChR-associated diseases
In contrast to the muscle-type nAChR, the pathophysiological functions of neuronal-type nAChRs are not well
defined. It is well documented that brain nAChRs participate in complex functions, such as attention, memory
and cognition, and clinical data suggest their involvement in the pathogenesis of several disorders (AD and
PD, schizophrenia, depression, etc.).
Alzheimer’s disease
AD, the most common cause of dementia, is characterized by a progressive decline in cognitive function, particularly affecting memory, attention and orientation,
whereas motor and sensory abilities are usually undisturbed. It appears that many biochemical events crucial
for neuronal communication and synaptic plasticity fail
during the course of the disease [372]. One prominent
hallmark of AD is an early and pronounced loss of
cholinergic function (the ‘cholinergic hypothesis’) [373].
Muscarinic AChRs have attracted most attention
because nAChRs are not as abundant as mAChRs in
the brain, and the contribution of deficits in nicotinergic transmission in AD has not received as much attention as deficits in signal transduction in the muscarinic
system. Nevertheless, the nicotinic system has been
shown to modulate attentional processes and is
involved in facilitation of memory. It is thus very likely
that deficits in nicotinic signaling are also involved in
the behavioural and cognitive deficits seen in AD.
A number of studies have reported reduced numbers
of central nAChRs in the aged and in AD patients. It
has been speculated that this reduction might be
caused by a preferential presynaptic location on
degenerating projection neurons [374]. Stimulation of
nAChRs (especially a7 subtype) and treatment with
nicotinic agonists are proved to protect neurons [375].
Because the deposition of brain amyloid plays a role
in the neurodegeneration associated with AD, the relationship between amyloid deposition and cholinergic
neuron activity is of great interest. Nicotine has been
shown to inhibit the development of cellular toxicity
induced by beta-amyloid peptides. Wang et al. [376]
showed that both the 42-amino acid beta-amyloid
peptide, Abeta(1–42), the predominant beta-amyloid
peptide species in amyloid plaques, and the a7 nAChR
are present in neuritic plaques and colocalize in
individual cortical neurons. Using extracts of human
brain tissue and cells that overexpress either a7 nAChR
or amyloid precursor protein, Abeta(1–42) and
a7 nAChR were coimmunoprecipitated by antibodies,
suggesting that they are tightly associated. Abeta(1–42)
and a7 nAChR bind with high affinity, and this interaction can be inhibited by a7 nAChR ligands. However, Lamb and co-workers [377] expressed various
nAChR subtypes in Xenopus oocytes (e.g. a4b2, a2b2,
a4a5b2 and a7) and showed that Abeta(1–42) blocks
the binding of ligands to various non-a7 nAChRs, but
not to a7 nAChRs. The block by Abeta(1–42) was
dependent on the subunit makeup and stoichiometry of
these receptors.
In addition, there is evidence that smokers have a
reduced risk of developing AD [378] and preventive
measures (including stimulants of nicotinic receptors)
[379] have been suggested in the treatment of AD. To
date, the acetylcholinesterase inhibitors have been the
most widely used anti-AD drugs and have been partially successful in slowing loss of cognition [375].
Schizophrenia
Schizophrenia is a chronic psychotic complex disorder
with a strong genetic predisposition. The exact molecular cause of any type of schizophrenia is still unknown.
The incidence of smoking in schizophrenic patients is
extremely high (80–90% versus 25–30% in the general
population) [379]. Studies have shown that tobacco use
transiently restores the schizophrenic patient’s cognitive
and sensory deficits, and cessation of smoking exacerbates the disease symptoms [380]. The a7 nAChR seems
to participate directly in the pathophysiology of schizophrenic disturbances. Leonard et al. [381] reported a
higher prevalence of functional promoter mutations in
a7 nAChRs in schizophrenic subjects than in controls.
Post-mortem binding studies have revealed a disturbance of nicotinic receptor expression affecting the a7
and a4b2 subunits in various cerebral areas [382].
Attention-deficit hyperactivity disorder
Attention-deficit hyperactivity disorder (ADHD) is
a disease characterized by a persistent pattern of
inattention and distractability and ⁄ or hyperactivity ⁄
impulsivity to such a degree that it impairs academic
or occupational functioning. ADHD sufferers typically
show evidence of this disorder in their childhood and
50% continue to demonstrate clinically significant
symptoms into adulthood [383]. The incidence of cigarette smoking in individuals with ADHD has been
found to be higher ( 40%) than in the general adult
FEBS Journal 274 (2007) 3799–3845 ª 2007 The Authors Journal compilation ª 2007 FEBS
3827
Nicotinic acetylcholine receptors
D. Kalamida et al.
population (25–30%) [384]. Animal studies have shown
that nAChR-related mechanisms are involved in attentional function [385]. The a4b2 and a7 nAChRs are
critical for attention and working memory in rats
[386,387]. Nicotine skin patches and a number of
nAChR agonists improve clinical ADHD symptoms
[388]. Wilens et al. [389] reported that the novel nicotinic cholinergic agent, ABT-418, a nicotinic agonist
with selectivity for the a4b2 nAChR, may reduce impulsivity, hyperactivity and attentional deficits in
adults with ADHD. Ueno et al. [390] used animal
models of ADHD and showed that nicotine improves
attention and memory in rats through activation of
a4b2, but not a7, nAChRs.
Parkinson’s disease
PD is a neurodegenerative movement disorder. In addition to the well-established loss of DAergic neurons,
several studies have shown that nAChRs play a critical
role in PD [391]. Epidemiological studies have shown
that smoking protects against PD [392]. A number of
a4b2 nAChR agonists have shown beneficial effects in
PD [393]. Xie et al. [394] have shown that the neuroprotective effect of nicotine in PD is receptor independent
and is due to its interaction with the mitochondrial
respiratory chain and its antioxidant effects.
Pemphigus vulgaris
Pemphigus vulgaris is an autoimmune disease of keratinocytes in which the cells of the epidermis loose
adherence (acantholysis), resulting in blistering of the
skin or oral mucosa. An autoimmune response to
a9 nAChRs has been identified [395]. Many neuronaltype nAChR subunits have been found at low levels in
keratinocytes, where their responses to endogenous
ACh and to nicotine influence cell adherence and
motility in vivo and may influence development, wound
healing and wrinkling in vivo [396]. Pemphigus patients
occasionally develop MG and ⁄ or thymoma [397].
Autoimmune autonomic neuropathy
Autonomic neuropathies are inherited or acquired neuropathies in which the autonomic nerve fibers, both
sympathetic and parasympathetic, are affected. The
neuropathies can be autoimmune, idiopathic or due to
diabetes, amyloidosis, drugs, etc. Autoimmune autonomic neuropathy (AAN) is sometimes associated with
a neoplasm and the patients have high titres of antibodies to ganglionic nicotinic nAChRs [398]. Autoantibodies to a3 nAChRs have been found in 40% of
3828
patients with idiopathic or paraneoplastic dysautonomia [399,400]. An animal model of AAN has been
developed and has shown the involvement of
a3 nAChR in the pathogenesis of the disease. Rabbits
immunized with the recombinant ECD of the human
nAChR a3 subunit produced antibodies against
nAChR and developed signs of experimental AAN
[401]. The same group soon demonstrated that experimental AAN is an antibody-mediated disorder by
documenting sympathetic, parasympathetic and enteric
autonomic dysfunction in mice injected with rabbit
IgG containing ganglionic nAChR antibodies. The
autonomic signs were associated with reversible failure
of nicotinic cholinergic synaptic transmission in the
superior mesenteric ganglia. In addition, mice injected
with IgG from two patients with AAN demonstrated a
milder phenotype, with evidence of urinary retention
and gastrointestinal dysmotility [402].
Hereditary epilepsies
Specific genes coding for ligand- and voltage-gated ion
channels that are associated with hereditary epileptic
phenotypes have been identified. Some rare idiopathic
epilepsies are associated with mutations in genes coding for different neuronal-type nAChR subunits. Most
mutations found to date are in the a4 subunit, the
most abundant subunit in the CNS. Specifically, the
identification of mutations in the a4 subunit in patients
with human benign familial neonatal convulsions or
autosomal dominant nocturnal frontal lobe epilepsy
raise the possibility that the observed gene defects are
causally linked with these two diseases or, alternatively, that a4 nAChR mutants increase the probability
of epileptic discharges [403].
Autism
Autism is a developmental disorder associated with
structural abnormalities of the brain. Cerebellar
abnormalities have been identified by neuroimaging
or neuropathology. The cholinergic neurotransmitter
system has been implicated on the basis of nAChR
loss in the cerebral cortex. Numbers of the a3 and
a4b2 nAChRs have been found to be significantly
reduced in autistic subjects compared with controls
[404]. An increase in a7 subunits has also been
observed [405].
Smoking addiction
Smoking is a major public health problem and the
a4b2 and a7 subtypes of nAChRs, which are the
FEBS Journal 274 (2007) 3799–3845 ª 2007 The Authors Journal compilation ª 2007 FEBS
Nicotinic acetylcholine receptors
D. Kalamida et al.
most abundant subtypes in the brain, are closely associated with nicotine addiction and nicotine-induced
behaviors [406]. a4b2 nAChRs have the highest sensitivity to nicotine and repeated nicotine exposure
increases the functional nicotinic receptors in the
brain. Functional upregulation of a4b2 nAChRs,
observed in the brains of both smokers and animals
chronically exposed to nicotine, is combined with a
sensitization of the mesolimbic dopamine response to
nicotine [407]. This response appears to be associated
with the overall addictive properties of nicotine (but
also of other drugs of abuse). The a7 nAChRs are
also overexpressed in small cell lung carcinoma of
smokers [408]; in this case, in vitro experiments have
suggested that the malignant growth can be ceased
using a-neurotoxins or a-conotoxins, by blocking
these receptors [409]. Nicotine itself, in a number of
commercially available forms (nicotine gum, transdermal patch, nicotine nasal spray, nicotine inhaler, etc.)
is effective as part of a strategy to promote smoking
cessation. However, compounds that could act as
a4b2 agonists offer a more promising approach.
Existing treatments have demonstrated only moderate
efficacy in assisting smokers to quit. Varenicline,
recently approved by the US FDA as an aid to smoking cessation treatment, has a novel mechanism of
action, targeting a4b2 nAChR [410]. It has both
agonistic and antagonistic properties that together are
believed to account for the reduction of craving and
withdrawal as well as blocking the rewarding effects
of smoking. Its targeted mechanism of action, superior efficacy and excellent tolerability make varenicline
a welcome and useful addition to the therapeutic
options for smoking cessation.
Future perspectives
For several decades, the nAChR has served as the prototypic molecule for neurotransmitter receptors. In this
review, we discuss its localization, structure, function
and pathogenicity, topics which have been investigated,
and many clarified, in the last half-century. Although
an enormous amount of data has been accumulated,
several fundamental questions remain unanswered.
These include the exact channel function, the exact
role of the different nAChR subtypes in different locations, the identification of the ‘key’ nAChR subtypes
in the various diseases in which they are involved, the
discovery or design of subtype-specific ligands, and the
atomic structure of the whole molecule. The solution
of the 3D structure (especially that of human neuronal
nAChR subtypes) is a major challenge, which is,
however, necessary for the development of novel
therapeutics. The efforts currently being invested make
us optimistic that considerable progress in this field
will be made in the near future. The knowledge
obtained will permit in-depth understanding of the
mechanisms of channel activation and function and
the design of selective ligands for therapeutic purposes.
Acknowledgements
Original studies in the authors’ laboratories described
in this review have been supported by grants from the
European Commission, the MDA of USA and the
Greek GSRT.
References
1 Changeux J & Edelstein SJ (2001) Allosteric mechanisms in normal and pathological nicotinic acetylcholine receptors. Curr Opin Neurobiol 11, 369–
377.
2 Lindstrom JM (1997) Nicotinic acetylcholine receptors
in health and disease. Mol Neurobiol 15, 193–222.
3 Wess J (1996) Molecular biology of muscarinic acetylcholine receptors. Crit Rev Neurobiol 10, 69–99.
4 Ishii M & Kurachi Y (2006) Muscarinic acetylcholine
receptors. Curr Pharm Des 12, 3573–3581.
5 Changeux JP, Bertrand D, Corringer PJ, Dehaene S,
Edelstein S, Léna C, Le Novère N, Marubio L, Picciotto M & Zoli M (1998) Brain nicotinic receptors:
structure and regulation, role in learning and reinforcement. Brain Res Rev 26, 198–216.
6 Karlin A & Akabas M (1996) Toward a structural
basis for the function of the nicotinic acetylcholine
receptors and their cousins. Neuron 15, 1231–1244.
7 Unwin N (1995) Acetylcholine receptor channel images
in the open state. Nature 373, 37–43.
8 Noda M, Ikeda T, Suzuki H, Takeshima H, Takahashi
T, Kuno M & Numa S (1986) Expression of functional
sodium channels from cloned cDNA. Nature 322, 826–
828.
9 Miledi R, Molinoff P & Potter LT (1971) Isolation of
the cholinergic receptor protein of Torpedo electric tissue. Nature 229, 554–557.
10 Kasai M, Changeux JP & Monnerie L (1969) In vitro
interaction of 1-anilino 8 naphthalene sulfonate with
excitable membranes isolated from the electric organ
of Electrophorus electricus. Biochem Biophys Res
Commun 36, 420–427.
11 Dennis M, Giraudat J, Kotzyba-Hibert F, Goeldner
M, Hirth C, Chang JY, Lazure C, Chretien M &
Changeux JP (1988) Amino acids of the Torpedo marmorata acetylcholine receptor subunit labeled by a
photoaffinity ligand for the acetylcholine binding site.
Biochemistry 27, 2346–2357.
FEBS Journal 274 (2007) 3799–3845 ª 2007 The Authors Journal compilation ª 2007 FEBS
3829
Nicotinic acetylcholine receptors
D. Kalamida et al.
12 Galzi JL & Changeux JP (1995) Neuronal nicotinic
receptors: molecular organization and regulations.
Neuropharmacology 34, 563–582.
13 Huganir RL & Greengard P (1990) Regulation of neurotransmitter receptor desensitization by protein phosphorylation. Neuron 5, 555–567.
14 Brejc K, van Dijk WJ, Klaasen RV, Schuurmans M,
van de Roost J, Smit AB & Sixma TK (2001) Crystal
structure of an ACh-binding protein reveals the ligandbinding domain of nicotinic receptors. Nature 411,
269–276.
15 Smit AB, Syed NI, Schaap D, van Minnen J, Klumperman J, Kits KS, Lodder H, van der Schors RC,
van Elk R, Sorgedrager B et al. (2001) A glia-derived
acetylcholine-binding protein that modulates synaptic
transmission. Nature 411, 261–268.
16 Celie P, van Rossum-Fikkert SE, van Dijk JW,
Brejc K & Smit AB (2004) Nicotine and carbamylcholine binding to nicotinic acetylcholine receptors as
studied in AChBP crystal structures. Neuron 41,
907–914.
17 Celie PH, Kasheverov IE, Mordvintsev DY, Hogg
RC, van Nierop P, van Elk R, van Rossum-Fikkert
SE, Zhmak MN, Bertrand D, Tsetlin V et al. (2005)
Crystal structure of nicotinic acetylcholine receptor
homolog AChBP in complex with an alphaconotoxin PnIA variant. Nat Struct Mol Biol 12,
582–588.
18 Ulens C, Hogg R, Bertrand D, Tsetlin V, Smit A &
Sixma T (2006) Structural determinants of selective
alpha-conotoxin binding to a nicotinic acetylcholine
receptor homolog AChBP. Proc Natl Acad Sci USA
103, 3615–3620.
19 Tasneem A, Iyer L, Jakobsson E & Aravind L (2004)
Identification of the prokaryotic ligand-gated ion channels and their implications for the mechanisms and origins of animal Cys-loop ion channels. Genome Biol 6,
R4.
20 Bocquet N, Prado de Carvalho L, Cartaud J, Neyton
J, Le Poupon C, Taly A, Grutter T, Changeux JP &
Corringer PJ (2007) A prokaryotic proton-gated ion
channel from the nicotinic acetylcholine receptor family. Nature 445, 116–119.
21 Unwin N (1993) Nicotinic acetylcholine receptor at 9
Å resolution. J Mol Biol 229, 1101–1124.
22 Beroukhim R & Unwin N (1995) Three-dimensional
location of the main immunogenic region of the acetylcholine receptor. Neuron 15, 323–331.
23 Miyazawa A, Fujiyoshi Y, Stowell M & Unwin N
(1999) Nicotinic acetylcholine receptor at 4.6 Å resolution: transverse tunnels in the channel wall. J Mol Biol
288, 765–786.
24 Miyazawa A, Fujiyoshi Y & Unwin N (2003) Structure
and gating mechanism of the acetylcholine receptor
pore. Nature 424, 949–955.
3830
25 West AP, Bjorkman PJ, Dougherty DA & Lester
HA (1997) Expression and circular dichroism of the
extracellular domain of the alpha subunit of the
nicotinic acetylcholine receptor. J Biol Chem 272,
25468–25473.
26 Schrattenholz A, Pfeiffer S, Pejovic V, Rudolph R,
Godovac-Zimmermann J & Maelicke A (1998) Expression and renaturation of the N-terminal extracellular
domain of Torpedo nicotinic acetylcholine receptor
alpha subunit. J Biol Chem 273, 32393–32399.
27 Alexeev T, Krivoshein A, Shevalier A, Kudelina I, Telyakova O, Vincent A, Utkin Y, Hucho F & Tsetlin V
(1999) Physicochemical and immunological studies of
the N-terminal domain of the Torpedo acetylcholine
receptor a-subunit expressed in Escherichia coli. Eur J
Biochem 259, 310–319.
28 Grant MA, Gentile LN, Shi QL, Pellegrini M & Hawrot E (1999) Expression and spectroscopic analysis of
soluble nicotinic acetylcholine receptor fragments
derived from the extracellular domain of the a-subunit.
Biochemistry 38, 10730–10742.
29 Yao Y, Wang J, Virronchatapan N, Samson A, Chill
J, Rothe E, Anglister J & Wang ZZ (2002) Yeast
expression and NMR analysis of the extracellular
domain of muscle nicotinic acetylcholine receptor a
subunit. J Biol Chem 277, 12613–12621.
30 Tsetlin VI, Dergousova NI, Azeeva EA, Kryukova
EV, Kudelina IA, Shibanova ED, Kasheverov IE &
Methfessel C (2002) Refolding of the Escherichia coli
expressed extracellular domain of alpha 7 nicotinic
acetylcholine receptor. Eur J Biochem 269, 2801–
2809.
31 Hertling-Jaweed S, Bandinin G, Muller-Fahrow A,
Dommes V & Hucho F (1988) Rapid preparation of
the nicotinic acetylcholine receptor for crystallization
in detergent solution. FEBS Lett 241, 29–32.
32 Paas Y, Cartaud J, Recouvreur M, Grailhe R, Dufresne V, Pebay-Peyroyla E, Landau EM & Changeux
JP (2003) Electron microscopic evidence for nucleation
and growth of 3D acetylcholine receptor microcrystals
in structured lipid–detergent matrices. Proc Natl Acad
Sci USA 100, 11309–11314.
33 Unwin N (2005) Refined structure of the nicotinic
acetylcholine receptor at 4 Å resolution. J Mol Biol
346, 967–989.
34 Corringer PJ, Le Novere N & Changeaux JP (2000)
Nicotinic receptors at the amino acid level. Annu Rev
Pharmacol Toxicol 40, 431–458.
35 Arias HR (2000) Localization of agonist and competitive antagonist binding sites on nicotinic acetylcholine
receptors. Neurochem Int 36, 595–645.
36 Galzi JL, Bertrand D, Devillers-Thiery A, Revah F,
Bertrand S & Changeux JP (1991) Functional significance of aromatic amino acids from three peptide
loops of the a7 neuronal nicotinic receptors site
FEBS Journal 274 (2007) 3799–3845 ª 2007 The Authors Journal compilation ª 2007 FEBS
Nicotinic acetylcholine receptors
D. Kalamida et al.
37
38
39
40
41
42
43
44
45
46
47
investigated by site directed mutagenesis. FEBS Lett
294, 198–202.
Barkas T, Mauron A, Roth B, Alliod C, Tzartos SJ &
Ballivet M (1987) Mapping the main immunogenic
region and toxin binding site of the nicotinic acetylcholine receptor. Science 235, 77–80.
Tzartos SJ, Kokla A, Walgrave SL & Conti-Tronconi
BM (1988) Localization of the main immunogenic
region of human muscle acetylcholine receptor to residues 67–76 of the alpha subunit. Proc Natl Acad Sci
USA 85, 2899–2903.
Tzartos SJ, Cung MT, Demange P, Loutrari H,
Mamalaki A, Marraud M, Papadouli I, Sakarellos C
& Tsikaris V (1991) The main immunogenic region
(MIR) of the nicotinic acetylcholine receptor and the
anti-MIR antibodies. Mol Neurobiol 5, 1–29.
Tzartos SJ, Barkas T, Cung M, Mamalaki A, Marraud
M, Papanastasiou D, Sakarellos C, Sakarellos-Daitsiotis M, Tsantili P & Tsikaris V (1998) Anatomy of the
antigenic structure of a large membrane autoantigen,
the muscle-type nicotinic acetylcholine receptor.
Immunol Rev 163, 89–120.
Wang F, Gerzanich V, Wells GB, Anand R, Peng X,
Keyser K & Lindstrom J (1996) Assembly of human
neuronal nicotinic receptor alpha5 subunits with
alpha3, beta2, and beta4 subunits. J Biol Chem 271,
17656–17665.
Wang F, Nelson M, Kuryatov A, Keyser K & Lindstrom J (1998) Chronic nicotine treatment up-regulates
human a3 b2 but not a3 b4 acetylcholine receptors
stably transfected in human embryonic kidney cells.
J Biol Chem 273, 28721–28732.
Sugiyama N, Boyd AE & Taylor P (1996) Anionic
residue in the alpha-subunit of the nicotinic acetylcholine receptor contributing to subunit assembly and
ligand binding. J Biol Chem 271, 26575–26581.
Kao PN & Karlin A (1986) Acetylcholine receptor
binding site contains a disulfide cross-link between
adjacent half-cystinyl residues. J Biol Chem 261, 8085–
8088.
Galzi JL, Revah F, Black D, Goeldner M, Hirth C &
Changeux JP (1990) Identification of a novel amino
acid alpha-tyrosine93 within the cholinergic ligandsbinding sites of the acetylcholine receptor by photoaffinity labeling. Additional evidence for a three-loop
model of the cholinergic ligands-binding sites. J Biol
Chem 265, 10430–10437.
Fu DX & Sine SM (1994) Competitive antagonists
bridge the alpha–gamma subunit interface of the acetylcholine receptor through quaternary ammonium–aromatic interactions. J Biol Chem 269, 26152–26157.
Celie PH, Klaassen RV, van Rossum-Fikkert SE, van
Elk R, van Nierop P, Smit AB & Sixma TK (2005)
Crystal structure of acetylcholine-binding protein from
Bulinus truncatus reveals the conserved structural scaf-
48
49
50
51
52
53
54
55
56
57
fold and sites of variation in nicotinic acetylcholine
receptors. J Biol Chem 280, 26457–26466.
Kostelidou K, Trakas N, Zouridakis M, Bitzopoulou
K, Sotiriadis A, Gavra I & Tzartos SJ (2006) Expression and characterisation of soluble forms of the extracellular domains of the b,c and e subunits of the
human muscle acetylcholine receptor. FEBS J 273,
3557–3568.
Tierney ML & Unwin N (2000) Electron microscopic
evidence for the assembly of soluble pentameric extracellular domains of the nicotinic acetylcholine receptor.
J Mol Biol 303, 185–196.
Avramopoulou V, Mamalaki A & Tzartos SJ (2004)
Soluble, oligomeric, and ligand-binding extracellular
domain of the human alpha7 acetylcholine receptor
expressed in yeast: replacement of the hydrophobic
cysteine loop by the hydrophilic loop of the ACh-binding protein enhances protein solubility. J Biol Chem
279, 38287–38293.
Levandoski MM, Lin Y, Moise L, McLaughlin JT,
Cooper E & Hawrot E (1999) Chimeric analysis of a
neuronal nicotinic acetylcholine receptor reveals amino
acids conferring sensitivity to alpha-bungarotoxin.
J Biol Chem 274, 26113–26119.
Zeng H, Moise L, Grant MA & Hawrot E (2001) The
solution structure of the complex formed between
alpha-bungarotoxin and an 18-mer cognate peptide
derived from the alpha 1 subunit of the nicotinic acetylcholine receptor from Torpedo californica. J Biol
Chem 276, 22930–22940.
Harel M, Kasher R, Nicolas A, Guss JM, Balass M,
Fridkin M, Smit AB, Brejc K, Sixma TK, KatchalskiKatzir E et al. (2001) The binding site of acetylcholine
receptor as visualized in the X-ray structure of a complex between alpha-bungarotoxin and a mimotope
peptide. Neuron 32, 265–275.
Balass M, Katchalski-Katzir E & Fuchs S (1997)
The alpha-bungarotoxin binding site on the nicotinic
acetylcholine receptor: analysis using a phageepitope library. Proc Natl Acad Sci USA 94, 6054–
6058.
Hansen SB, Sulzenbachre G, Huxford T, Marchot P,
Taylor P & Bourne Y (2005) Structures of Aplysia
AChBP complexes with nicotinic agonists and antagonists reveal distinctive binding interfaces and conformations. EMBO J 24, 3635–3646.
Bourne Y, Talley T, Hansen SB, Taylor P & Marchot
P (2005) Crystal structure of Cbtx–AChBP complex
reveals essential interactions between snake alpha-neurotoxins and nicotinic receptors. EMBO J 24, 1512–
1522.
Schrattenholz A, Godovac-Zimmermann J, Schaèfer
HJ, Albuquerque EX & Maelicke A (1993) Photoaffinity labeling of Torpedo acetylcholine receptor by
physostigmine. Eur J Biochem 216, 671–677.
FEBS Journal 274 (2007) 3799–3845 ª 2007 The Authors Journal compilation ª 2007 FEBS
3831
Nicotinic acetylcholine receptors
D. Kalamida et al.
58 Buisson B & Bertrand D (1998) Allosteric modulation
of neuronal nicotinic acetylcholine-receptors. J Physiol
(Paris) 92, 89–100.
59 Lena C & Changeux JP (1993) Allosteric modulators
of the nicotinic acetylcholine receptor. Trends Neurosci
16, 181–186.
60 Iorga B, Herlem D, Barré E & Guillou C (2006)
Acetylcholine nicotinic receptors: finding the putative
binding site of allosteric modulators using the
‘blind docking’ approach. J Mol Model 12, 366–
372.
61 Tsetlin VI & Hucho F (2004) Snake and snail toxins
acting on nicotinic acetylcholine receptors: fundamental aspects and medical applications. FEBS Lett 557,
9–13.
62 Neumann D, Barchan D, Safran A, Gershoni JM &
Fuchs S (1986) Mapping of the a-bungarotoxin binding site within the a subunit of the acetylcholine receptor. Proc Natl Acad Sci USA 83, 3008–3011.
63 Tzartos SJ & Remoundos MS (1990) Fine localization
of the major a-bungarotoxin binding site to residues
a189–a195 of the Torpedo acetylcholine receptor.
J Biol Chem 265, 21462–21467.
64 Ruan KH, Spurlino J, Quiocho FA & Atassi MZ
(1990) Acetylcholine receptor–a-bungarotoxin interactions: determination of the region-to-region contacts
by peptide–peptide interactions and molecular modeling of the receptor cavity. Proc Natl Acad Sci USA
87, 6156–6160.
65 Moise L, Piserchio A, Basus B & Hawrot E (2002)
NMR structural analysis of a-bungarotoxin and its
complex with the principal a-neurotoxin-binding
sequence on the a7 subunit of a neuronal nicotinic
acetylcholine receptor. J Biol Chem 277, 12406–
12417.
66 Marinou M & Tzartos SJ (2003) Identification of
regions involved in the binding of alpha-bungarotoxin
to the human alpha7 neuronal nicotinic acetylcholine
receptor using synthetic peptides. Biochem J 372, 543–
554.
67 Gotti C, Frigerio F, Bolognesi M, Longhi R, Racchetti
G & Clementi F (1988) Nicotinic acetylcholine receptor: a structural model for a-subunit peptide 188–201,
the putative binding site for cholinergic agents. FEBS
Lett 228, 118–122.
68 Wilson PT & Lentz TL (1988) Binding of a-bungarotoxin to synthetic peptides corresponding to residues
173–204 of the a subunit of Torpedo, calf, and human
acetylcholine receptor and restoration of high-affinity
binding by sodium dodecyl sulphate. Biochemistry 27,
6667–6674.
69 Osaka H, Sugiyama N & Taylor P (1998) Distinctions
in agonist and antagonist specificity conferred by anionic residues of the nicotinic acetylcholine receptor.
J Biol Chem 273, 12758–12765.
3832
70 Blount P & Merlie JP (1990) Mutational analysis of
muscle nicotinic acetylcholine receptor subunit assembly. J Cell Biol 111, 2613–2622.
71 Gehle VM & Sumikawa K (1991) Site-directed mutagenesis of the conserved N-glycosylation site on the
nicotinic acetylcholine receptor subunits. Brain Res
Mol Brain Res 11, 17–25.
72 Gehle VM, Walcott EC, Nishizaki T & Sumikawa K
(1997) N-Glycosylation at the conserved sites ensures
the expression of properly folded functional ACh
receptors. Brain Res Mol Brain Res 45, 219–229.
73 Prives J & Bar-Sagi D (1983) Effect of tunicamycin, an
inhibitor of protein glycosylation, on the biological
properties of acetylcholine receptor in cultured muscle
cells. J Biol Chem 258, 1775–1780.
74 Shtrom SS & Hall ZW (1996) Formation of a ligandbinding site for the acetylcholine receptor in vitro.
J Biol Chem 271, 25506–25514.
75 Psaridi-Linardaki L, Mamalaki A, Remoundos M &
Tzartos SJ (2002) Expression of soluble ligand- and
antibody-binding extracellular domain of human muscle acetylcholine receptor a subunit in the yeast Pichia
pastoris: role of glycosylation in a-bungarotoxin binding. J Biol Chem 277, 26980–26986.
76 Servent D & Menez A (2001) Snake neurotoxins that
interact with nicotinic acetylcholine receptor. In Handbook of Neurotoxicology, Vol. 1 (Massaro EJ, ed.),
pp. 385–425. Humana Press, Totowa, NJ.
77 Changeaux JP & Edelstein SJ (2005) Chemical structure
of the agonist binding site. In Nicotinic Acetylcholine
Receptors, pp. 51–63. Odile Jacob, New York, NY.
78 McIntosh JM, Santos AD & Olivera BM (1999) Conus
peptides targeted to specific nicotinic acetylcholine
receptor subtypes. Annu Rev Biochem 68, 59–88.
79 Luo S, Nguyen TA, Cartier GE, Olivera BM, Yoshikami D & McIntosh JM (1999) Single-residue alteration in alpha-conotoxin PnIA switches its nAChR
subtype selectivity. Biochemistry 38, 14542–14548.
80 Hogg RC, Hopping G, Alewood PF, Adams DJ &
Bertrand D (2003) Alpha-conotoxins PnIA and
[A10L]PnIA stabilize different states of the alpha7–
L247T nicotinic acetylcholine receptor. J Biol Chem
278, 26908–26914.
81 McIntosh JM, Yoshikami D, Mahe E, Nielsen DB,
Rivier JE, Gray WR & Olivera B (1994) A nicotinic
acetylcholine receptor ligand of unique specificity,
alpha-conotoxin ImI. J Biol Chem 269, 16733–16739.
82 Ellison M, Gao F, Wang HL, Sine SM, McIntosh
JM & Olivera BM (2004) Alpha-conotoxins ImI and
ImII target distinct regions of the human alpha7
nicotinic acetylcholine receptor and distinguish
human nicotinic receptor subtypes. Biochemistry 43,
16019–16026.
83 Hansen SB, Talley TT, Radic Z & Taylor P (2004)
Structural and ligand recognition characteristics of an
FEBS Journal 274 (2007) 3799–3845 ª 2007 The Authors Journal compilation ª 2007 FEBS
Nicotinic acetylcholine receptors
D. Kalamida et al.
84
85
86
87
88
89
90
91
92
93
94
95
96
acetylcholine-binding protein from Aplysia californica.
J Biol Chem 279, 24197–24202.
Dutertre S, Nicke A & Lewis RJ (2005) Beta2 subunit
contribution to 4 ⁄ 7 alpha-conotoxin binding to the
nicotinic acetylcholine receptor. J Biol Chem 280,
30460–30468.
Mukhtasimova N, Free C & Sine SM (2005) Initial
coupling of binding to gating mediated by conserved
residues in the muscle nicotinic receptor. J Gen Physiol
126, 23–39.
Unwin N, Miyazawa A, Li J & Fujiyoshi Y (2002)
Activation of the nicotinic acetylcholine receptor
involves a switch in conformation of the a subunits.
J Mol Biol 319, 1165–1176.
Raftery MA, Hunkapiller MW, Strader CD & Hood
LE (1980) Acetylcholine receptor: complex of homologous subunits. Science 208, 1454–1456.
Mishina M, Takai T, Imoto K, Noda M, Takahashi
T, Numa S, Methfessel C & Sakmann B (1986)
Molecular distinction between fetal and adult forms of
muscle acetylcholine receptor. Nature 321, 406–411.
Witzemann V, Barg B, Nishikawa Y, Sakmann B &
Numa S (1987) Differential regulation of muscle
acetylcholine receptor gamma- and epsilon-subunit
mRNAs. FEBS Lett 223, 104–112.
Sine SM & Claudio T (1991) Gamma- and delta-subunits regulate the affinity and the cooperativity of ligand binding to the acetylcholine receptor. J Biol Chem
266, 19369–19377.
Ackermann EJ & Taylor P (1997) Nonidentity of
the a-neurotoxin binding sites on the nicotinic acetylcholine receptor revealed by modification in a-neurotoxin and receptor structures. Biochemistry 36,
12836–12844.
Sine SM, Ohno K, Bouzat C, Auerbach A, Milone M,
Pruitt JN & Engel AG (1995) Mutation of the acetylcholine receptor e subunit causes a slow-channel
myasthenic syndrome by enhancing agonist-binding
affinity. Neuron 15, 229–239.
Wheeler SV, Jane SD, Cross KM, Chad JE & Foreman
RC (1994) Membrane clustering and bungarotoxin
binding by the nicotinic acetylcholine receptor: role of
the beta subunit. J Neurochem 63, 1891–1899.
Green WN, Ross AF & Claudio T (1991) Acetylcholine receptor assembly is stimulated by phosphorylation of its c subunit. Neuron 7, 659–666.
Ramanathan VK & Hall ZW (1999) Altered glycosylation sites of the d subunit of the acetylcholine receptor
(ACHR) reduce ad association and receptor assembly.
J Biol Chem 274, 20513–20520.
Missias AC, Chu GC, Klocke BJ, Sanes JR & Merlie
JP (1996) Maturation of the acetylcholine receptor in
skeletal muscle: regulation of the AChR c-to-e switch.
Dev Biol 179, 223–238.
97 Yumoto N, Wakatsuki S & Sehara-Fujisawa A (2005)
The acetylcholine c-to-e switch occurs in individual
endplates. Biochem Biophys Res Commun 331, 1522–
1527.
98 Sanes JR, Johnson YR, Kotzbauer PT, Mudd J, Hanley T, Martinou JC & Merlie JP (1991) Selective
expression of an acetylcholine receptor-lacZ transgene
in synaptic nuclei of adult muscle fibres. Development
113, 1181–1191.
99 Newland CF, Beeson D, Vincent A & Newsom-Davis
J (1993) Human nicotinic acetylcholine receptor a-subunit isoforms: origins and expression. Nucleic Acids
Res 21, 5463–5467.
100 Newland CF, Beeson D, Vincent A & Newsom-Davis
J (1995) Functional and non-functional isoforms of the
human muscle acetylcholine receptor. J Physiol 489,
767–778.
101 Gattenlöhner S, Schneider C, Thamer C, Klein R,
Roggendorf W, Gohlke F, Niethammer C, Czub S,
Vincent A, Müller-Hermelink HK et al. (2002) Expression of foetal type acetylcholine receptor to type I
muscle fibres in human neuromuscular disorders. Brain
125, 1309–1319.
102 Johnson MA, Polgar J, Weightman D & Appleton D
(1973) Data on the distribution of fibre types in thirtysix human muscles. An autopsy study. J Neurol Sci 18,
111–129.
103 Adams L, Carlson BM, Henderson L & Goldman D
(1995) Adaptation of nicotinic acetylcholine receptor,
myogenin and MRF4 gene expression to long-term
denervation. J Cell Biol 131, 1341–1349.
104 MacLennan C, Beeson D, Buigis AM, Vincent A &
Newsom-Davis J (1997) Acetylcholine receptor expression in human extraocular muscles and their susceptibility to myasthenia gravis. Ann Neurol 41, 423–431.
105 Lindstrom JM (2000) Acetylcholine receptors and
myasthenia. Muscle Nerve 23, 453–477.
106 Hughes BW, Kusner LL & Kaminski HJ (2006)
Molecular architecture of the neuromuscular junction.
Muscle Nerve 33, 445–461.
107 Fambrough DM (1979) Control of acetylcholine receptors in skeletal muscle. Physiol Rev 59, 165–227.
108 Ponomareva ON, Ma H, Dakour R, Raabe TD, Lai C
& Rimer M (2005) Stimulation of acetylcholine receptor transcription by neuregelin-2 requires an N-box
response element and is regulated by alternative
splicing. Neuroscience 134, 495–503.
109 Willmann R & Fuhrer C (2002) Neuromuscular
synaptogenesis: clustering of acetylcholine receptors
revisited. Cell Mol Life Sci 59, 1296–1316.
110 Watty A, Neubauer G, Dreger M, Zimmer M,
Wilm M & Burden SJ (2000) The in vitro and in vivo
phosphotyrosine map of activated MuSK. Proc Natl
Acad Sci USA 97, 4585–4590.
FEBS Journal 274 (2007) 3799–3845 ª 2007 The Authors Journal compilation ª 2007 FEBS
3833
Nicotinic acetylcholine receptors
D. Kalamida et al.
111 Tsim KWK, Choi RCY, Siow NL, Cheng AWM, Ling
KKY, Jiang JXS, Tung EKK, Lee HHC, Xie QH,
Simon J et al. (2003) ATP induces post-synaptic gene
expressions in vertebrate skeletal neuromuscular junctions. J Neurocyt 32, 603–617.
112 Jaworski A & Burden SJ (2006) Neuromuscular synapse formation in mice lacking motor-neuron and skeletal-muscle-derived neuregelin-1. J Neurosci 26, 655–
661.
113 Kukhtina V, Kottwitz D, Strauss H, Heise B, Chebotareva N, Tsetlin V & Hucho F (2006) Intracellular
domain of nicotinic acetylcholine receptor: the importance of being unfolded. J Neurochem 97, 63–67.
114 Fischbach GD & Schuetze SM (1980) A post-natal
decrease in acetylcholine channel open time at rat endplates. J Physiol 303, 125–137.
115 Koenen M, Peter C, Villaroel A, Witzemann V & Sakmann B (2005) Acetylcholine receptor channel subtype
directs the innervation pattern of skeletal muscle.
EMBO Reports 6, 570–576.
116 Schuetze SM & Role LW (1987) Developmental regulation of nicotinic acetylcholine receptors. Annu Rev
Neurosci 10, 403–457.
117 Missias AC, Mudd J, Cunningham JM, Steinbach J,
Merlei JP & Sanes JR (1997) Deficient development
and maintenance of postsynaptic specializations in
mutant mice lacking an ‘adult’ acetylcholine receptor
subunit. Development 124, 5075–5086.
118 Grassi F & Degasperi V (2000) Modulation of fetal
and adult acetylcholine and Mg2+ at developing
mouse end-plates. Pflügers Arch Eur J Physiol 440,
704–709.
119 Herlitze S, Villarroel A, Witzemann V, Koenen M &
Sakmann B (1996) Structural determinants of channel
conductance in fetal and adult rat muscle acetylcholine
receptors. J Physiol 492, 775–787.
120 Takahashi M, Kubo T, Mizoguchi A, Carlson CG &
Ohnishi K (2002) Spontaneous muscle action potentials fail to develop without fetal-type acetylcholine
receptors. EMBO Reports 3, 674–681.
121 Imoto K, Busch C, Sakmann B, Mishina M, Konno
T, Nakai J, Bujo H, Mori Y, Fukuda K & Numa S
(1988) Rings of negatively charged amino acids determine the acetylcholine receptor channel conductance.
Nature 335, 645–651.
122 Grassi F (1999) 5-Hydroxytryptamine blocks the fetal
more potently than the adult mouse muscle acetylcholine receptor. Pflügers Arch Eur J Physiol 437, 903–
909.
123 Salpeter MM (1987) Development and neural control
of the neuromuscular junction and of the junctional
acetylcholine receptor. In The Vertebrate Neuromuscular Junction (Salpeter MM, ed.), pp. 55–115.
Alan R. Liss Inc., New York, NY.
3834
124 Witzemann V, Schwarz H, Koenen M, Berberich C,
Villarroel A, Wernig A, Brenner HR & Sakmann B
(1996) Acetylcholine receptor e-subunit deletion causes
muscle weakness and atrophy in juvenile and adult
mice. Proc Natl Acad Sci USA 93, 13286–13291.
125 Schwarz H, Giese G, Müller H, Koenen M & Witzemann V (2000) Different functions of fetal and adult
subtypes for the formation and maintenance of neuromuscular synapses revealed in e-subunit-deficient mice.
Eur J Neurosci 12, 3107–3116.
126 Salpeter MM, Kasprzak H, Feng H & Fertuck H
(1979) End-plates after esterase inactivation in vivo:
correlation between esterase concentration, functional
response and fine structure. J Neurocytol 8, 95–115.
127 Di Castro A, Martinello K, Grassi F, Eusebi F &
Engel AG (2007) Pathogenic point mutations in a
transmembrane domain of the e subunit increase the
Ca2+ permeability of the human endplate ACh
receptor. J Physiol in press.
128 Jaramillo F, Vicini S & Schuetze SM (1988) Embryonic
acetylcholine receptors guarantee spontaneous contractions in rat developing muscle. Nature 335, 66–68.
129 Merlie JP & Lindstrom J (1983) Assembly in vivo of
mouse muscle acetylcholine receptor: identification of
an a subunit species that may be an assembly intermediate. Cell 34, 747–757.
130 Green WN & Claudio T (1993) Acetylcholine receptor
assembly: subunit folding and oligomerisation occur
sequentially. Cell 74, 57–69.
131 Green WN & Wanamaker CP (1997) The role of the
cysteine loop in acetylcholine receptor assembly. J Biol
Chem 272, 20945–20953.
132 Wanamaker CP & Green WN (2005) N-Linked glycosylation is required for nicotinic receptor assembly but
not for subunit associations with calnexin. J Biol Chem
280, 33800–33810.
133 Green WN & Wanamaker CP (1998) Formation of the
nicotinic acetylcholine receptor binding sites. J Neurosci 18, 5555–5564.
134 Mitra M, Wanamaker CP & Green WN (2001) Rearrangement of nicotinic receptor a subunits during formation of the ligand binding sites. J Neurosci 21,
3000–3008.
135 Saedi MS, Conroy WG & Lindstrom J (1991) Assembly of Torpedo acetylcholine receptors in Xenopus
oocytes. J Cell Biol 112, 1007–1015.
136 Blount P, Smith M & Merlie JP (1990) Assembly intermediates of the mouse muscle nicotinic acetylcholine
receptor in stably transfected fibroblasts. J Cell Biol
111, 2601–2611.
137 Gu Y, Forsayeth JR, Verrall S, Yu XM & Hall ZW
(1991) Assembly of the mammalian muscle acetylcholine receptor in transfected COS cells. J Cell Biol 114,
799–807.
FEBS Journal 274 (2007) 3799–3845 ª 2007 The Authors Journal compilation ª 2007 FEBS
Nicotinic acetylcholine receptors
D. Kalamida et al.
138 Blount P & Merlie JP (1988) Native folding of an acetylcholine receptor a subunit expressed in the absence
of other subunits. J Biol Chem 263, 1072–1080.
139 Paulson HL, Ross AF, Green WN & Claudio T (1991)
Analysis of early events in acetylcholine receptor
assembly. J Cell Biol 113, 1371–1384.
140 Christianson JC & Green WN (2004) Regulation of
nicotinic receptor expression by the ubiquitin-proteasome system. EMBO J 23, 4156–4165.
141 Williamson PT, Meier BH & Watts A (2004) Structural and functional studies of the nicotinic acetylcholine receptor by solid-state NMR. Eur Biophys J 33,
247–254.
142 Tsouloufis T, Mamalaki A, Remoundos M & Tzartos
SJ (2000) Reconstitution of conformationally dependent epitopes on the N-terminal extracellular domain of
the human muscle acetylcholine receptor a subunit
expressed in Escherichia coli: implications for myasthenia gravis therapeutic approaches. Int Immunol 12,
1255–1265.
143 Psaridi-Linardaki L, Trakas N, Mamalaki A & Tzartos SJ (2005) Specific immunoadsorption of the autoantibodies from myasthenic patients using the
extracellular domain of the human muscle acetylcholine receptor alpha-subunit. Development of an antigen-specific therapeutic strategy. J Neuroimmunol 159,
183–191.
144 Li ZL, Li YJ, Guo CY, Shi YW, Xu MQ, Trommer
WE & Yuan JM (2004) Soluble expression and affinity
purification of functional domain of human acetylcholine receptor a-subunit by the modulation of maltose
binding protein. Biotechnol Lett 26, 1765–1769.
145 Sideris S, Lagoumintzis G, Kordas G, Kostelidou K,
Sotiriadis A, Poulas K & Tzartoz S (2007) Isolation
and functional characterization of anti-acetylcholine
receptor subunit specific autoantibodies from
myasthenic patients: receptor loss in cell culture.
J Neuroim, doi: 10.1016/j.jneuroim.2007.06.014.
146 Jansen KU, Conroy WG, Claudio T, Fox TD, Fujita N,
Hamill O, Lindstrom JM, Luther M, Nelson N & Ryan
KA (1989) Expression of the four subunits of the Torpedo californica nicotinic acetylcholine receptor in Saccharomyces cerevisiae. J Biol Chem 264, 15022–15027.
147 Kurosaki T, Fukuda K, Konno T, Mori Y, Tanaka K,
Mishina M & Numa S (1987) Functional properties of
nicotinic acetylcholine receptor subunits expressed in
various combinations. FEBS Lett 214, 253–258.
148 Claudio T (1992) Stable expression of heterologous
multisubunit protein complexes established by calcium
phosphate- or lipid-mediated cotransfection. Methods
Enzymol 207, 391–408.
149 Claudio T, Paulson HL, Green WN, Ross AF, Hartman DS & Hayden D (1989) Fibroblasts transfected
with Torpedo acetylcholine receptor beta-, gamma-,
and delta-subunit cDNAs express functional receptors
150
151
152
153
154
155
156
157
158
159
160
161
when infected with a retroviral alpha recombinant.
J Cell Biol 108, 2277–2290.
Buller AL & White MM (1990) Functional acetylcholine receptors expressed in Xenopus oocytes after injection of Torpedo beta, gamma, and delta subunit RNAs
are a consequence of endogenous oocyte gene expression. Mol Pharmacol 37, 423–428.
Sumikawa K & Gehle VM (1992) Assembly of mutant
subunits of the nicotinic acetylcholine receptor lacking
the conserved disulfide loop structure. J Biol Chem
267, 6286–6290.
White MM, Mayne KM, Lester HA & Davidson N
(1985) Mouse–Torpedo hybrid acetylcholine receptors: functional homology does not equal sequence
homology. Proc Natl Acad Sci USA 82, 4852–4856.
Mayne KM, Yoshii K, Yu L, Lester HA & Davidson
N (1987) Expression of mouse–Torpedo acetylcholine
receptor subunit chimeras and hybrids in Xenopus
oocytes. Brain Res 388, 191–197.
Yu L, Leonard RJ, Davidson N & Lester HA (1991)
Single-channel properties of mouse–Torpedo acetylcholine receptor hybrids expressed in Xenopus oocytes.
Brain Res Mol Brain Res 10, 203–211.
Butler DH, Lasalde JA, Butler JK, Tamamizu S,
Zimmerman G & McNamee MG (1997) Mouse–
Torpedo chimeric alpha-subunit used to probe channelgating determinants on the nicotinic acetylcholine
receptor primary sequence. Cell Mol Neurobiol 17,
13–33.
Sumikawa K & Miledi R (1989) Change in desensitization of cat muscle acetylcholine receptor caused by
coexpression of Torpedo acetylcholine receptor subunits in Xenopus oocytes. Proc Natl Acad Sci USA 86,
367–371.
Paulson HL & Claudio T (1990) Temperature-sensitive
expression of all-Torpedo and Torpedo–rat hybrid AChR
in mammalian muscle cells. J Cell Biol 110, 1705–1717.
Wang YD & Claudio TJ (1993) Xenopus muscle acetylcholine receptor alpha subunits bind ligands with different affinities. J Biol Chem 268, 18782–18793.
Imoto K, Sakmann B, Mishina M, Mori Y, Konno T,
Fukuda K, Kurasaki M, Bujo H, Fujita Y & Numa S
(1986) Location of a delta-subunit region determining
ion transport through the acetylcholine receptor channel. Nature 324, 670–674.
Loutrari H, Tzartos SJ & Claudio T (1992) Use of
Torpedo–mouse hybrid acetylcholine receptors
reveals immunodominance of the alpha subunit in
myasthenia gravis antisera. Eur J Immunol 22,
2946–2956.
Loutrari H, Kokla A, Trakas N & Tzartos SJ (1997)
Expression of human–Torpedo hybrid acetylcholine
receptor (AChR) for analysing the subunit specificity
of antibodies in sera from patients with myasthenia
gravis (MG). Clin Exp Immunol 109, 538–546.
FEBS Journal 274 (2007) 3799–3845 ª 2007 The Authors Journal compilation ª 2007 FEBS
3835
Nicotinic acetylcholine receptors
D. Kalamida et al.
162 Jackson MB, Imoto K, Mishina M, Konno T, Numa
S & Sakmann B (1990) Spontaneous and agonistinduced openings of an acetylcholine receptor channel
composed of bovine muscle alpha-, beta- and deltasubunits. Pflugers Arch 417, 129–135.
163 Criado M, Koenen M & Sakmann B (1990) Assembly
of an adult type acetylcholine receptor in a mouse cell
line transfected with rat muscle epsilon-subunit DNA.
FEBS Lett 270, 95–99.
164 Charnet P, Labarca C & Lester HA (1992) Structure
of the gamma-less nicotinic acetylcholine receptor:
learning from omission. Mol Pharmacol 41, 708–717.
165 Liu Y & Brehm P (1993) Expression of subunit-omitted mouse nicotinic acetylcholine receptors in Xenopus
laevis oocytes. J Physiol 470, 349–363.
166 Liu E, Hamill OP & Salpeter MM (1994) Mouse muscle epsilon- and gamma-containing acetylcholine receptors expressed in Xenopus laevis oocytes do not differ
in their degradation half-lives. Neurosci Lett 174,
77–80.
167 Garcia-Colunga J & Miledi R (1996) Serotonergic
modulation of muscle acetylcholine receptors of different subunit composition. Proc Natl Acad Sci USA 93,
3990–3994.
168 Forsayeth JR, Franco A Jr, Rossi AB, Lansman JB &
Hall ZW (1990) Expression of functional mouse muscle acetylcholine receptors in Chinese hamster ovary
cells. J Neurosci 10, 2771–2779.
169 Witzemann V, Stein E, Barg B, Konno T, Koenen M,
Kues W, Criado M, Hofmann M & Sakmann B (1990)
Primary structure and functional expression of the
alpha-, beta-, gamma-, delta- and epsilon-subunits
of the acetylcholine receptor from rat muscle. Eur J
Biochem 194, 437–448.
170 Carlson CG & Feng Y (1993) Asynaptic expression of
the adult nicotinic acetylcholine receptor in long-term
cultures of mammalian myotubes. Brain Res Dev Brain
Res 72, 245–252.
171 Gu Y, Franco A Jr, Gardner PD, Lansman JB, Forsayeth JR & Hall ZW (1990) Properties of embryonic
and adult muscle acetylcholine receptors transiently
expressed in COS cells. Neuron 5, 147–157.
172 Kullberg R, Owens JL, Camacho P, Mandel G &
Brehm P (1990) Multiple conductance classes of mouse
nicotinic acetylcholine receptors expressed in Xenopus
oocytes. Proc Natl Acad Sci USA 87, 2067–2071.
173 Santos S & Aizenman E (2002) Functional expression
of muscle-type nicotinic acetylcholine receptors in rat
forebrain neurons in vitro. Methods Find Exp Clin
Pharmacol 24, 63–66.
174 Wang ZZ, Hardy SF & Hall ZW (1996) Assembly of
the nicotinic acetylcholine receptor: the first transmembrane domains of truncated a and d subunits are
required for heterodimer formation in vivo. J Biol
Chem 271, 27575–27584.
3836
175 Plazas PV, Katz E, Gomez-Casati ME, Bouzat C &
Elgoyhen AB (2005) Stoichiometry of the alpha9alpha10 nicotinic cholinergic receptor. J Neurosci 25,
10905–10912.
176 Grinevich VP, Letchworth SR, Lindenberger KA,
Menager J, Mary V, Sadieva KA, Buhlman LM,
Bohme GA, Pradier L, Benavides J, et al. (2005)
Heterologous expression of human {alpha}6{beta}4{beta}3{alpha}5 nicotinic acetylcholine receptors: binding properties consistent with their natural expression
require quaternary subunit assembly including the
{alpha}5 subunit. J Pharmacol Exp Ther 312, 619–
626.
177 Xiao Y & Kellar KJ (2004) The comparative pharmacology and up-regulation of rat neuronal nicotinic
receptor subtype binding sites stably expressed in
transfected mammalian cells. J Pharmacol Exp Ther
310, 98–107.
178 Wu J, Liu Q, Yu K, Hu J, Kuo YP, Segerberg M, St
John PA & Lukas RJ (2006) Roles of nicotinic acetylcholine receptor beta subunits in function of human
alpha4-containing nicotinic receptors. J Physiol 576,
103–118.
179 Groot-Kormelink PJ, Luyten WH, Colquhoun D &
Sivilotti LG (1998) A reporter mutation approach
shows incorporation of the ‘orphan’ subunit beta3 into
a functional nicotinic receptor. J Biol Chem 273,
15317–15320.
180 Ramirez-Latorre J, Yu CR, Qu X, Perin F, Karlin A
& Role L (1996) Functional contributions of alpha5
subunit to neuronal acetylcholine receptor channels.
Nature 380, 347–351.
181 Fischer H, Orr-Urtreger A, Role L & Huck S (2005)
Selective deletion of the a5 subunit differentially
affects somatic dendritic versus axonaly targeted nicotinic Ach receptors in mouse. J Physiol 563, 119–137.
182 Wang N, Orr-Urtreger A, Chapman J, Rabinowitz R,
Nachman R & Korczyn AD (2002) Autonomic
function in mice lacking alpha5 neuronal nicotinic
acetylcholine receptor subunit. J Physiol 542,
347–354.
183 Williams BM, Temburni MK, Levey MS, Bertrand S,
Bertrand D & Jacob MH (1998) The long internal
loop of the alpha 3 subunit targets nAChRs to subdomains within individual synapses on neurons in vivo.
Nat Neurosci 1, 557–562.
184 Xu J, Zhu Y & Heinemann SF (2006) Identification of
sequence motifs that target neuronal nicotinic receptors to dendrites and axons. J Neurosci 26, 9780–9793.
185 Adams DJ & Nutter TJ (1992) Calcium permeability
and modulation of nicotinic acetylcholine receptorchannels in rat parasympathetic neurons. J Physiol
(Paris) 86, 67–76.
186 Castro NG & Albuquerque EX (1995) Alpha-bungarotoxin-sensitive hippocampal nicotinic receptor channel
FEBS Journal 274 (2007) 3799–3845 ª 2007 The Authors Journal compilation ª 2007 FEBS
Nicotinic acetylcholine receptors
D. Kalamida et al.
187
188
189
190
191
192
193
194
195
196
197
198
199
has a high calcium permeability. Biophys J 68, 516–
524.
Rathouz MM & Berg DK (1994) Synaptic-type acetylcholine receptors raise intracellular calcium levels in
neurons by two mechanisms. J Neurosci 14, 6935–
6945.
Dajas-Bailador FA, Mogg AJ & Wonnacott S (2002)
Intracellular Ca2+ signals evoked by stimulation of
nicotinic acetylcholine receptors in SH-SY5Y cells:
contribution of voltage-operated Ca2+ channels and
Ca2+ stores. J Neurochem 81, 606–614.
Brain KL, Trout SJ, Jackson VM, Dass N & Cunnane
TC (2001) Nicotine induces calcium spikes in single
nerve terminal varicosities: a role for intracellular calcium stores. Neuroscience 106, 395–403.
Shoop RD, Chang KT, Ellisman MH & Berg DK
(2001) Synaptically driven calcium transients via
nicotinic receptors on somatic spines. J Neurosci 21,
771–781.
Beker F, Weber M, Fink RH & Adams DJ (2003)
Muscarinic and nicotinic ACh receptor activation
differentially mobilize Ca2+ in rat intracardiac
ganglion neurons. J Neurophysiol 90, 1956–1964.
Vijayaraghavan S, Pugh PC, Zhang ZW, Rathouz
MM & Berg DK (1992) Nicotinic receptors that bind
alpha-bungarotoxin on neurons raise intracellular free
Ca2+. Neuron 8, 353–362.
Barrantes GE, Murphy CT, Westwick J & Wonnacott
S (1995) Nicotine increases intracellular calcium in rat
hippocampal neurons via voltage-gated calcium channels. Neurosci Lett 196, 101–104.
Tsuneki H, Klink R, Lena C, Korn H & Changeux JP
(2000) Calcium mobilization elicited by two types of
nicotinic acetylcholine receptors in mouse substantia
nigra pars compacta. Eur J Neurosci 12, 2475–2485.
Sharma G & Vijayaraghavan S (2001) Nicotinic cholinergic signaling in hippocampal astrocytes involves
calcium-induced calcium release from intracellular
stores. Proc Natl Acad Sci USA 98, 4148–4153.
Quick MW & Lester RA (2002) Desensitization of
neuronal nicotinic receptors. J Neurobiol 53, 457–478.
Khiroug L, Giniatullin R, Klein RC, Fayuk D &
Yakel JL (2003) Functional mapping and Ca2+ regulation of nicotinic acetylcholine receptor channels in rat
hippocampal CA1 neurons. J Neurosci 23, 9024–9031.
Champtiaux N, Gotti C, Cordero-Erausquin M, David
DJ, Przybylski C, Lena C, Clementi F, Moretti M,
Rossi FM, Le Novere N et al. (2003) Subunit composition of functional nicotinic receptors in dopaminergic
neurons investigated with knock-out mice. J Neurosci
23, 7820–7829.
Salminen O, Murphy KL, McIntosh JM, Drago J,
Marks MJ, Collins AC & Grady SR (2004) Subunit
composition and pharmacology of two classes of striatal presynaptic nicotinic acetylcholine receptors medi-
200
201
202
203
204
205
206
207
208
209
210
211
ating dopamine release in mice. Mol Pharmacol 65,
1526–1535.
Breese CR, Adams C, Logel J, Drebing C, Rollins Y,
Barnhart M, Sullivan B, Demasters BK, Freedman R
& Leonard S (1997) Comparison of the regional
expression of nicotinic acetylcholine receptor alpha7
mRNA and [125I]-alpha-bungarotoxin binding in
human post-mortem brain. J Comp Neurol 387, 385–
398.
Chen D & Patrick JW (1997) The alpha-bungarotoxinbinding nicotinic acetylcholine receptor from rat brain
contains only the alpha7 subunit. J Biol Chem 272,
24024–24029.
Kawai H, Zago W & Berg DK (2002) Nicotinic alpha
7 receptor clusters on hippocampal GABAergic neurons: regulation by synaptic activity and neurotrophins. J Neurosci 22, 7903–7912.
Elgoyhen AB, Johnson DS, Boulter J, Vetter DE &
Heinemann S (1994) Alpha 9: an acetylcholine receptor
with novel pharmacological properties expressed in rat
cochlear hair cells. Cell 79, 705–715.
Elgoyhen AB, Vetter DE, Katz E, Rothlin CV,
Heinemann SF & Boulter J (2001) Alpha10: a
determinant of nicotinic cholinergic receptor function
in mammalian vestibular and cochlear mechanosensory hair cells. Proc Natl Acad Sci USA 98,
3501–3506.
Battaglioli E, Gotti C, Terzano S, Flora A, Clementi F
& Fornasari D (1998) Expression and transcriptional
regulation of the human alpha3 neuronal nicotinic
receptor subunit in T lymphocyte cell lines. J Neurochem 71, 1261–1270.
Flora A, Schulz R, Benfante R, Battaglioli E, Terzano
S, Clementi F & Fornasari D (2000) Neuronal and
extraneuronal expression and regulation of the human
alpha5 nicotinic receptor subunit gene. J Neurochem
75, 18–27.
Flora A, Schulz R, Benfante R, Battaglioli E, Terzano
S, Clementi F & Fornasari D (2000) Transcriptional
regulation of the human alpha5 nicotinic receptor subunit gene in neuronal and non-neuronal tissues. Eur J
Pharmacol 393, 85–95.
Fornasari D, Battaglioli E, Flora A, Terzano S &
Clementi F (1997) Structural and functional characterization of the human alpha3 nicotinic subunit gene promoter. Mol Pharmacol 51, 250–261.
Ficklin MB, Zhao S & Feng G (2005) Ubiquilin-1
regulates nicotine-induced up-regulation of neuronal
nicotinic acetylcholine receptors. J Biol Chem 280,
34088–34095.
Dineley KT & Patrick JW (2000) Amino acid determinants of alpha 7 nicotinic acetylcholine receptor surface expression. J Biol Chem 275, 13974–13985.
Ren XQ, Cheng SB, Treuil M, Mukherjee J, Rao J,
Braunewell KH, Lindstrom JM & Anand R (2005)
FEBS Journal 274 (2007) 3799–3845 ª 2007 The Authors Journal compilation ª 2007 FEBS
3837
Nicotinic acetylcholine receptors
212
213
214
215
216
217
218
219
220
221
222
223
3838
D. Kalamida et al.
Structural determinants of alpha4beta2 nicotinic acetylcholine receptor trafficking. J Neurosci 25, 6676–
6686.
Castillo M, Mulet J, Gutierrez LM, Ortiz JA, Castelan
F, Gerber S, Sala S, Sala F & Criado M (2005) Dual
role of the RIC-3 protein in trafficking of serotonin
and nicotinic acetylcholine receptors. J Biol Chem 280,
27062–27068.
Williams ME, Burton B, Urrutia A, Shcherbatko A,
Chavez-Noriega LE, Cohen CJ & Aiyar J (2005) Ric-3
promotes functional expression of the nicotinic acetylcholine receptor alpha7 subunit in mammalian cells.
J Biol Chem 280, 1257–1263.
Bruses JL, Chauvet N & Rutishauser U (2001) Membrane lipid rafts are necessary for the maintenance of
the (alpha) 7 nicotinic acetylcholine receptor in somatic
spines of ciliary neurons. J Neurosci 21, 504–512.
Zhu D, Xiong WC & Mei L (2006) Lipid rafts serve as
a signaling platform for nicotinic acetylcholine receptor clustering. J Neurosci 26, 4841–4851.
Liu Z, Tearle AW, Nai Q & Berg DK (2005) Rapid
activity-driven SNARE-dependent trafficking of nicotinic receptors on somatic spines. J Neurosci 25, 1159–
1168.
Flores CM, Rogers SW, Pabreza LA, Wolfe BB &
Kellar KJ (1992) A subtype of nicotinic cholinergic
receptor in rat brain is composed of alpha 4 and beta
2 subunits and is up-regulated by chronic nicotine
treatment. Mol Pharmacol 41, 31–37.
Avila AM, Davila-Garcia MI, Ascarrunz VS, Xiao Y
& Kellar KJ (2003) Differential regulation of nicotinic
acetylcholine receptors in PC12 cells by nicotine and
nerve growth factor. Mol Pharmacol 64, 974–986.
Cormier A, Paas Y, Zini R, Tillement JP, Lagrue G,
Changeux JP & Grailhe R (2004) Long-term exposure
to nicotine modulates the level and activity of acetylcholine receptors in white blood cells of smokers and
model mice. Mol Pharmacol 66, 1712–1718.
Harkness PC & Millar NS (2002) Changes in conformation and subcellular distribution of alpha4beta2
nicotinic acetylcholine receptors revealed by chronic
nicotine treatment and expression of subunit chimeras.
J Neurosci 22, 10172–10181.
Meyer EL, Xiao Y & Kellar KJ (2001) Agonist regulation of rat alpha3 beta4 nicotinic acetylcholine receptors stably expressed in human embryonic kidney 293
cells. Mol Pharmacol 60, 568–576.
Peng X, Gerzanich V, Anand R, Whiting PJ & Lindstrom J (1994) Nicotine-induced increase in neuronal
nicotinic receptors results from a decrease in the rate
of receptor turnover. Mol Pharmacol 46, 523–530.
Kuryatov A, Luo J, Cooper J & Lindstrom J (2005)
Nicotine acts as a pharmacological chaperone to
up-regulate human alpha4beta2 acetylcholine receptors. Mol Pharmacol 68, 1839–1851.
224 Sallette J, Bohler S, Benoit P, Soudant M, Pons S, Le
Novere N, Changeux JP & Corringer PJ (2004) An
extracellular protein microdomain controls up-regulation of neuronal nicotinic acetylcholine receptors by
nicotine. J Biol Chem 279, 18767–18775.
225 Nashmi R, Dickinson ME, McKinney S, Jareb M,
Labarca C, Fraser SE & Lester HA (2003) Assembly
of alpha4beta2 nicotinic acetylcholine receptors
assessed with functional fluorescently labeled subunits:
effects of localization, trafficking, and nicotine-induced
upregulation in clonal mammalian cells and in cultured
midbrain neurons. J Neurosci 23, 11554–11567.
226 Zhang ZW, Coggan JS & Berg DK (1996) Synaptic
currents generated by neuronal acetylcholine receptors
sensitive to alpha-bungarotoxin. Neuron 17, 1231–
1240.
227 Ullian EM, McIntosh JM & Sargent PB (1997) Rapid
synaptic transmission in the avian ciliary ganglion is
mediated by two distinct classes of nicotinic receptors.
J Neurosci 17, 210–219.
228 Alkondon M, Pereira EF & Albuquerque EX (1998)
Alpha-bungarotoxin- and methyllycaconitine-sensitive
nicotinic receptors mediate fast synaptic transmission
in interneurons of rat hippocampal slices. Brain Res
810, 257–263.
229 Frazier CJ, Buhler AV, Weiner JL & Dunwiddie TV
(1998) Synaptic potentials mediated via alpha-bungarotoxin-sensitive nicotinic acetylcholine receptors in rat
hippocampal interneurons. J Neurosci 18, 8228–8235.
230 Jones S, Sudweeks S & Yakel JL (1999) Nicotinic
receptors in the brain: correlating physiology with
function. Trends Neurosci 22, 555–561.
231 Lena C, Changeux JP & Mulle C (1993) Evidence for
‘preterminal’ nicotinic receptors on GABAergic axons
in the rat interpeduncular nucleus. J Neurosci 13,
2680–2688.
232 Wonnacott S (1997) Presynaptic nicotinic ACh receptors. Trends Neurosci 20, 92–98.
233 Sher E, Chen Y, Sharples TJ, Broad LM, Benedetti G,
Zwart R, McPhie GI, Pearson KH, Baldwinson T &
De Filippi G (2004) Physiological roles of neuronal
nicotinic receptor subtypes: new insights on the nicotinic modulation of neurotransmitter release, synaptic
transmission and plasticity. Curr Top Med Chem 4,
283–297.
234 Jensen AA, Frolund B, Liljefors T & Krogsgaard-Larsen P (2005) Neuronal nicotinic acetylcholine receptors: structural revelations, target identifications, and
therapeutic inspirations. J Med Chem 48, 4705–4745.
235 Soliakov L & Wonnacott S (1996) Voltage-sensitive
Ca2+ channels involved in nicotinic receptor-mediated
[3H]dopamine release from rat striatal synaptosomes.
J Neurochem 67, 163–170.
236 Kulak JM, McIntosh JM, Yoshikami D & Olivera
BM (2001) Nicotine-evoked transmitter release from
FEBS Journal 274 (2007) 3799–3845 ª 2007 The Authors Journal compilation ª 2007 FEBS
Nicotinic acetylcholine receptors
D. Kalamida et al.
237
238
239
240
241
242
243
244
245
246
247
248
249
synaptosomes: functional association of specific presynaptic acetylcholine receptors and voltage-gated calcium channels. J Neurochem 77, 1581–1589.
Turner TJ (2004) Nicotine enhancement of dopamine
release by a calcium-dependent increase in the size
of the readily releasable pool of synaptic vesicles.
J Neurosci 24, 11328–11336.
Sharma G & Vijayaraghavan S (2003) Modulation of
presynaptic store calcium induces release of glutamate
and postsynaptic firing. Neuron 38, 929–939.
Soliakov L & Wonnacott S (2001) Involvement of protein kinase C in the presynaptic nicotinic modulation
of [3H]-dopamine release from rat striatal synaptosomes. Br J Pharmacol 132, 785–791.
Cox ME & Parsons SJ (1997) Roles for protein kinase
C and mitogen-activated protein kinase in nicotineinduced secretion from bovine adrenal chromaffin cells.
J Neurochem 69, 1119–1130.
Levy RB & Aoki C (2002) Alpha7 nicotinic acetylcholine receptors occur at postsynaptic densities of
AMPA receptor-positive and -negative excitatory
synapses in rat sensory cortex. J Neurosci 22, 5001–
5015.
Ji D, Lape R & Dani JA (2001) Timing and location
of nicotinic activity enhances or depresses hippocampal
synaptic plasticity. Neuron 31, 131–141.
Arenella LS, Oliva JM & Jacob MH (1993) Reduced
levels of acetylcholine receptor expression in chick ciliary ganglion neurons developing in the absence of
innervation. J Neurosci 13, 4525–4537.
Corriveau RA & Berg DK (1993) Coexpression of
multiple acetylcholine receptor genes in neurons: quantification of transcripts during development. J Neurosci
13, 2662–2671.
Devay P, Qu X & Role L (1994) Regulation of
nAChR subunit gene expression relative to the development of pre- and postsynaptic projections of
embryonic chick sympathetic neurons. Dev Biol 162,
56–70.
Howard MJ, Gershon MD & Margiotta JF (1995)
Expression of nicotinic acetylcholine receptors and
subunit mRNA transcripts in cultures of neural crest
cells. Dev Biol 170, 479–495.
Smith J, Fauquet M, Ziller C & Le Douarin NM
(1979) Acetylcholine synthesis by mesencephalic neural
crest cells in the process of migration in vivo. Nature
282, 853–855.
Greenberg ME, Ziff EB & Greene LA (1986) Stimulation of neuronal acetylcholine receptors induces rapid
gene transcription. Science 234, 80–83.
Pich EM, Pagliusi SR, Tessari M, Talabot-Ayer D,
Hooft van Huijsduijnen R & Chiamulera C (1997)
Common neural substrates for the addictive
properties of nicotine and cocaine. Science 275,
83–86.
250 Harlan RE & Garcia MM (1998) Drugs of abuse and
immediate–early genes in the forebrain. Mol Neurobiol
16, 221–267.
251 Salminen O, Seppa T, Gaddnas H & Ahtee L (1999)
The effects of acute nicotine on the metabolism of
dopamine and the expression of Fos protein in striatal
and limbic brain areas of rats during chronic nicotine
infusion and its withdrawal. J Neurosci 19, 8145–8151.
252 Dunckley T & Lukas RJ (2003) Nicotine modulates
the expression of a diverse set of genes in the neuronal
SH-SY5Y cell line. J Biol Chem 278, 15633–15640.
253 Kumer SC & Vrana KE (1996) Intricate regulation of
tyrosine hydroxylase activity and gene expression.
J Neurochem 67, 443–462.
254 Gueorguiev VD, Zeman RJ, Meyer EM & Sabban EL
(2000) Involvement of alpha7 nicotinic acetylcholine
receptors in activation of tyrosine hydroxylase and
dopamine beta-hydroxylase gene expression in PC12
cells. J Neurochem 75, 1997–2005.
255 Gueorguiev VD, Zeman RJ, Hiremagalur B, Menezes
A & Sabban EL (1999) Differing temporal roles of
Ca2+ and cAMP in nicotine-elicited elevation of tyrosine hydroxylase mRNA. Am J Physiol 276, C54–C65.
256 Griffiths J & Marley PD (2001) Ca2+-dependent activation of tyrosine hydroxylase involves MEK1. Neuroreport 12, 2679–2683.
257 Gueorguiev VD, Cheng SY & Sabban EL (2006) Prolonged activation of cAMP-response element-binding
protein and ATF-2 needed for nicotine-triggered elevation of tyrosine hydroxylase gene transcription in
PC12 cells. J Biol Chem 281, 10188–10195.
258 Alkondon M & Albuquerque EX (2004) The nicotinic
acetylcholine receptor subtypes and their function in
the hippocampus and cerebral cortex. Prog Brain Res
145, 109–120.
259 Alkondon M & Albuquerque EX (2001) Nicotinic
acetylcholine receptor alpha7 and alpha4beta2 subtypes differentially control GABAergic input to CA1
neurons in rat hippocampus. J Neurophysiol 86, 3043–
3055.
260 Alkondon M, Pereira EF & Albuquerque EX (2003)
NMDA and AMPA receptors contribute to the nicotinic cholinergic excitation of CA1 interneurons in the
rat hippocampus. J Neurophysiol 90, 1613–1625.
261 Sweatt JD (2004) Mitogen-activated protein kinases in
synaptic plasticity and memory. Curr Opin Neurobiol
14, 311–317.
262 Chang KT & Berg DK (2001) Voltage-gated channels block nicotinic regulation of CREB phosphorylation and gene expression in neurons. Neuron 32,
855–865.
263 Nakayama H, Numakawa T, Ikeuchi T & Hatanaka
H (2001) Nicotine-induced phosphorylation of extracellular signal-regulated protein kinase and CREB in
PC12h cells. J Neurochem 79, 489–498.
FEBS Journal 274 (2007) 3799–3845 ª 2007 The Authors Journal compilation ª 2007 FEBS
3839
Nicotinic acetylcholine receptors
D. Kalamida et al.
264 Nakayama H, Shimoke K, Isosaki M, Satoh H, Yoshizumi M & Ikeuchi T (2006) Subtypes of neuronal
nicotinic acetylcholine receptors involved in nicotineinduced phosphorylation of extracellular signal-regulated protein kinase in PC12h cells. Neurosci Lett 392,
101–104.
265 Dajas-Bailador FA, Soliakov L & Wonnacott S (2002)
Nicotine activates the extracellular signal-regulated
kinase 1 ⁄ 2 via the alpha7 nicotinic acetylcholine
receptor and protein kinase A, in SH-SY5Y cells
and hippocampal neurones. J Neurochem 80,
520–530.
266 Hu M, Liu QS, Chang KT & Berg DK (2002) Nicotinic regulation of CREB activation in hippocampal
neurons by glutamatergic and nonglutamatergic pathways. Mol Cell Neurosci 21, 616–625.
267 Dajas-Bailador FA, Lima PA & Wonnacott S (2000)
The alpha7 nicotinic acetylcholine receptor subtype
mediates nicotine protection against NMDA excitotoxicity in primary hippocampal cultures through a Ca2+
dependent mechanism. Neuropharmacology 39, 2799–
2807.
268 Ferchmin PA, Perez D, Eterovic VA & de Vellis J
(2003) Nicotinic receptors differentially regulate
N-methyl-d-aspartate damage in acute hippocampal
slices. J Pharmacol Exp Ther 305, 1071–1078.
269 Zoli M, Picciotto MR, Ferrari R, Cocchi D & Changeux JP (1999) Increased neurodegeneration during ageing in mice lacking high-affinity nicotine receptors.
EMBO J 18, 1235–1244.
270 Oddo S & LaFerla FM (2006) The role of nicotinic
acetylcholine receptors in Alzheimer’s disease. J Physiol (Paris) 99, 172–179.
271 Wang HY, Lee DH, D’Andrea MR, Peterson PA,
Shank RP & Reitz AB (2000) Beta-amyloid(1–42)
binds to alpha7 nicotinic acetylcholine receptor with
high affinity. Implications for Alzheimer’s disease
pathology. J Biol Chem 275, 5626–5632.
272 Snyder EM, Nong Y, Almeida CG, Paul S, Moran T,
Choi EY, Nairn AC, Salter MW, Lombroso PJ, Gouras
GK et al. (2005) Regulation of NMDA receptor trafficking by amyloid-beta. Nat Neurosci 8, 1051–1058.
273 Corrigall WA, Coen KM, Adamson KL, Chow BL &
Zhang J (2000) Response of nicotine self-administration in the rat to manipulations of mu-opioid and
gamma-aminobutyric acid receptors in the ventral tegmental area. Psychopharmacology (Berlin) 149, 107–
114.
274 Laviolette SR & van der Kooy D (2003) Blockade of
mesolimbic dopamine transmission dramatically
increases sensitivity to the rewarding effects of nicotine
in the ventral tegmental area. Mol Psychiatry 8, 50–59.
275 Kalivas PW (1993) Neurotransmitter regulation of
dopamine neurons in the ventral tegmental area. Brain
Res Brain Res Rev 18, 75–113.
3840
276 Mansvelder HD, Keath JR & McGehee DS (2002)
Synaptic mechanisms underlie nicotine-induced
excitability of brain reward areas. Neuron 33, 905–
919.
277 Klink R, de Kerchove d’Exaerde A, Zoli M & Changeux JP (2001) Molecular and physiological diversity of
nicotinic acetylcholine receptors in the midbrain dopaminergic nuclei. J Neurosci 21, 1452–1463.
278 Laviolette SR & van der Kooy D (2004) The neurobiology of nicotine addiction: bridging the gap from
molecules to behaviour. Nat Rev Neurosci 5, 55–65.
279 McGehee DS, Heath MJ, Gelber S, Devay P & Role
LW (1995) Nicotine enhancement of fast excitatory
synaptic transmission in CNS by presynaptic receptors.
Science 269, 1692–1696.
280 Girod R, Barazangi N, McGehee D & Role LW
(2000) Facilitation of glutamatergic neurotransmission
by presynaptic nicotinic acetylcholine receptors. Neuropharmacology 39, 2715–2725.
281 Kaiser S & Wonnacott S (2000) Alpha-bungarotoxinsensitive nicotinic receptors indirectly modulate
[3H]dopamine release in rat striatal slices via glutamate
release. Mol Pharmacol 58, 312–318.
282 Oakman SA, Faris PL, Kerr PE, Cozzari C & Hartman BK (1995) Distribution of pontomesencephalic
cholinergic neurons projecting to substantia nigra differs significantly from those projecting to ventral tegmental area. J Neurosci 15, 5859–5869.
283 Blaha CD, Allen LF, Das S, Inglis WL, Latimer MP,
Vincent SR & Winn P (1996) Modulation of dopamine
efflux in the nucleus accumbens after cholinergic stimulation of the ventral tegmental area in intact, pedunculopontine tegmental nucleus-lesioned, and laterodorsal
tegmental nucleus lesioned rats. J Neurosci 16, 714–
722.
284 Forster GL & Blaha CD (2000) Laterodorsal tegmental stimulation elicits dopamine efflux in the rat nucleus accumbens by activation of acetylcholine and
glutamate receptors in the ventral tegmental area. Eur
J Neurosci 12, 3596–3604.
285 Pidoplichko VI, DeBiasi M, Williams JT & Dani JA
(1997) Nicotine activates and desensitizes midbrain
dopamine neurons. Nature 390, 401–404.
286 Whaley K, Lappin D & Barkas T (1981) C2 synthesis
by human monocytes is modulated by a nicotinic cholinergic receptor. Nature 293, 580–583.
287 Skok M, Grailhe R, Agenes F & Changeux JP (2006)
The role of nicotinic acetylcholine receptors in lymphocyte development. J Neuroimmunol 171, 86–98.
288 Sharma G & Vijayaraghavan S (2002) Nicotinic receptor signaling in nonexcitable cells. J Neurobiol 53, 524–
534.
289 Gahring LC & Rogers SW (2005) Neuronal nicotinic
acetylcholine receptor expression and function on nonneuronal cells. Aaps J 7, 885–894.
FEBS Journal 274 (2007) 3799–3845 ª 2007 The Authors Journal compilation ª 2007 FEBS
Nicotinic acetylcholine receptors
D. Kalamida et al.
290 Wang Y, Pereira EF, Maus AD, Ostlie NS, Navaneetham D, Lei S, Albuquerque EX & Conti-Fine BM
(2001) Human bronchial epithelial and endothelial cells
express alpha7 nicotinic acetylcholine receptors. Mol
Pharmacol 60, 1201–1209.
291 Tournier JM, Maouche K, Coraux C, Zahm JM, Cloez-Tayarani I, Nawrocki-Raby B, Bonnomet A, Burlet
H, Lebargy F, Polette M et al. (2006) alpha3alpha5beta2-Nicotinic acetylcholine receptor contributes to
the wound repair of the respiratory epithelium by
modulating intracellular calcium in migrating cells.
Am J Pathol 168, 55–68.
292 Chernyavsky AI, Arredondo J, Marubio LM &
Grando SA (2004) Differential regulation of
keratinocyte chemokinesis and chemotaxis through
distinct nicotinic receptor subtypes. J Cell Sci 117,
5665–5679.
293 Borovikova LV, Ivanova S, Zhang M, Yang H,
Botchkina GI, Watkins LR, Wang H, Abumrad N,
Eaton JW & Tracey KJ (2000) Vagus nerve stimulation attenuates the systemic inflammatory response to
endotoxin. Nature 405, 458–462.
294 Wang H, Yu M, Ochani M, Amella CA, Tanovic M,
Susarla S, Li JH, Yang H, Ulloa L & Al-Abed Y
(2003) Nicotinic acetylcholine receptor alpha7 subunit
is an essential regulator of inflammation. Nature 421,
384–388.
295 Wang H, Liao H, Ochani M, Justiniani M, Lin X,
Yang L, Al-Abed Y, Metz C, Miller EJ, Tracey KJ
et al. (2004) Cholinergic agonists inhibit HMGB1
release and improve survival in experimental sepsis.
Nat Med 10, 1216–1221.
296 Ulloa L (2005) The vagus nerve and the nicotinic antiinflammatory pathway. Nat Rev Drug Discov 4, 673–
684.
297 Lotze MT & Tracey KJ (2005) High-mobility group
box 1 protein (HMGB1): nuclear weapon in the
immune arsenal. Nat Rev Immunol 5, 331–342.
298 Guarini S, Altavilla D, Cainazzo MM, Giuliani D,
Bigiani A, Marini H, Squadrito G, Minutoli L,
Bertolini A, Marini R et al. (2003) Efferent vagal fibre
stimulation blunts nuclear factor-kappaB activation
and protects against hypovolemic hemorrhagic shock.
Circulation 107, 1189–1194.
299 Saeed RW, Varma S, Peng-Nemeroff T, Sherry B,
Balakhaneh D, Huston J, Tracey KJ, Al-Abed Y &
Metz CN (2005) Cholinergic stimulation blocks
endothelial cell activation and leukocyte recruitment
during inflammation. J Exp Med 201, 1113–1123.
300 de Jonge WJ, van der Zanden EP, The FO, Bijlsma
MF, van Westerloo DJ, Bennink RJ, Berthoud HR,
Uematsu S, Akira S, van den Wijngaard RM et al.
(2005) Stimulation of the vagus nerve attenuates
macrophage activation by activating the Jak2-STAT3
signaling pathway. Nat Immunol 6, 844–851.
301 Villablanca AC (1998) Nicotine stimulates DNA
synthesis and proliferation in vascular endothelial cells
in vitro. J Appl Physiol 84, 2089–2098.
302 Macklin KD, Maus AD, Pereira EF, Albuquerque EX
& Conti-Fine BM (1998) Human vascular endothelial
cells express functional nicotinic acetylcholine receptors. J Pharmacol Exp Ther 287, 435–439.
303 Heeschen C, Weis M, Aicher A, Dimmeler S & Cooke
JP (2002) A novel angiogenic pathway mediated by
non-neuronal nicotinic acetylcholine receptors. J Clin
Invest 110, 527–536.
304 Heeschen C, Jang JJ, Weis M, Pathak A, Kaji S, Hu
RS, Tsao PS, Johnson FL & Cooke JP (2001) Nicotine
stimulates angiogenesis and promotes tumor growth
and atherosclerosis. Nat Med 7, 833–839.
305 Dasgupta P, Kinkade R, Joshi B, Decook C, Haura E
& Chellappan S (2006) Nicotine inhibits apoptosis
induced by chemotherapeutic drugs by up-regulating
XIAP and survivin. Proc Natl Acad Sci USA 103,
6332–6337.
306 Dasgupta P, Rastogi S, Pillai S, Ordonez-Ercan D,
Morris M, Haura E & Chellappan S (2006) Nicotine
induces cell proliferation by b-arrestin-mediated activation of Src and Rb–Raf-1 pathways. J Clin Invest 116,
2208–2217.
307 Peng JH, Fryer JD, Hurst RS, Schroeder KM,
George AA, Morrissy S, Groppi VE, Leonard SS &
Lukas RJ (2005) High-affinity epibatidine binding of
functional, human alpha7-nicotinic acetylcholine
receptors stably and heterologously expressed de
novo in human SH-EP1 cells. J Pharmacol Exp Ther
313, 24–35.
308 Zhao L, Kuo Y-P, George A, Peng J-H, Purandare
MS, Schroeder K, Lukas R & Wu J (2003) Functional
properties of homomeric, human alpha 7-nicotinic
acetylcholine receptors heterologously expressed in the
SH-EP1 human epithelial cell line. J Pharmacol Exp
Ther 305, 1132–1141.
309 Aztiria EM, Sogayar MC & Barrantes FJ (2000)
Expression of a neuronal nicotinic acetylcholine receptor in insect and mammalian host cell systems. Neurochem Res 25, 171–180.
310 Sweileh W, Wenberg K, Xu J, Forsayeth J, Hardy S &
Loring RH (2000) Multistep expression and assembly
of neuronal nicotinic receptors is both host-cell- and
receptor-subtype-dependent. Brain Res Mol Brain Res
75, 293–302.
311 Person AM, Bills KL, Liu H, Botting SK, Lindstrom J
& Wells GB (2005) Extracellular domain nicotinic
acetylcholine receptors formed by alpha4 and beta2
subunits. J Biol Chem 280, 39990–40002.
312 Wells GB, Anand R, Wang F & Lindstrom J (1998)
Water-soluble nicotinic acetylcholine receptor formed
by alpha7 subunit extracellular domains. J Biol Chem
273, 964–973.
FEBS Journal 274 (2007) 3799–3845 ª 2007 The Authors Journal compilation ª 2007 FEBS
3841
Nicotinic acetylcholine receptors
D. Kalamida et al.
313 Zhou Y, Nelson ME, Kuryatov A, Choi C, Cooper J
& Lindstrom J (2003) Human alpha4beta2 acetylcholine receptors formed from linked subunits. J Neurosci
23, 9004–9015.
314 Groot-Kormelink PJ, Broadbent S, Beato M & Sivilotti LG (2006) Constraining the expression of nicotinic
acetylcholine receptors by using pentameric constructs.
Mol Pharmacol 69, 558–563.
315 Craig PJ, Bose S, Zwart R, Beattie RE, Folly EA,
Johnson LR, Bell E, Evans NM, Benedetti G, Pearson
KH et al. (2004) Stable expression and characterisation
of a human alpha 7 nicotinic subunit chimera: a tool
for functional high-throughput screening. Eur J Pharmacol 502, 31–40.
316 Fischer M, Corringer PJ, Schott K, Bacher A &
Changeux JP (2001) A method for soluble overexpression of the alpha7 nicotinic acetylcholine receptor
extracellular domain. Proc Natl Acad Sci USA 98,
3567–3570.
317 Utkin YN, Kukhtina VV, Kryukova EV, Chiodini F,
Bertrand D, Methfessel C & Tsetlin VI (2001) ‘Weak
toxin’ from Naja kaouthia is a nontoxic antagonist of
alpha 7 and muscle-type nicotinic acetylcholine receptors. J Biol Chem 276, 15810–15815.
318 Conti-Fine BM, Milani M & Kaminski HJ (2006)
Myasthenia gravis: past, present, and future. J Clin
Invest 116, 2843–2854.
319 Vincent A, Palaceb J & Hilton J (2001) Myasthenia
gravis. Lancet 357, 2122–2128.
320 Engel AG & Sine SM (2005) Current understanding of
congenital myasthenic syndromes. Curr Opin Pharmacol 5, 308–321.
321 Mossman S, Vincent A & Newson-Davis J (1986)
Myasthenia gravis without acetylcholine receptor antibody: a distinct disease entity. Lancet 18, 116–119.
322 Hoch W, McConville J & Helms S (2001) Autoantibodies to the receptor tyrosine kinase MuSK in
patients with myasthenia gravis without acetylcholine
receptor antibodies. Nat Med 7, 365–368.
323 Romi F, Skeie GO, Gilhus NE & Aarli JA (2005) Striational antibodies in myasthenia gravis: reactivity and
possible clinical significance. Arch Neurol 62, 442–446.
324 Poulas K, Tsibri E, Kokla A, Papanastasiou D, Tsouloufis T, Marinou M, Tsantili P, Papapetropoulos T &
Tzartos SJ (2001) Epidemiology of seropositive myasthenia gravis in Greece. J Neurol Neurosurg Psychiatry
71, 352–356.
325 Vincent A, Beeson D & Lang B (2000) Molecular
targets for autoimmune and genetic disorders of neuromuscular transmission. Eur J Biochem 267, 6717–
6728.
326 Patrick J & Lindstrom J (1973) Autoimmune response
to acetylcholine receptor. Science 180, 871–872.
327 Lindstrom JM (1999) Experimental induction and
treatment of myasthenia gravis. In Myasthenia Gravis
3842
328
329
330
331
332
333
334
335
336
337
338
and Myasthenic Disorders (Engel A, ed.), pp. 111–130.
Oxford University Press, Oxford.
Lindstrom J, Engel A, Seybold M, Lennon V & Lambert E (1976) Pathological mechanisms in EAMG II:
passive transfer of experimental autoimmune myasthenia gravis in rats with anti-acetylcholine receptor antibodies. J Exp Med 144, 739–753.
Tzartos S, Hochschwender S, Vasquez P & Lindstrom
J (1987) Passive transfer of experimental autoimmune
myasthenia gravis by monoclonal antibodies to the
main immunogenic region of the acetylcholine receptor. J Neuroimmunol 15, 185–194.
Weinberg C & Hall Z (1979) Antibodies from patients
with myasthenia gravis recognize determinants unique
to extrajunctional acetylcholine receptors. Proc Natl
Acad Sci USA 76, 504–508.
Mesnard-Rouiller L, Bismith J, Wakkach A, PoeaGuyon S & Berrih-Aknin S (2004) Thymic myoid cells
express high levels of muscle genes. J Neuroimmunol
148, 97–105.
Levinson AI, Song D, Gaulton G & Zheng Y (2004)
The intrathymic pathogenesis of myasthenia gravis.
Clin Dev Immunol 11, 215–220.
Colombara M, Antonini V, Riviera AP, Mainiero F,
Strippoli R, Merola M, Fracasso G, Poffe O, Brutti
N, Tridente G et al. (2005) Constitutive activation of
p38 and ERK1 ⁄ 2 MAPKs in epithelial cells of myasthenic thymus leads to IL-6 and RANTES overexpression: effects on survival and migration of peripheral T
and B cells. J Immunol 175, 7021–7028.
Poea-Guyon S, Christadoss P, Le Panse R, Guyon T,
De Baets M, Wakkach A, Bidault J, Tzartos S & Berrih-Aknin S (2005) Effects of cytokines on acetylcholine receptor expression: implications for myasthenia
gravis. J Immunol 174, 5941–5949.
Bernasconi P, Barberis M, Baggi F, Passerini L, Cannone M, Arnoldi E, Novellino L, Cornelio F & Mantegazza R (2005) Increased toll-like receptor 4
expression in thymus of myasthenic patients with
thymitis and thymic involution. Am J Pathol 167, 129–
139.
Inoue M, Okumura M, Miyoshi S, Shiono H, Fukuhara K, Kadota Y, Shirakura R & Matsuda H (1999)
Impaired expression of MHC class II molecules in
response to interferon-gamma (IFN-gamma) on
human thymoma neoplastic epithelial cells. Clin Exp
Immunol 117, 1–7.
Sommer N, Willcox N, Harcourt GC & Newson-Davis
J (1990) Myasthenic thymus and thymoma are selectively enriched in acetylcholine receptor-reactive T
cells. Ann Neurol 28, 312–319.
Nagvekar N, Moody AM, Moss P, Roxanis I, Curnow
J, Beeson D, Pantic N, Newson-Davis J, Vincent A &
Willcox N (1998) A pathogenetic role for the thymoma
in myasthenia gravis. Autosensitization of IL-4- produ-
FEBS Journal 274 (2007) 3799–3845 ª 2007 The Authors Journal compilation ª 2007 FEBS
Nicotinic acetylcholine receptors
D. Kalamida et al.
339
340
341
342
343
344
345
346
347
348
349
350
cing T-cell clones recognizing extracellular acetylcholine receptor epitopes presented by minority class II isotypes. J Clin Invest 101, 2268–2277.
Maiti PK, Feferman T, Im SH, Souroujon MC &
Fuchs S (2004) Immunosuppression of rat myasthenia gravis by oral administration of a syngeneic acetylcholine receptor fragment. J Neuroimmunol 152,
112–120.
Cohen-Kaminsky S & Jambou F (2005) Prospects for
a T-cell receptor vaccination against myasthenia gravis.
Expert Rev Vaccines 4, 473–492.
Tzartos SJ, Sophianos D & Efthimiadis A (1985) Role
of the main immunogenic region of acetylcholine
receptor in myasthenia gravis. An Fab monoclonal
antibody protects against antigenic modulation by
human sera. J Immunol 134, 2343–2349.
Papanastasiou D, Poulas K, Kokla A & Tzartos SJ
(2000) Prevention of passively transferred experimental
autoimmune myasthenia gravis by Fab fragments of
monoclonal antibodies directed against the main
immunogenic region of the acetylcholine receptor.
J Neuroimmunol 104, 124–132.
Fostieri E, Tzartos SJ, Berrih-Aknin S, Beeson D &
Mamalaki A (2005) Isolation of potent human Fab
fragments against a novel highly immunogenic region
on human muscle acetylcholine receptor, which protect
the receptor from myasthenic autoantibodies. Eur J
Immunol 35, 632–643.
Graus YF, de Baets MH, Parren PW, Berrih-Aknin S,
Wokke J, van Breda Vriesman PJ & Burton DR
(1997) Human anti-nicotinic acetylcholine receptor
recombinant Fab fragments isolated from thymusderived phage display libraries from myasthenia gravis
patients reflect predominant specificities in serum and
block the action of pathogenic serum antibodies.
J Immunol 158, 1919–1929.
Aricha R, Feferman T, Souroujon MC & Fuchs S
(2006) Overexpression of phosphodiesterases in experimental autoimmune myasthenia gravis: suppression of
disease by a phosphodiesterase inhibitor. FASEB J 20,
374–376.
Hantai D, Richard P, Koenig J & Eymand B (2004)
Congenital myasthenic syndromes. Curr Opin Neurol
17, 539–551.
Ohno K & Engel AG (2004) Congenital myasthenic
syndromes: gene mutations. Neuromusc Disord 14,
117–122.
Engel AG (1994) Congenital myasthenic syndromes.
Neurol Clinics North Am 12, 401–436.
Engel AG (1999) Congenital myasthenic syndromes.
J Child Neurol 14, 38–41.
Beeson D, Hantai D, Lochmuller H & Engel AG
(2005) 126th International Workshop: congenital
myasthenic syndromes, 24–26 September 2004, Naarden, the Netherlands. Neuromusc Disord 15, 498–512.
351 Milone M, Wang HL, Ohno K, Fukudome T, Pruitt
JN, Bren N, Sine SM & Engel AG (1997) Slow-channel syndrome caused by enhanced activation, desensitization, and agonist binding affinity due to mutation in
the M2 domain of the acetylcholine receptor alpha
subunit. J Neurosci 17, 5651–5665.
352 Croxen R, Newland C, Beeson D, Oosterhuis H,
Chauplanaz G, Vincent A & Newsom-Davis J (1997)
Mutations in different functional domains of the
human muscle acetylcholine receptor e subunit in
patients with the slow-channel congenital myasthenic
syndrome. Hum Mol Genet 6, 767–774.
353 Wang HL, Auerbach A, Bren N, Ohno K, Engel AG
& Sine SM (1997) Mutation in the M1 domain of the
acetylcholine receptor alpha subunit decreases the rate
of agonist dissociation. J Gen Physiol 109, 757–766.
354 Croxen R, Hatton C, Shelley C, Brydson M, Chauplannaz G, Oosterhuis H, Vincent A, Newsom-Davis
J, Colquhoun D & Beeson D (2002) Recessive inheritance and variable penetrance of slow-channel congenital myasthenic syndromes. Neurology 59, 162–
168.
355 Engel AG, Ohno K, Shen XM & Sine SM (2003) Congenital myasthenic syndromes: multiple molecular targets at the neuromuscular junction. Ann NY Acad Sci
998, 138–160.
356 Akabas MH, Kaufmann C, Archdeacon P & Karlin A
(1994) Identification of acetylcholine receptor channellining residues in the entire M2 segment of the alpha
subunit. Neuron 13, 919–927.
357 Wang HL, Milone M, Ohno K, Shen XM, Tsujino A,
Batocchi AP, Tonali P, Brengman JM, Engel AG &
Sine SM (1999) Acetylcholine receptor M3 domain:
stereochemical and volume contributions to channel
gating. Nat Neurosci 2, 226–233.
358 Milone M, Wang HL, Ohno K, Prince RJ, Shen XM,
Brengman JM, Griggs RC & Engel AG (1998) Mode
switching kinetics produced by a naturally occurring
mutation in the cytoplasmic loop of the human acetylcholine receptor subunit. Neuron 20, 575–588.
359 Brownlow S, Webster R, Croxen R, Brydson M, Neville B, Lin JP, Vincent A, Newsom-Davis J & Beeson
D (2001) Acetylcholine receptor subunit mutations
underlie a fast-channel myasthenic syndrome and
arthrogryposis multiplex congenita. J Clin Invest 108,
125–130.
360 Shen XM, Ohno K, Milone M, Brengman JM, Spilsbury PR & Engel AG (2001) Fast-channel syndrome.
Neurology 56, A60.
361 Middleton L, Ohno K, Christodoulou K, Brengman J,
Milone M, Neocleous V, Serdaroglu P, Deymeer F,
Ozdemir C, Mubaidin A et al. (1999) Chromosome
17p-linked myasthenias stem from defects in the acetylcholine receptor epsilon-subunit gene. Neurology 53,
1076–1082.
FEBS Journal 274 (2007) 3799–3845 ª 2007 The Authors Journal compilation ª 2007 FEBS
3843
Nicotinic acetylcholine receptors
D. Kalamida et al.
362 Abicht A, Stucka R, Karcagi V, Herczegfalvi A, Horvath R, Mortier W, Schara U, Ramaekers V, Jost W,
Brunner J et al. (1999) A common mutation
(e1267delG) in congenital myasthenic patients of Gipsy
ethnic origin. Neurology 53, 1564–1569.
363 Croxen R, Newland C, Betty M, Vincent A, NewsomDavis J & Beeson D (1999) Novel functional 3-subunit
polypeptide generated by a single nucleotide deletion
in acetylcholine receptor deficiency congenital myasthenic syndrome. Ann Neurol 46, 639–647.
364 Nichols P, Croxen R, Vincent A, Rutter R, Hutchinson M, Newson-Davis J & Beeson D (1999) Mutation
in the AChR e-subunit promoter in CMS. Ann Neurol
45, 439–443.
365 Ohno K, Anlar B & Engel AG (1999) CMS caused by
a mutation in the Ets-binding site of the promoter
region of the acetylcholine receptor e-subunit gene.
Neuromusc Disord 9, 131–135.
366 Ealing J, Webster R, Browlow S, Abdelgany A,
Oosterhuis H, Muntoni F, Vaux DJ, Vincent A &
Beeson D (2002) Mutations in congenital myasthenic
syndromes reveal an e subunit C-terminal cysteine,
C470, crucial for maturation and surface expression of
adult AChR. Hum Mol Genet 11, 3087–3096.
367 Ohno K, Brengman J, Tsujino A & Engel AG
(1998) Human endplate AChE deficiency caused by
mutations in the collagen like tail subunit of the
asymmetric enzyme. Proc Natl Acad Sci USA 95,
9654–9659.
368 Morar B, Gresham D, Angelishera D, Tourner I, Gooding R, Guergueltchera V & Schmidt C (2004) Mutation history of the Roma ⁄ Gypsies. Am J Hum Genet
75, 596–609.
369 Harper CM & Engel AG (2000) Treatment of 31 congenital myasthenic syndrome patients with 3,4-diaminopyridine. Neurology 54, A395.
370 Ohno K, Engel AG, Shen XM, Selcen D, Brengman J,
Harper CM, Tsujino A & Milone M (2002) Rapsyn
mutations in humans cause endplate acetylcholinereceptor deficiency and myasthenic syndrome. Am J
Hum Genet 70, 875–885.
371 Chevessier F, Faraut B, Ravel-Chapuis A, Richard P,
Gaudon K, Bauche S, Prioleau C, Herbst R, Goillot
E, Ioos C et al. (2004) MuSK, a new target for mutations causing congenital myasthenic syndrome. Hum
Mol Genet 13, 3229–3240.
372 Blennow K, de Leon MJ & Zetterberg H (2006) Alzheimer’s disease. Lancet 368, 387–403.
373 Terry AV Jr & Buccafusco JJ (2003) The cholinergic
hypothesis of age and Alzheimer’s disease-related cognitive deficits: recent challenges and their implications
for novel drug development. J Pharmacol Exp Ther
306, 821–827.
374 Whitehouse PJ, Martino AM, Antuono PG, Lowenstein PR, Coyle JT, Price DL & Kellar KJ (1986)
3844
375
376
377
378
379
380
381
382
383
384
385
386
Nicotinic acetylcholine binding sites in Alzheimer’s disease. Brain Res 371, 146–151.
Kihara T, Shimohama S, Sawada H, Honda K, Nakamizo T, Shibasaki H, Kume T & Akaike A (2001)
Alpha 7 nicotinic receptor transduces signals to
phosphatidylinositol 3-kinase to block Abeta-amyloid-induced neurotoxicity. J Biol Chem 276, 13541–
13546.
Wang HY, Lee DH, Davis CB & Shank RP (2000)
Amyloid peptide Abeta(1–42) binds selectively and
with picomolar affinity to alpha7 nicotinic acetylcholine receptors. J Neurochem 75, 1155–1161.
Lamb PW, Melton MA & Yakel JL (2005) Inhibition
of neuronal nicotinic acetylcholine receptor channels
expressed in Xenopus oocytes by beta-amyloid1–42
peptide. J Mol Neurosci 27, 13–21.
Lee PN (1994) Smoking and Alzheimer’s disease: a
review of the epidemiological evidence. Neuroepidemiology 13, 131–144.
de Leon J, Dadvand M, Canuso C, White AO, Stanilla
JK & Simpson GM (1995) Schizophrenia and smoking: an epidemiological survey in a state hospital. Am
J Psychiatry 152, 453–455.
Adler LE, Hoffer LD, Wiser A & Freedman R (1993)
Normalization of auditory physiology by cigarette
smoking in schizophrenic patients. Am J Psychiatry
150, 1856–1861.
Leonard S, Gault J, Hopkins J, Logel J, Vianzon R,
Short M, Drebing C, Berger R, Venn D, Sirota P et al.
(2002) Association of promoter variants in the alpha7
nicotinic acetylcholine receptor subunit gene with an
inhibitory deficit found in schizophrenia. Arch Gen
Psychiatry 59, 1085–1096.
Freedman R, Hall M, Adler LE & Leonard S (1995)
Evidence in post-mortem brain tissue for decreased
numbers of hippocampal nicotinic receptors in schizophrenia. Biol Psychiatry 38, 22–33.
Barkley RA, Fischer M, Smallish L & Fletcher K
(2002) The persistence of attention-deficit ⁄ hyperactivity
disorder into young adulthood as a function of reporting source and definition of disorder. J Abnorm
Psychol 111, 279–289.
Pomerleau OF, Downey KK, Stelson FW & Pomerleau CS (1995) Cigarette smoking in adult patients
diagnosed with attention deficit hyperactivity disorder.
J Substance Abuse 7, 373–378.
Blondel A, Sanger DJ & Moser PC (2000) Characterisation of the effects of nicotine in the five-choice serial
reaction time task in rats: antagonist studies. Psychopharmacology (Berlin) 149, 293–305.
Levin ED, Christopher NC, Weaver T, Moore J &
Brucato F (1999) Ventral hippocampal ibotenic acid
lesions block chronic nicotine-induced spatial working
memory improvement in rats. Brain Res Cogn Brain
Res 7, 405–410.
FEBS Journal 274 (2007) 3799–3845 ª 2007 The Authors Journal compilation ª 2007 FEBS
Nicotinic acetylcholine receptors
D. Kalamida et al.
387 Grottick AJ & Higgins GA (2000) Effect of subtype
selective nicotinic compounds on attention as assessed
by the five-choice serial reaction time task. Behav Brain
Res 117, 197–208.
388 Levin ED & Rezvani AH (2000) Development of nicotinic drug therapy for cognitive disorders. Eur J Pharmacol 393, 141–146.
389 Wilens TE, Biederman J, Spencer TJ, Bostic J, Prince
J, Monuteaux MC, Soriano J, Fine C, Abrams A,
Rater M et al. (1999) A pilot controlled clinical trial of
ABT-418, a cholinergic agonist, in the treatment of
adults with attention deficit hyperactivity disorder.
Am J Psychiatry 156, 1931–1937.
390 Ueno K, Togashi H, Matsumoto M, Ohashi S, Saito
H & Yoshioka M (2002) Alpha4beta2 nicotinic acetylcholine receptor activation ameliorates impairment of
spontaneous alternation behavior in stroke-prone
spontaneously hypertensive rats, an animal model of
attention deficit hyperactivity disorder. J Pharmacol
Exp Ther 302, 95–100.
391 Picciotto MR & Zoli M (2002) Nicotinic receptors in
aging and dementia. J Neurobiol 53, 641–655.
392 Tanner CM, Goldman SM, Aston DA, Ottman R,
Ellenberg J, Mayeux R & Langston JW (2002)
Smoking and Parkinson’s disease in twins. Neurology
58, 581–588.
393 Schneider JS, Van Velson M, Menzaghi F & Lloyd
GK (1998) Effects of the nicotinic acetylcholine receptor agonist SIB-1508Y on object retrieval performance
in MPTP-treated monkeys: comparison with levodopa
treatment. Ann Neurol 43, 311–317.
394 Xie YX, Bezard E & Zhao BL (2005) Investigating the
receptor-independent neuroprotective mechanisms of
nicotine in mitochondria. J Biol Chem 280, 32405–
32412.
395 Grando SA (2000) Autoimmunity to keratinocyte acetylcholine receptor in pemphigus. Dermatology 201,
290–295.
396 Grando SA (2001) Receptor-mediated action of nicotine in human skin. Int J Dermatol 40, 691–693.
397 Vu TN, Lee TX, Ndoye A, Shultz LD, Pittelkow MR,
Dahl MV, Lynch PJ & Grando SA (1998) The pathophysiological significance of nondesmoglein targets of
pemphigus autoimmunity. Development of antibodies
against keratinocyte cholinergic receptors in patients
with pemphigus vulgaris and pemphigus foliaceus.
Arch Dermatol 134, 971–980.
398 Low PA, Vernino S & Suarez G (2003) Autonomic
dysfunction in peripheral nerve disease. Muscle Nerve
27, 646–661.
399 Vernino S, Adamski J, Kryzer TJ, Fealey RD & Lennon VA (1998) Neuronal nicotinic ACh receptor antibody in subacute autonomic neuropathy and cancerrelated syndromes. Neurology 50, 1806–1813.
400 Vernino S, Low PA, Fealey RD, Stewart JD, Farrugia
G & Lennon VA (2000) Autoantibodies to ganglionic
acetylcholine receptors in autoimmune autonomic neuropathies. N Engl J Med 343, 847–855.
401 Lennon VA, Ermilov LG, Szurszewski JH & Vernino
S (2003) Immunization with neuronal nicotinic acetylcholine receptor induces neurological autoimmune disease. J Clin Invest 111, 907–913.
402 Vernino S, Ermilov LG, Sha L, Szurszewski JH, Low
PA & Lennon VA (2004) Passive transfer of autoimmune autonomic neuropathy to mice. J Neurosci 24,
7037–7042.
403 Steinlein OK, Mulley JC, Propping P, Wallace RH,
Phillips HA, Sutherland GR, Scheffer IE & Berkovic
SF (1995) A missense mutation in the neuronal nicotinic acetylcholine receptor alpha 4 subunit is associated with autosomal dominant nocturnal frontal lobe
epilepsy. Nat Genet 11, 201–203.
404 Lee M, Martin-Ruiz C, Graham A, Court J, Jaros E,
Perry R, Iversen P, Bauman M & Perry E (2002) Nicotinic receptor abnormalities in the cerebellar cortex in
autism. Brain 125, 1483–1495.
405 Martin-Ruiz CM, Lee M, Perry RH, Baumann M,
Court JA & Perry EK (2004) Molecular analysis of
nicotinic receptor expression in autism. Brain Res Mol
Brain Res 123, 81–90.
406 Buisson B & Bertrand D (2002) Nicotine addiction:
the possible role of functional upregulation. Trends
Pharmacol Sci 23, 130–136.
407 Balfour DJK (2004) The neurobiology of tobacco
dependence: a preclinical perspective on the role of
the dopamine projections to the nucleus accumbens.
Nicotine Tob Res 6, 899–912.
408 Sciamanna MA, Griesmann GE, Williams CL &
Lennon VA (1997) Nicotinic acetylcholine receptors
of muscle and neuronal (alpha7) types coexpressed in
a small cell lung carcinoma. J Neurochem 69,
2302–2311.
409 Sandall DW, Satkunanathan N, Keays DA, Polidano
MA, Liping X, Pham V, Down JG, Khalil Z, Livett
BG & Gayler KR (2003) A novel alpha-conotoxin
identified by gene sequencing is active in suppressing
the vascular response to selective stimulation of sensory nerves in vivo. Biochemistry 42, 6904–6911.
410 Tonstad S (2007) Varenicline for smoking cessation.
Expert Rev Neurother 7, 121–127.
FEBS Journal 274 (2007) 3799–3845 ª 2007 The Authors Journal compilation ª 2007 FEBS
3845