ArticlePDF AvailableLiterature Review

Understanding the Bases of Function and Modulation of α7 Nicotinic Receptors: Implications for Drug Discovery

Authors:

Abstract

The nicotinic acetylcholine receptor (nAChR) belongs to a superfamily of pentameric ligand gated ion channels involved in many physiological and pathological processes. Among nAChRs, receptors comprised of the α7 subunit are unique due to their high Ca(2+) permeability and fast desensitization. nAChR agonists elicit a transient ion flux response that is further sustained by the release of calcium from intracellular sources. Due to the dual ionotropic/metabotropic nature of α7 receptors, signaling pathways are activated. The α7 subunit is highly expressed in the nervous system, mostly in regions implicated in cognition and memory, and has therefore attracted attention as a novel drug target. Additionally, its dysfunction is associated with several neuropsychiatric and neurological disorders, such as schizophrenia and Alzheimer's disease. α7 is also expressed in non-neuronal cells, particularly immune cells, where it plays a role in immunity, inflammation and neuroprotection. Thus, α7 potentiation has emerged as a therapeutic strategy for several neurological and inflammatory disorders. With unique activation properties, the receptor is a sensitive drug target carrying different potential binding sites for chemical modulators, particularly agonists and positive allosteric modulators. Although macroscopic and single-channel recordings have provided significant information underlying molecular mechanisms and binding sites of modulatory compounds, we know just the tip of the iceberg. Further concerted efforts are necessary to effectively exploit α7 as a drug target for each pathological situation. In this article, we focus mainly on the molecular basis of activation and drug modulation of α7, key pillars for rational drug design.
1521-0111/90/3/288299$25.00 http://dx.doi.org/10.1124/mol.116.104240
MOLECULAR PHARMACOLOGY Mol Pharmacol 90:288299, September 2016
Copyright ª2016 by The American Society for Pharmacology and Experimental Therapeutics
MINIREVIEWA LATIN AMERICAN PERSPECTIVE ON ION CHANNELS
Understanding the Bases of Function and Modulation of a7
Nicotinic Receptors: Implications for Drug Discovery
Jeremías Corradi and Cecilia Bouzat
Instituto de Investigaciones Bioquímicas de Bahía Blanca, Universidad Nacional del Sur, CONICET/UNS, Bahía Blanca,
Argentina
Received March 7, 2016; accepted May 5, 2016
ABSTRACT
The nicotinic acetylcholine receptor (nAChR) belongs to a
superfamily of pentameric ligand-gated ion channels involved
in many physiologic and pathologic processes. Among nAChRs,
receptors comprising the a7 subunit are unique because of their
high Ca
21
permeability and fast desensitization. nAChR agonists
elicit a transient ion flux response that is further sustained by the
release of calcium from intracellular sources. Owing to the dual
ionotropic/metabotropic nature of a7 receptors, signaling path-
ways are activated. The a7 subunit is highly expressed in the
nervous system, mostly in regions implicated in cognition and
memory and has therefore attracted attention as a novel drug
target. Additionally, its dysfunction is associated with several
neuropsychiatric and neurologic disorders, such as schizophre-
nia and Alzheimers disease. a7 is also expressed in non-neuronal
cells, particularly immune cells, where it plays a role in immunity,
inflammation, and neuroprotection. Thus, a7 potentiation has
emerged as a therapeutic strategy for several neurologic and
inflammatory disorders. With unique activation properties, the
receptor is a sensitive drug target carrying different potential
binding sites for chemical modulators, particularly agonists and
positive allosteric modulators. Although macroscopic and single-
channel recordings have provided significant information about
the underlying molecular mechanisms and binding sites of mod-
ulatory compounds, we know just the tip of the iceberg. Further
concerted efforts are necessary to effectively exploit a7 as a drug
target for each pathologic situation. In this article, we focus mainly
on the molecular basis of activation and drug modulation of a7,
key pillars for rational drug design.
Introduction
Nicotine has been a key molecule for the advancement of
pharmacology since the beginning of the 20th century, when
Langley (1905), through fundamental experiments, concluded
that muscle contraction was mediated by a receptive sub-
stancepresent on the muscle. The muscle nicotinic acetylcho-
line receptor (nAChR) was thus a pillar in the discovery of
neurotransmitter receptors (Langley, 1905). Still, it was not until
1970 that the first neurotransmitter receptor, nAChR, was
identified (Changeux et al., 1970; Miledi and Potter, 1971). With
the later advent of the molecular biology revolution in the 1980s,
the nAChR family was first identified and an extended family of
homologous pentameric receptors was revealed (Patrick et al.,
1983; Le Novère and Changeux, 1995). This class of receptors
was first known as Cys-loop receptors because all family sub-
units contain a conserved pair of disulfide-bonded cysteines
separated by 13 residues. The discovery of orthologs in pro-
karyotes (Tasneem et al., 2005), which lack the double cyste-
ines, has extended the Cys-loop family to the superfamily of
pentameric ligand-gated ion channels (pLGIC).
In vertebrates, the pLGIC superfamily includes cationic chan-
nels, nAChRs and serotonin 5-HT
3
receptors, and anionic chan-
nels activated by GABA or glycine (Le Novère and Changeux,
2001; Lester et al., 2004; Sine and Engel, 2006; Bartos et al., 2009).
Their vital role in converting chemical recognition into an
electrical impulse makes these receptors prime loci for learn-
ing, memory, and disease processes, as well as targets for clini-
cally relevant drugs.
The nAChR is widely distributed throughout the animal
kingdom, from nematodes to humans (Le Novère and Changeux,
1995). nAChRs are expressed in many regions of the central
This work was supported by grants from Universidad Nacional del Sur
(UNS), Consejo Nacional de Investigaciones Científicas y Técnicas (CONI-
CET), FONCYT, and the Bill and Melinda Gates Foundation to C.B.
dx.doi.org/10.1124/mol.116.104240.
ABBREVIATIONS: ACh, acetylcholine; a-BTX, a-bungarotoxin; ECD, extracellular domain; JAK, Janus kinase; LY-2087101, (2-amino-5-keto)
thiazole), [2-[(4-fluorophenyl)amino]-4-methyl-5-thiazolyl]-3-thienyl-methanone; nAChR, nicotinic acetylcholine receptor; NAM, negative allosteric
modulators; NS-1738, 1-(5-chloro-2-hydroxyphenyl)-3-(2-chloro-5-trifluoromethylphenyl)urea; PAM, positive allosteric modulator; pLGIC, pentameric
ligand-gated ion channels; PNU-120596, 1-(5-chloro-2,4-dimethoxyphenyl)-3-(5-methylisoxazol-3-yl)urea; SAM, silent allosteric modulator;
STAT, signal transducer and activator of transcription; TMD, transmembranedomain;TQS,4-(naphthalen-1-yl)-3a,4,5,9b-tetrahydro-3H-
cyclopenta[c]quinoline-8-sulfonamide.
288
at ASPET Journals on August 18, 2016molpharm.aspetjournals.orgDownloaded from
and peripheral nervous system, in addition to non-neuronal
tissues. The muscle nAChR plays a major role in neuromus-
cular transmission and is the target of muscle relaxants (Sine,
2012), whereas nAChRs in the brain represent a broad hetero-
geneous family of ubiquitously expressed receptors. nAChR
responses to endogenous acetylcholine (Ach) and choline and
exogenous nicotine are involved in a number of physiologic
processes and pharmacological effects (Dani and Bertrand,
2007; Albuquerque et al., 2009; Hurst et al., 2013).
The homopentameric a7, one of the most abundant nAChRs
in the nervous system, is also expressed in many non-neuronal
cells. Its unique activation properties, high calcium perme-
ability, ionotropic/metabotropic dual action, ubiquitous distri-
bution, and involvement in a range of neurologic, psychiatric,
and inflammatory disorders have made a7animportant
emerging drug target; the participation of a7 in pathologic
conditions and the therapeutic potential of a7 ligands has been
well documented (see for example Taly and Changeux, 2008;
Wallace and Porter, 2011; Lendvai et al., 2013; Wallace and
Bertrand, 2013; Uteshev, 2014; Dineley et al., 2015). In this
article, wefocus on the unique properties of activation and drug
modulation of a7 and its relationship withdisease and therapy.
nAChR Structure and Function
nAChR subunits are classified as two types, aand non-a,
with the a-type containing a disulfide bridge in the agonist
binding site. Five muscular (a1, b,g,«, and d) and eleven
neuronal (a2a7, a9, a10, and b2b4) nAChR subunits have
been identified in the mammalian genome (ligand-gated
ion channel database, http://www.ebi.ac.uk/compneur-srv/
LGICdb/cys-loop.php).
nAChRs are assembled from five identical (a7ora9) or
different subunits (at least two a-type subunits), and can form a
variety of different heteromeric receptors with a broad spec-
trum of pharmacological and kinetic properties (Fig. 1). The
resolution of the three-dimensional structures of pLGICs has
been the subject of intense efforts over the last decade (Brejc
et al., 2001; Dellisanti et al., 2007; Hilf and Dutzler, 2008, 2009;
Bocquet et al., 2009; Hibbs and Gouaux, 2011; Corringer et al.,
2012; Hassaine et al., 2014; Miller and Aricescu, 2014; Sauguet
et al., 2014; Cecchini and Changeux, 2015). However, no high-
resolution structure of the full length a7hasbeenreportedto
date; an extracellular domain of a7/AChBP chimera (Li et al.,
2011; Nemecz and Taylor, 2011) and a nuclear magnetic
resonance (NMR) structure of a7 transmembrane domain have
been described (Bondarenko et al., 2014).
All pLGICs share a conserved organization with five
subunits symmetrically arranged around a central ion pore
(Fig. 2). Functional domains include the extracellular domain
(ECD), which carries the agonist binding sites at subunit
interfaces; the transmembrane domain (TMD), which con-
tains the ion pore and the gate; and the intracellular domain
(ICD), which contains determinants of channel conductance
and sites for regulation and intracellular signaling (Paulo
et al., 2009; Jones et al., 2010; King et al., 2015) (Fig. 2). The
interface between the ECD and TMD, also referred to as the
coupling region, is important for coupling agonist binding to
channel opening (Bouzat et al., 2004; Lee and Sine, 2005;
Castillo et al., 2006; Bartos et al., 2009), as well as for
determining open channel lifetime and rate of desensitization
(Bouzat et al., 2008; Yan et al., 2015) (Fig. 2).
The possible structural events that translate neurotrans-
mitter binding at the ECD into opening of the transmembrane
ion channel 60 Å away is an issue of intense research that has
been discussed in recent reviews (Corringer et al., 2012;
Althoff et al., 2014; Sauguet et al., 2014; Cecchini and Changeux,
2015). On the basis of the Monod-Wyman-Changeux model
(Monod et al., 1965), the functional response of a pLGIC can be
interpreted as a selection from a few global and discrete
conformations elicited by the binding of agonist: closed, open,
and desensitized, the latter showinghighagonistaffinityatthe
same time being impermeable to ions (Zhang et al., 2013) (Fig. 3).
Intermediate states between closed and open or open and
desensitized states have been proposed for nAChRs and several
pLGICs (Lape et al., 2008; Corradi et al., 2009; Mukhtasimova
et al., 2009; Cecchini and Changeux, 2015). Therefore, the
number of states in the main conformations and the rate of
transition between states determine receptor kinetics, and this
tunes each receptor to its physiologic role. In turn, drugs, by
binding to different states, can differently modulate receptor
function.
a7 in the Nervous System in Healthy and Disease
States
a7 is one of the most abundant nAChRs in the central
nervous system. It is particularly expressed in regions impli-
cated in cognitive function and memory, such as hippocampus,
cortex, and several subcortical limbic regions (see Lendvai
Fig. 1. Models of pentameric arrangements of homomeric and some
heteromeric nAChRs. a7 and a9 are the only a-type subunits capable of
forming functional homomeric receptors, which contain five identical
binding sites. In a7, occupancy of only one site is required for activation.
Examples of possible combinations of aand non-asubunits in heteromeric
arrangements are shown. An a-type subunit is required for the principal
face of the binding site. The muscle nAChR contains two functional binding
sites at a/dand a/«(g) interfaces. Some subunits can assemble with
different stoichiometries, such as a4andb2. In addition to the arrange-
ment shown, (a4)
3
(b2)
2
receptors are also functional.
a7 Modulation 289
at ASPET Journals on August 18, 2016molpharm.aspetjournals.orgDownloaded from
et al., 2013). It is also expressed on non-neuronal cells,
including astrocytes, microglia, oligodendrocyte precursor
cells, and endothelial cells, where it plays a role in immunity,
inflammation, and neuroprotection (Shytle et al., 2004; Shen
and Yakel, 2012; Dineley et al., 2015).
In neurons, a7 receptors localize presynaptically on GABAer-
gic and glutamatergic terminals in the hippocampus and other
regions to facilitate release of neurotransmitters. Postsynapti-
cally, a7 receptors mediate fast synaptic transmission, and in
perisynaptic locations they modulate other inputs to neurons and
activate a variety of signaling pathways through volume trans-
mission (Gotti and Clementi, 2004; Jones and Wonnacott, 2004;
Dani and Bertrand, 2007; Dickinson et al., 2008; Albuquerque
et al., 2009; Sinkus et al., 2015) (Fig. 4).
a7 contributes to cognitive functioning, sensory processing
information, attention, working memory, and reward path-
ways, and a large body of evidence shows that enhancing a7
activity improves attention, cognitive performance, and neu-
ronal resistance to injury (reviewed in Uteshev, 2014).
Significant reduction of a7 in the brain, particularly in the
hippocampus, has been reported in Alzheimer disease (Guan
et al., 2000; Kadir et al., 2006) and schizophrenic patients
(Schaaf, 2014; Dineley et al., 2015). The a7 gene, CHRNA7 on
chromosome 15, is genetically linked to multiple disorders
with cognitive deficits, including schizophrenia, intellectual
disability, bipolar disorder, autism spectrum disorders, attention
deficit hyperactivity disorder, epilepsy, Alzheimer disease, and
sensory processing deficit (Sinkus et al., 2009, 2015; Schaaf,
2014; Dineley et al., 2015; Deutsch et al., 2016). A partial dupli-
cation of CHRNA7 resulting in the chimeric gene CHRFAM7A,
which is present only in humans, has been associated with
schizophrenia (Sinkus et al., 2009; Neri et al., 2012). Its gene
product, dupa7, lacks part of the binding site and may act as a
negative modulator of a7 (Wang et al., 2014).
Despite its homomeric character, a7 can assemble with
other subunits to form heteromeric receptors. In particular,
a7b2 heteromeric receptors have been detected in several
Fig. 2. Structural model of pLGICs. The ECD is folded into a
highly conserved immunoglobulin-like b-sandwich. The agonist
binding site is found in a cavity at an interface between two
adjacent subunits (Sine, 2012). The principal or positive face is
provided by an a-type subunit and includes three loops that
span bstrands (named as Loops AC) that harbor predomi-
nantly aromatic residues essential for binding and gating. The
adjacent subunit, which forms the complementary or minus
face, contributes with residues clustered in segments called
Loops DF (Brejc et al., 2001; Sine, 2012). ACh docked into the
binding site is shown (a7/AChBP chimera, pdb code 3SQ6). Key
aromatic residues from the principal face are Tyr188, Trp149,
and Tyr93, and from the minus face, Trp55. The transmem-
brane domain (TMD) is composed of four transmembrane-
spanning helices (TM1TM4). The TM2 forms the walls of the
ion pore, which contains the gate (ring of leucines at 99position)
and determinants for selectivity. The outer ring of fifteen
a-helices (TM1, TM3, and TM4) shields the inner TM2 ring
from the lipids (reviewed in Althoff et al., 2014). The interface
between the ECD and TMD, also named as coupling region,
includes the conserved Cys-loop (b6b7 loop), b1b2andb8b9
loops from the ECD, and the M2M3 linker from the TMD. The
long intracellular region (ICD) between TM3 and TM4 is highly
variable and particularly short in prokaryotic pLGICs. It is
thought to be associated with cytoskeletal proteins and in-
volved in channel modulation in all pLGICs (Kabbani et al.,
2013). As shown in the figure, in a7, this region contains
determinants of channel conductance (Andersen et al., 2011,
2013) and mediates several intracellular signals (Paulo et al.,
2009; Jones et al., 2010).
Fig. 3. Minimal model of nAChR activation. pLGICs have three main
classes of conformational states: Closed (C), open (O), and desensitized (D).
Intermediate states have also been detected.
290 Corradi and Bouzat
at ASPET Journals on August 18, 2016molpharm.aspetjournals.orgDownloaded from
brain areas (Liu et al., 2009, 2012; Moretti et al., 2014;
Thomsen et al., 2015; Zoli et al., 2015). Interestingly, a7b2
are highly sensitive to blockade by Ab142, suggesting that
they may play a unique role in the neuropathology of Alzheimer
disease (Liu et al., 2009). Additionally, these heteromeric
receptors exhibit high sensitivity to volatile anesthetics, and
therefore could be targets for anesthetic action (Mowrey et al.,
2013). Thus, a7b2 nAChR may represent a novel molecular
target requiring selective a7b2 ligands.
Extraneuronal a7 and Its Pleiotropic Roles
a7 is present in various non-neuronal tissues, such as glia
(Sharma and Vijayaraghavan, 2001), blood cells (Kawashima
and Fujii, 2004; De Rosa et al., 2005; Báez-Pagán et al., 2015),
keratinocytes (Maus et al., 1998), epithelial cells and fibro-
blasts (Zia et al., 1997), endothelial cells (Macklin et al., 1998),
cells of the digestive system and lung cells (reviewed in
Wessler and Kirkpatrick, 2008), spermatogonia, spermato-
cytes, and seminiferous tubular and Sertoli cells (Schirmer
et al., 2011). The functional role of a7 in these cells is being
intensively investigated and has been associated with differ-
entiation, migration, adhesion, cell contact, apoptosis, and
angiogenesis processes (Ni et al., 2010; Egea et al., 2015;
Zdanowski et al., 2015).
In particular, a7, present in all types of immune cells,
including lymphocytes (T and B cells), dendritic cells and
macrophages (reviewed in Egea et al., 2015), has attracted
considerable attention as an important drug target for in-
flammation. a7 is an important player in the cholinergic anti-
inflammatory pathway,which is a linkbetween vagal efferent
fibers and innate immune system (Martelli et al., 2014). a7
modulates intracellular signal pathways [Janus kinase 2
(JAK2)/signal transducer and activator of transcription
(STAT3) and PI3K/Akt] in immune cells, which results in
potent anti-inflammatory effects through cytokine production
inhibition and overexpression of heme oxygenase 1 (Báez-
Pagán et al., 2015; Egea etal., 2015) (Fig. 5). A brain cholinergic
pathway also exists that regulates microglial activation
through a7 (Shytle et al., 2004). This pathway is crucial for
neuroprotection (Park et al., 2007) and is probably important
in Parkinson disease (Stuckenholz et al., 2013), oxygen and
glucose deprivation (Parada et al., 2013), and global ischemia
(Guan et al., 2015).
Therefore, a7 nAChR is emerging as an important drug
target for the modulation of inflammation in different path-
ologic contexts, including sepsis, ischemia/reperfusion, rheu-
matoid arthritis, and pancreatitis.
a7 Pharmacology and Ion Selectivity
Hallmark features of a7 receptors include high Ca
21
permeability, relatively low sensitivity to ACh, full activation
by choline, high-affinity for a-bungarotoxin (a-BTX), relative-
ly low affinity for nicotine, and fast desensitization that occurs
on the submillisecond time scale.
Dose-response curves show EC
50
values of 100200 mM for
ACh [Hill coefficient (nH) 1] (Andersen et al., 2013), 0.4
1.6 mM for choline, and 1891 mM for nicotine (Wonnacott and
Barik, 2007). Several a7-specific agonists have been synthe-
tized, including PNU-282987 (EC
50
128 nM), AR-R17779
(EC
50
1020 mM), compound A (EC
50
14 nM to 0.95 mM),
and partial agonists GTS-21 (EC
50
626 mM), and SSR180711
(EC
50
14mM). Selective competitive antagonists are a-BTX
(IC
50
1100 nM), which has been widely used to detect a7
in tissues, and methyllycaconitine (MLA, IC
50
10200 nM)
(Wonnacott and Barik, 2007).
a7 allows flux of Na
1
and K
1
and is highly permeable to
Ca
21
. The PCa
21
/PNa
1
ratio is 1020, which is greater than
that of other nAChRs and similar to N-methyl-D-aspartate
receptors (Séguéla et al., 1993; Albuquerque et al., 1997). The
high Ca
21
permeability underlies most of a7 functions: facil-
itation of neurotransmitter release, depolarization of post-
synaptic cells, and initiation of many cellular processes through
its action as a second messenger (Gotti and Clementi, 2004).
The transient increase in intracellular Ca
21
is converted into a
sustained, wide-ranging phenomenon by calcium release from
intracellular stores through a calcium-induced calcium release
mechanism, a process involving IP
3
and ryanodine receptors
(Tsuneki et al., 2000; Dajas-Bailador et al., 2002; Guerra-
Álvarez et al., 2015) (Fig. 5).
The concept of a7 as a dual metabotropic/ionotropic receptor
is attracting increasing attention (Kabbani et al., 2013). a7
binds both Gaand Gbg proteins through the M3M4 loop and
enables a downstream calcium signaling response that can
persist beyond the expected time course of channel activation
(Fig. 5) (Kabbani et al., 2013; King et al., 2015). Moreover,
in lymphocyte T cells, mobilization of Ca
21
through the a7
channel is not necessarily required for the nicotine-induced
release of Ca
21
from the internal stores (Razani-Boroujerdi
et al., 2007). Channel-independent signal transduction has
been related to the role of a7 in inflammation (de Jonge and
Ulloa, 2007; Thomsen and Mikkelsen, 2012) and in neurite
growth (Nordman and Kabbani, 2012; Kabbani et al., 2013).
a7 is not only permeable to Ca
21
but is also modulated by
the ion; Ca
21
has been shown to regulate agonist efficacy and
cooperativity (Bonfante-Cabarcas et al., 1996; Albuquerque
et al., 1997). As in other pLGICs (Zimmermann et al., 2012),
the divalent modulatory sites may be located at the ECD.
a7 Channel Kinetics
Heterologous expression of a7 in oocytes and mammalian
cells combined with electrophysiological experiments has
provided information about the receptors unique activation
properties. The surface expression of recombinant a7 requires
Fig. 4. Neurotransmission mediated by a7 in the mammalian brain. a7
receptors can be postsynaptic, presynaptic (with a role in regulation of
neurotransmitter release), or perisynaptic when they are involved in
volume transmission.
a7 Modulation 291
at ASPET Journals on August 18, 2016molpharm.aspetjournals.orgDownloaded from
coexpression of the chaperone RIC-3 (Williams et al., 2005), a
transmembrane protein that is required for efficient receptor
folding, assembly, and functional expression (Castillo et al.,
2005; Millar, 2008). Recently, another chaperone, NACHO,
has been identified. NACHO is a transmembrane protein of
neuronal endoplasmic reticulum that mediates assembly of
a7 by promoting protein folding, maturation through the Golgi
complex, and expression at the cell surface (Gu et al., 2016).
Typical a7 receptor-mediated currents decay rapidly in the
presence of agonist as a result of desensitization (Fig. 6A).
Given the fast kinetics, the temporal resolution of agonist
exchange and most recording systems limit the accurate
estimation of the true desensitization rate (Zhou et al., 1998;
Lovinger et al., 2002; Bouzat et al., 2008), which may partially
account for the great variability of desensitization rates found
in the literature. Outside-out patches rapidly perfused with
ACh, which allows for a more accurate determination of the
desensitization rate, show current decay time constants of
0.4 milliseconds (Bouzat et al., 2008). Owing to their fast
decay, a7 peak responses occur in advance of complete solu-
tion exchange. Therefore, more accurate EC
50
values are
obtained if the net charge, which represents the time in-
tegration of all channel activation, rather than the peak
current, is used for the analysis of the concentration-effect
relationship (Papke and Porter Papke, 2002) (Fig. 6A).
At the single-channel level, channel activity appears as
isolated brief pulses (0.10.3 milliseconds) flanked by long
closed periods and, less often, as two or three brief pulses in
quick succession (bursts) (Fig. 6B). Thus, a7 has a very low
open probability. Single-channel openings exhibit a broad
distribution of current amplitudes, probably owing to limited
time resolution of the brief openings, with a maximum of 10 pA
at 70 mV (Mike et al., 2000; Bouzat et al., 2008; Andersen et al.,
2013; daCosta and Sine, 2013; Yan et al., 2015).
Although there is no general consensus for an a7 kinetic
model, an interesting aspect is that the temporal pattern of
single ACh-activated currents is similar at 10 mMor1mM
ACh (Bouzat et al., 2008) (Fig. 6B). This lack of concentration-
dependence combined with the fact that most receptor acti-
vation episodes consist of a single opening with a duration
similar to the desensitization time constant suggests that
desensitization is the predominant pathway for channel
closing, a unique feature among nAChRs. Control of open-
channel lifetime through desensitization has potential conse-
quences for inter-response latency at a synapse where the
neurotransmitter pulse is transient. Recovery from desensiti-
zation depends on agonist concentration and exposure dura-
tion, since different desensitized states may exist. Desensitized
a7 receptors expressed in human embryonic kidney cells
recover with a time constant of 1 second (Bouzat et al.,
2008), whereas 1530 seconds are required for full recovery in
the hippocampus (Frazier et al., 1998). Thus, after a7 brief
response, a latency of several seconds is required to generate
another response of full amplitude. The fast desensitization
and brief open duration may avoid cell toxicity caused by
increased intracellular Ca
21
owing to a7 overstimulation.
Fig. 5. Dual ionotropic/metabotropic nature of a7: Intracellular pathway signaling mediated by a7 activation. a7 allows transient flux of Na
+
,K
+
, and
Ca
2+
. The transient increase of calcium may also lead to a sustained calcium response by a calcium-induced calcium release (CICR) mechanism through
IP
3
receptors. As a metabotropic receptor, a7 mediates intracellular signals by binding to G aand Gbg proteins and several other partners (Kabbani et al.,
2013; King et al., 2015). In immune cells, a7 has been shown to be involved in several intracellular pathways; although not yet fully deciphered
mechanistically, these pathways lead to potent anti-inflammatory effects. For example, a7 has been shown to activate JAK2/STAT3 in some immune
cells, which leads to blockade of nuclear factor (NF)-kB nuclear translocation and inhibition of NF-kB, resulting in inhibition of proinflammatory cytokine
production. Also, a7 has been shown to activate the PI3K/Akt pathway that promotes Nrf-2 translocation to the nucleus and overexpression of heme
oxygenase 1 (HO-1), resulting in potent anti-inflammatory effects (see de Jonge and Ulloa, 2007; Báez-Pagán et al., 2015; Egea et al., 2015). DAG,
diacylglycerol; GPCR, G protein-coupled receptor; PKC, protein kinase C; ROS, reactive oxygen species.
292 Corradi and Bouzat
at ASPET Journals on August 18, 2016molpharm.aspetjournals.orgDownloaded from
Another intriguing aspect of a7 activation is the relation-
ship between ACh occupancy and activation. We set out to
determine how many of the five identical agonist binding sites
are required to activate a7 by developing a strategy that
utilizes coexpression of an inactivated binding-site subunit
and a reporter amplitude subunit. This allows for the de-
termination of the number of ACh-occupied sites from the
amplitude of each individual single-channel opening (Rayes
et al., 2009; Andersen et al., 2011, 2013). The results revealed
that ACh occupancy of only one of five a7 binding sites is
necessary for activation, and that open-channel lifetime of
a single-occupied receptor is indistinguishable from that of
receptors containing five intact binding sites (Andersen et al.,
2013). Also, occupancy of a single site by the antagonist
methylcaconitine (Palma et al., 1996) or a-BTX (daCosta
et al., 2015) will inhibit a7 function.
Whole-cell experiments have revealed the physiologic con-
sequence of having more sites than those required for
activation; saturation of receptors in cells expressing a wild-
type and a binding-site mutant subunit (a7Y188T) is achieved
at higher ACh concentrations when the proportion of recep-
tors with fewer functional binding sites increases (Andersen
et al., 2013; Williams et al., 2011a). This suggests that a7is
a highly sensitive sensor of ACh and is therefore adapted to
function with submaximal occupancy of its sites, a property
appropriate for volume transmission.
The unique activation properties of a7 also suggest that any
slight change in the energy barriers between active, closed,
and/or desensitized states may have a deep impact on receptor
function, which makes these receptors very sensitive drug
targets.
a7 Modulation as a Therapeutic Strategy
In addition to agonists and antagonists that bind to the
orthosteric site, a large number of compounds modulate a7
function by binding to allosteric sites. These compounds may
act as: 1) positive allosteric modulators (PAMs) that potenti-
ate currents only in the presence of the agonist; 2) allosteric
agonists that activate receptors from nonorthosteric sites; 3)
negative allosteric modulators (NAMs), which either act as
open-channel blockers by binding to the pore or inhibit
activation allosterically; and 4) silent allosteric modulators
(SAMs) that have no effect on orthosteric agonist responses
but block allosteric modulation (Fig. 7).
Since stimulation of a7 improves attention, cognitive per-
formance, and neuronal resistance to injury in addition to
eliciting robust analgesic and anti-inflammatory effects, a7
potentiation has emerged as a potential therapeutic strategy,
and the search for novel potentiators is an active research
field. The potential therapeutic use of several a7 partial
agonists and PAMs on animals and humans has been docu-
mented in several recent reviews (e.g., Wallace and Porter,
2011; Thomsen and Mikkelsen, 2012; Lendvai et al., 2013;
Dineley et al., 2015) (Table 1). Still, no drug has reached phase
III clinical stage.
Compared with agonists, PAMs are promising therapeutic
tools because they: 1) better maintain the temporal spatial
characteristics of endogenous activation; 2) show higher
selectivity, since the orthosteric site is more conserved among
nAChRs than allosteric sites (Yang et al., 2012); 3) allow
greater diversity in structure and final effects; 4) reduce
tolerance attributable to a7 desensitization; and 5) act as
neuronal protectors (Kalappa et al., 2013; Sun et al., 2013;
Uteshev, 2014). In particular, because neuronal damage
elevates choline levels near the site of injury, the presence of
a PAM may not require coapplication of an agonist for the
mediation of a local neuroprotective effect (Uteshev, 2014).
Therefore, it is increasingly accepted that targeting allosteric
sites can provide novel medications with greater structural
diversity and specificity.
PAMs have been classified on the basis of their macroscopic
effects as type I or type II (Fig. 8). Type I PAMs mainly
enhance agonist-induced peak currents without significantly
affecting current decay and do not reactivate desensitized
receptors, whereas type II PAMs delay desensitization and
reactivate desensitized receptors (Bertrand and Gopalakrishnan,
2007; Arias and Bouzat, 2010; Williams et al., 2011b). The
ratio of the changes in net charge/peak current induced by
type I PAMs is close to one, whereas it is higher than one in
the presence of type II PAMs (Andersen et al., 2016; Williams
et al., 2011c).
Single-channel recordings provide an invaluable tool for un-
derstanding the foundations of these macroscopic effects. In
the presence of either type I or type II PAMs, ACh-activated a7
channels show prolonged open durations and appear in longer
activation episodes, revealing that both PAM types affect
activation kinetics (Andersen et al., 2016) (Fig. 8). The most
efficacious PAM to date is PNU-120596, a type II PAM (Hurst
et al., 2005). This compound elicits significantly prolonged
openingsthatappeargroupedinbursts,whichinturncoalesce
into long activation periods of several seconds, referred to as
clusters (daCosta et al., 2011, 2015; Williams et al., 2011b;
Pałczy
nska et al., 2012; Andersen et al., 2016). However, the
Fig. 6. a7 single-channel and macroscopic currents. (A) Typical whole-cell
currents elicited by different ACh concentrations (from 10 to 1000 mM)
from cells transfected with human a7. At left, a schematic representation
of a macroscopic current with the measured parameters. Both the maximal
current (peak) and the net charge (net) can be used to construct dose-
response curves as shown in the figure. (B) Single-channel activity from
cell-attached patches of cells expressing human a7 appears as very brief
(0.10.3 milliseconds) and isolated openings or less often as short bursts.
Channel openings are shown as upward deflections. Membrane potential,
70 mV; filter, 9 kHz. At right, a typical open duration histogram (Bouzat
et al., 2008).
a7 Modulation 293
at ASPET Journals on August 18, 2016molpharm.aspetjournals.orgDownloaded from
M254
F456
I281
V288
S223
C460
A226 TM1
TM2 TM3 TM4
Extracellular pockets
Intrasubunit cavity
Pore
PAMs
NAMs
SAMs
Allosteric agonists
NAMs
PAMs
MODULATORY SITES
PNU-120596
Open-channel blockers
Fig. 7. Allosteric modulatory sites in a7. Structural model of the a7 receptor viewed from the side representing several potential sites for allosteric
modulators. At the TMD, the intrasubunit transmembrane cavity is a site for a great variety of compounds that may elicit different pharmacological
effects (potentiation (PAMs), inhibition (NAMs), no effect (SAMs), or activation (allosteric agonists)). PNU-120596 docked into this cavity is shown. Open-
channel blockers inhibit response by transiently blocking the flux of ions through the pore. At the ECD, different potential sites for inhibitors and
potentiators have been proposed (Ludwig et al., 2010; Spurny et al., 2015).
TABLE 1
a7 modulators with potential clinical applications.
Drug Potential Therapeutic Effects and Uses References
Agonists GTS-21/DMXB-A Cognitive disorders, schizophrenia, Alzheimer
disease, attention deficit hyperactivity disorder Olincy et al. (2006)
Pain van Westerloo et al. (2006)
Inflammatory processes Vukelic et al. (2013)
Terry et al. (2015)
AR-R17779 Cognitive disorders Levin et al. (1999)
Inflammatory processes Hashimoto et al. (2014)
Atherosclerotic vascular diseases Van Kampen et al. (2004)
PNU-282987 Cognitive disorders Hajós et al. (2005)
Inflammatory processes Terry et al. (2015)
SSR180711 Cognitive disorders Pichat et al. (2007)
Terry et al. (2015)
ABBF Cognitive disorders Boess et al. (2007)
Terry et al. (2015)
EVP-6124 Cognitive disorders Prickaerts et al. (2012)
Terry et al. (2015)
TC-5619 Cognitive disorders Hauser et al. (2009)
Terry et al. (2015)
RG3487 Cognitive and sensorimotor gating disorders Wallace et al. (2011)
Terry et al. (2015)
PHA-568487 Cognitive disorders Karamihalev et al. (2014)
Terry et al. (2015)
CP-810123 Cognitive disorders ODonnell et al. (2010)
Terry et al. (2015)
AZD0328 Cognitive disorders Sydserff et al. (2009)
Terry et al. (2015)
Tropisetron Cognitive disorders Shiina et al. (2010)
Hashimoto, (2015)
Terry et al. (2015)
ABT-107 Cognitive disorders Bitner et al. (2010)
Neuroprotection Quik et al. (2015)
Terry et al. (2015)
JN403 Cognitive disorders Feuerbach et al. (2007, (2009)
Anticonvulsive
Pain
Type I PAMs Genistein Neuroprotection Menze et al. (2015, 2016)
Memory disorders Terry et al. (2015)
NS-1738 Cognitive disorders Timmermann et al. (2007)
Terry et al. (2015)
AVL-3288 Cognitive disorders Ng et al. (2007)
Terry et al. (2015)
Galantamine Cognitive disorders Nikiforuk et al. (2015)
Alzheimer disease Terry et al. (2015)
Type II PAMs PNU-120596 Cognitive disorders Nikiforuk et al. (2015)
Inflammatory processes Callahan et al. (2013)
Terry et al. (2015)
PAM-2 Cognitive disorders Targowska-Duda et al. (2016)
Pain Bagdas et al. (2015)
Inflammatory processes
294 Corradi and Bouzat
at ASPET Journals on August 18, 2016molpharm.aspetjournals.orgDownloaded from
single-channel profile in the presence of a weaker type II PAM,
PAM-2 (3-furan-2-yl-N-p-tolyl-acrylamide) (Arias et al., 2011),
more closely resembles that of type I 5-HI or NS-1738 PAMs
than that of type II PNU-120596 (Fig. 8). Thus, when analyzed
at the molecular level, potentiation is more complex than
initially believed. Moreover, PAMs showing macroscopic in-
termediate type I/II properties were proposed (Dunlop et al.,
2009; Malysz et al., 2009; Dinklo et al., 2011; Sahdeo et al.,
2014; Chatzidaki and Millar, 2015). Therefore, the classifica-
tion of type I and type II appears to be an oversimplification
resulting mainly from macroscopic observations, and a more
thorough classification may be required.
It is a generally accepted statement that type II PAMs may
increase the energetic barrier for desensitization, which allows
successive opening/closing events (daCosta et al., 2011) and/or
reversal of some forms of agonist-induced desensitization
(Williams et al., 2011b). Also, it has been proposed that PNU-
120596 binds predominantly to a fast desensitized state and
induces a set of conformations in which the opening of the pore
is energetically more favorable (Szabo et al., 2014). On the other
hand, only the decrease inthe energetic barrier for opening has
been proposed as the underlying mechanism for enhancement
of peak currents by type I PAMs (Williams et al., 2011b; Hurst
et al., 2013). Such a decrease might explain the appearance of
bursts of openings owing to rapid reopening of the closed
channel. However, it would only explain the increase in open-
channel duration if reopening were so fast that the associated
brief closings could not be detected, thus making openings ap-
pear longer. Alternatively, the increase in open duration could
be the result of either slight changes in desensitization that are
not detectable from whole-cell macroscopic currents or to the
induction of different open states. Thus, there seems to be more
than one mechanism by which PAMs prolong open-channel
lifetime and activation episodes.
a7 PAM potentiation is particularly dependent on temper-
ature. For PNU-120596, such dependence is revealed by
decreased potentiated macroscopic currents (Dunlop et al.,
2009; Williams et al., 2012) and the absence of long clusters
(Andersen et al., 2016) at physiologic temperature compared
with room temperature. Thus, for better extrapolation to the
in vivo situation, in vitro studies should be carried out at
physiologic temperatures.
a7 Allosteric Binding Sites
Computational studies (Dey and Chen, 2011), electrophysio-
logical studies from mutant or chimeric receptors (Bertrand
et al., 2008; Young et al., 2008; Collins et al., 2011; daCosta and
Sine, 2013), crystallographic studies, (Spurny et al., 2015), and
NMR studies (Bondarenko et al., 2014) have suggested the
existence of several allosteric binding sites, some of which are
common to other pLGICs (Sauguet et al., 2014).
The Intrasubunit Transmembrane Cavity. In silico
and electrophysiological studies show that several PAMs, in-
cluding LY-2087101, PNU-120596, and TQS bind to an intra-
subunit transmembrane cavity (Young et al., 2008) (Fig. 7). An
a7 receptor with mutations at five residues lining this cavity
was not potentiated by PNU-120596, PAM-2, or type I PAM
NS-1738, indicating common structural determinants for
their potentiation (daCosta et al., 2011; Andersen et al., 2016).
The macrocyclic lactone ivermectin (a type I PAM) also appears
to bind in close proximity to this intrasubunit site (Collins and
Millar, 2010), although it binds at an intersubunit transmem-
brane site in the glutamate-activated Cys-loop receptor (Hibbs
and Gouaux, 2011).
This cavity is also the site for allosteric agonists that me-
diate a7 activation in the absence of an orthosteric agonist
(Gill et al., 2011, 2012, 2013; Pałczy
nska et al., 2012). Single-
channel activity of a7 in the presence of allosteric agonists
resembles that of the receptor in the presence of ACh and a
PAM. Both are characterized by long bursts instead of isolated
brief ACh-elicited openings, indicating activation with signif-
icantly reduced desensitization (Pałczy
nska et al., 2012).
Moreover, a7-selective allosteric modulators showing subtle
structural changes and displaying distinct pharmacological
effects (typical of type I PAMs, type II PAMs, NAMs, SAMs,
and allosteric agonists) may bind to this common site (Gill
et al., 2013; Gill-Thind et al., 2015). Thus, the intrasubunit trans-
membrane cavity appears to be a promiscuous binding site
with a strategic location for allosteric modulation. Further-
more, this cavity may be a common modulatory site within the
pLGIC superfamily from which a large variety of different
compounds that bind to and mediate a great spectrum of
allosteric effects (Corradi et al., 2011; Nury et al., 2011;
Jayakar et al., 2013; Sauguet et al., 2014).
Fig. 8. Macroscopic and single-channel current profiles of human a7in
the presence of typical type I and type II PAMs. Macroscopic current
profiles have been used to classify PAMs into type II (i.e., PNU-120596 and
PAM-2), which increase the decay time constant, or type I, which only
increase the peak current (5-HI and NS-1738). Left: Macroscopic currents
elicited by ACh in the absence (black) and presence of the specified PAM
(gray traces). Right: Traces of 50100 mM ACh-activated single a7
channels in the absence or in the presence of 1 mM PNU-120596, 5 mM
PAM-2, 2 mM 5-HI, 10 mM NS-1738. Membrane potential: 70 mV.
Channel openings are shown as upward deflections. All PAMs enhance
open channel lifetime and elicit activation episodes formed by successive
opening events.
a7 Modulation 295
at ASPET Journals on August 18, 2016molpharm.aspetjournals.orgDownloaded from
Extracellular Allosteric Binding Sites. The putative
binding site of galantamine, an a7 potentiator, is located at
the outer surface of the ECD in the vicinity of the ACh site
(Hansen and Taylor, 2007; Ludwig et al., 2010). Three
different allosteric sites in the ECD of the a7/AChBP chimera
were also identified by X-ray crystallography (Spurny et al.,
2015; Fig. 7). Although all the allosteric binders behaved on
human a7 as negative allosteric modulators, it was proposed
that their chemical modification could lead to a change in
functional activity.
Binding to potential ECD sites has been proposed for the
type I PAMs 5-HI and NS-1738, although other reports
support a TMD location (Placzek et al., 2004; Bertrand et al.,
2008; Hu and Lovinger, 2008; Gronlien et al., 2010; Collins
et al., 2011; Andersen et al., 2016). A virtual screening re-
vealed that some PAMs that bind to the TMD, such as PNU-
120596 and TQS, also dock into potential allosteric sites at
the ECD (Dey and Chen, 2011). Therefore, for any given PAM,
multiple binding sites or domains may be involved in the
conformational changes associated with potentiation, which
could account for these controversial results. Also, until the
location of an allosteric binding site is unequivocally defined,
it is advisable to refer to structural determinants of potentia-
tion instead of a binding site.
Despite the large body of experimental evidence supporting
a7 potentiation as a promising therapeutic strategy, there are
still many unsolved challenges: 1) Potentiation by exogenous
agonists may inhibit a7 response owing to desensitization.
Thus, PAMs might have therapeutic benefits in situations
where stronger agonist responses are desirable. 2) Given the
presence of other nAChRs and homologous receptors, high
PAM selectivity is required. However, PAMs targeting multi-
ple receptors might show better efficacy (Iturriaga-Vásquez
et al., 2015; Möller-Acuña et al., 2015). 3) Excessive receptor
activation, particularly with efficacious nondesensitizing
PAMs, might lead to cytotoxicity, which is an issue of concern
and controversy (Ng et al., 2007; Liu et al., 2009; Williams
et al., 2012; Guerra-Álvarez et al., 2015; Uteshev, 2016). 4)
The ubiquitous distribution of a7 and its interplay with
different signal pathways could make the cell response to a
given PAM variable among cell types or conditions. Given the
broad spectrum of effects and molecular mechanisms of PAMs,
it is probable that each patient or pathologic situation could
require a unique PAM.
Concluding Remarks
a7 has emerged as an important drug target for improving
cognition and memory in several neuropsychiatric disorders,
and as a target for inflammatory processes. a7 is unique owing
to its high calcium permeability and fast desensitization, and
it behaves as a ACh-sensitive sensor harboring a built-in
filtering mechanism against excessive stimulation. Transient
calcium responses are further sustained by the release of
calcium from intracellular sources, and several signaling
pathways are also activated because a7 has a dual ionotropic/
metabotropic nature. Its ubiquitous location and pleiotropic
effects make a7 an interesting but complex drug target. A bet-
ter understanding of the molecular basis underlying allosteric
modulation and its wide spectrum of effects, as well as the
availability of high resolution structures of a7, will help in the
rational design of therapeutics for the receptor.
Acknowledgments
ASPET thanks Dr. Katie Strong for copyediting of this article.
Authorship Contributions
Wrote or contributed to the writing of the manuscript: Bouzat,
Corradi.
References
Albuquerque EX, Alkondon M, Pereira EF, Castro NG, Schrattenholz A, Barbosa
CT, Bonfante-Cabarcas R, Aracava Y, Eisenberg HM, and Maelicke A (1997)
Properties of neuronal nicotinic acetylcholine receptors: pharmacological char-
acterization and modulation of synaptic function. JPharmacolExpTher28 0:
11171136.
Albuquerque EX, Pereira EFR, Alkondon M, and Rogers SW (2009) Mammalian nico-
tinic acetylcholine receptors: from structure to function. Physiol Rev 89:73120.
Althoff T, Hibbs RE, Banerjee S, and Gouaux E (2014) X-ray structures of GluCl in
apo states reveal a gating mechanism of Cys-loop receptors. Nature 512:333337.
Andersen N, Corradi J, Bartos M, Sine SM, and Bouzat C (2011) Functional rela-
tionships between agonist binding sites and coupling regions of homomeric Cys-
loop receptors. J Neurosci 31:36623669.
Andersen N, Corradi J, Sine SM, and Bouzat C (2013) Stoichiometry for activation of
neuronal a7 nicotinic receptors. Proc Natl Acad Sci USA 110:2081920824.
AndersenND, Nielsen BE, Corradi J, Tolosa MF, Feuerbach D, Arias HR, and Bouzat C
(2016) Exploring the positive allosteric modulation of human a7 nicotinic receptors
from a single-channel perspective. Neuropharmacology 107:189200.
Arias HR and Bouzat C (2010) Activation and modulation of the nicotinic receptor.
J Pediatr Biochem 1:5373.
Arias HR, Gu R-X, Feuerbach D, Guo B-B, Ye Y, and Wei D-Q (2011) Novel positive
allosteric modulators of the human a7 nicotinic acetylcholine receptor. Bio-
chemistry 50:52635278.
Báez-Pagán CA, Delgado-Vélez M, and Lasalde-Dominicci JA (2015) Activation of the
macrophage a7 nicotinic acetylcholine receptor and control of inflammation.
J Neuroimmune Pharmacol 10:468476.
Bagdas D, Targowska-Duda KM, López JJ, Perez EG, Arias HR, and Damaj MI
(2015) The Antinociceptive and Antiinflammatory Properties of 3-furan-2-yl-N-p-
tolyl-acrylamide, a Positive Allosteric Modulator of a7 Nicotinic Acetylcholine
Receptors in Mice. Anesth Analg 121:13691377.
Bartos M, Corradi J, and Bouzat C (2009) Structural basis of activation of cys-loop
receptors: the extracellular-transmembrane interface as a coupling region. Mol
Neurobiol 40:236252.
Bertrand D, Bertrand S, Cassar S, Gubbins E, Li J, and Gopalakrishnan M (2008)
Positive allosteric modulation of the a7 nicotinic acetylcholine receptor: ligand
interactions with distinct binding sites and evidence for a prominent role of the
M2-M3 segment. Mol Pharmacol 74:14071416.
Bertrand D and Gopalakrishnan M (2007) Allosteric modulation of nicotinic acetyl-
choline receptors. Biochem Pharmacol 74:11551163.
Bitner RS, Bunnelle WH, Decker MW, Drescher KU, Kohlhaas KL, Markosyan S,
Marsh KC, Nikkel AL, Browman K, Radek R, et al. (2010) In vivo pharmacological
characterization of a novel selective a7 neuronal nicotinic acetylcholine receptor
agonist ABT-107: preclinical considerations in Alzheimers disease. J Pharmacol
Exp Ther 334:875886.
Bocquet N, Nury H, Baaden M, Le Poupon C, Changeux J-P, Delarue M,
and Corringer P-J (2009) X-ray structure of a pentameric ligand-gated ion channel
in an apparently open conformation. Nature 457:111114.
Boess FG, De Vry J, Erb C, Flessner T, Hendrix M, Luithle J, Methfessel C, Riedl B,
Schnizler K, Van Der Staay F, et al. (2007) The Novel a7 Nicotinic Acetylcholine
Receptor Agonist benzofuran-2-carboxamide Improves Working and Recognition
Memory in Rodents. Pharmacology 321:716725.
Bondarenko V, Mowrey DD, Tillman TS, Seyoum E, Xu Y, and Tang P (2014) NMR
structures of the human a7 nAChR transmembrane domain and associated anes-
thetic binding sites. Biochim Biophys Acta 1838:13891395.
Bonfante-Cabarcas R, Swanson KL, Alkondon M, and Albuquerque EX (1996) Di-
versity of nicotinic acetylcholine receptors in rat hippocampal neurons. IV. Regu-
lation by external Ca11 of alpha-bungarotoxin-sensitive receptor function and of
rectification induced by internal Mg11.J Pharmacol Exp Ther 277:432444.
Bouzat C (2012) New insights into the structural bases of activation of Cys-loop
receptors. J Physiol Paris 106:2333.
Bouzat C, Bartos M, Corradi J, and Sine SM (2008) The interface between extra-
cellular and transmembrane domains of homomeric Cys-loop receptors governs
open-channel lifetime and rate of desensitization. J Neurosci 28:78087819.
Bouzat C, Gumilar F, Spitzmaul G, Wang H-L, Rayes D, Hansen SB, Taylor P,
and Sine SM (2004) Coupling of agonist binding to channel gating in an ACh-
binding protein linked to an ion channel. Nature 430:896900.
Brejc K, van Dijk WJ, Klaassen RV, Schuurmans M, van Der Oost J, Smit AB,
and Sixma TK (2001) Crystal structure of an ACh-binding protein reveals the
ligand-binding domain of nicotinic receptors. Nature 411:269276.
Callahan PM, Hutchings EJ, Kille NJ, Chapman JM, and Terry AV, Jr (2013) Pos-
itive allosteric modulator of a7 nicotinic-acetylcholine receptors, PNU-120596
augments the effects of donepezil on learning and memory in aged rodents and
non-human primates. Neuropharmacology 67:201212.
Castillo M, Mulet J, Bernal JA, Criado M, Sala F, and Sala S (2006) Improved gating
of a chimeric alpha7-5HT3A receptor upon mutations at the M2-M3 extracellular
loop. FEBS Lett 580:256260.
Castillo M, Mulet J, Gutiérrez LM, Ortiz JA, Castelán F, Gerber S, Sala S, Sala F,
and Criado M (2005) Dual role of the RIC-3 protein in trafficking of serotonin and
nicotinic acetylcholine receptors. J Biol Chem 280:2706227068.
296 Corradi and Bouzat
at ASPET Journals on August 18, 2016molpharm.aspetjournals.orgDownloaded from
Cecchini M and Changeux J-P (2015) The nicotinic acetylcholine receptor and its
prokaryotic homologues: Structure, conformational transitions & allosteric modu-
lation. Neuropharmacology 96 (Pt B):137149.
Changeux JP, Kasai M, and Lee CY (1970) Use of a snake venom toxin to charac-
terize the cholinergic receptor protein. Proc Natl Acad Sci USA 67:12411247.
Chatzidaki A and Millar NS (2015) Allosteric modulation of nicotinic acetylcholine
receptors. Biochem Pharmacol 97:408417.
Collins T and Millar NS (2010). Nicotinic acetylcholine receptor transmembrane
mutations convert ivermectin from a positive to a negative allosteric modulator.
Mol Pharmacol 78:198204.
Collins T, Young GT, and Millar NS (2011) Competitive binding at a nicotinic
receptor transmembrane site of two a7-selective positive allosteric modulators
with differing effects on agonist-evoked desensitization. Neuropharmacology 61:
13061313.
Corradi J, Andersen N, and Bouzat C (2011) A novel mechanism of modulation of
5-HTA receptors by hydrocortisone. Biophys J 100:4251.
Corradi J, Gumilar F, and Bouzat C (2009) Single-channel kinetic analysis for activa-
tion and desensitization of homomeric 5-HT(3)A receptors. Biophys J 97:13351345.
Corringer P-J, Poitevin F, Prevost MS, Sauguet L, Delarue M, and Changeux J-P
(2012) Structure and pharmacology of pentameric receptor channels: from bacteria
to brain. Structure 20:941956.
daCosta CJB, Free CR, Corradi J, Bouzat C, and Sine SM (2011) Single-channel and
structural foundations of neuronal a7 acetylcholine receptor potentiation. J Neu-
rosci 31:1387013879.
daCosta CJB, Free CR, and Sine SM (2015) Stoichiometry for a-bungarotoxin block of
a7 acetylcholine receptors. Nat Commun 6:8057.
daCosta CJB and Sine SM (2013) Stoichiometry for drug potentiation of a pentameric
ion channel. Proc Natl Acad Sci USA 110:65956600.
Dajas-Bailador FA, Mogg AJ, and Wonnacott S (2002) Intracellular Ca21signals evoked
by stimulation of nicotinic acetylcholine receptors in SH-SY5Y cells: contribution of
voltage-operated Ca21channels and Ca21stores. J Neurochem 81:606614.
Dani JA and Bertrand D (2007) Nicotinic acetylcholine receptors and nicotinic cho-
linergic mechanisms of the central nervous system. Annu Rev Pharmacol Toxicol
47:699729.
de Jonge WJ and Ulloa L (2007) The alpha7 nicotinic acetylcholine receptor as a
pharmacological target for inflammation. Br J Pharmacol 151:915929.
Dellisanti CD, Yao Y, Stroud JC, Wang Z-Z, and Chen L (2007) Crystal structure of
the extracellular domain of nAChR alpha1 bound to alpha-bungarotoxin at 1.94 A
resolution. Nat Neurosci 10:953962.
De Rosa MJ, Esandi MdelC, Garelli A, Rayes D, and Bouzat C (2005) Relationship between
alpha 7 nAChR and apoptosis in human lymphocytes. J Neuroimmunol 160:154161.
Deutsch SI, Burket JA, Benson AD, and Urbano MR (2016) The 15q13.3 deletion
syndrome: Deficient a(7)-containing nicotinic acetylcholine receptor-mediated
neurotransmission in the pathogenesis of neurodevelopmental disorders. Prog
Neuropsychopharmacol Biol Psychiatry 64:109117.
Dey R and Chen L (2011) In search of allosteric modulators of a7-nAChR by solvent
density guided virtual screening. J Biomol Struct Dyn 28:695715.
Dickinson JA, Kew JNC, and Wonnacott S (2008) Presynaptic alpha 7- and beta 2-
containing nicotinic acetylcholine receptors modulate excitatory amino acid release
from rat prefrontal cortex nerve terminals via distinct cellular mechanisms. Mol
Pharmacol 74:348359.
Dineley KT, Pandya AA, and Yakel JL (2015) Nicotinic ACh receptors as therapeutic
targets in CNS disorders. Trends Pharmacol Sci 36:96108.
Dinklo T, Shaban H, Thuring JW, Lavreysen H, Stevens KE, Zheng L, Mackie C,
Grantham C, Vandenberk I, Meulders G, et al. (2011) Characterization of 2-[[4-
fluoro-3-(trifluoromethyl)phenyl]amino]-4-(4-pyridinyl)-5-thiazolemethanol (JNJ-
1930942), a novel positive allosteric modulator of the a7 nicotinic acetylcholine
receptor. J Pharmacol Exp Ther 336:560574.
Dunlop J, Lock T, Jow B, Sitzia F, Grauer S, Jow F, Kramer A, Bowlby MR, Randall
A, Kowal D, et al. (2009) Old and new pharmacology: positive allosteric modulation
of the a7 nicotinic acetylcholine receptor by the 5-hydroxytryptamine(2B/C) re-
ceptor antagonist SB-206553 (3,5-dihydro-5-methyl-N-3-pyridinylbenzo[1,2-b:4,5-
b9]di pyrrole-1(2H)-carboxamide). J Pharmacol Exp Ther 328:766776.
Egea J, Buendia I, Parada E, Navarro E, León R, and Lopez MG (2015) Anti-
inflammatory role of microglial alpha7 nAChRs and its role in neuroprotection.
Biochem Pharmacol 97:463472.
Feuerbach D, Lingenhoehl K, Olpe HR, Vassout A, Gentsch C, Chaperon F, Nozulak
J, Enz A, Bilbe G, McAllister K, et al. (2009) The selective nicotinic acetylcholine
receptor a7 agonist JN403 is active in animal models of cognition, sensory gating,
epilepsy and pain. Neuropharmacology 56:254263.
Feuerbach D, Nozulak J, Lingenhoehl K, McAllister K, and Hoyer D (2007) JN403, in
vitro characterization of a novel nicotinic acetylcholine receptor a7 selective ago-
nist. Neurosci Lett 416:6165.
Frazier CJ, Rollins YD, Breese CR, Leonard S, Freedman R, and Dunwiddie TV
(1998) Acetylcholine activates an alpha-bungarotoxin-sensitive nicotinic current in
rat hippocampal interneurons, but not pyramidal cells. J Neurosci 18:11871195.
Gill JK, Dhankher P, Sheppard TD, Sher E, and Millar NS (2012) A series of a7
nicotinic acetylcholine receptor allosteric modulators with close chemical similarity
but diverse pharmacological properties. Mol Pharmacol 81:710718.
Gill JK, Chatzidaki A, Ursu D, Sher E, and Millar NS (2013) Contrasting properties
of a7-selective orthosteric and allosteric agonists examined on native nicotinic
acetylcholine receptors. PLoS One 8:e55047 10.1371/journal.pone.0055047.
Gill JK, Savolainen M, Young GT, Zwart R, Sher E, and Millar NS (2011) Agonist
activation of alpha7 nicotinic acetylcholine receptors via an allosteric trans-
membrane site. Proc Natl Acad Sci USA 108:58675872.
Gill-Thind JK, Dhankher P, DOyley JM, Sheppard TD, and Millar NS (2015)
Structurally similar allosteric modulators of a7 nicotinic acetylcholine receptors
exhibit five distinct pharmacological effects. J Biol Chem 290:35523562.
Gotti C and Clementi F (2004) Neuronal nicotinic receptors: from structure to pa-
thology. Prog Neurobiol 74:363396.
Grønlien JH, Ween H, Thorin-Hagene K, Cassar S, Li J, Briggs CA, Gopalakrishnan
M, and Malysz J (2010) Importance of M2-M3 loop in governing properties of
genistein at the a7 nicotinic acetylcholine receptor inferred from a7/5-HT3A chi-
mera. Eur J Pharmacol 647:3747.
Gu S, Matta JA, Lord B, Harrington AW, Sutton SW, Davini WB, and Bredt DS
(2016) Brain a7 Nicotinic Acetylcholine Receptor Assembly Requires NACHO.
Neuron 89:948955.
Guan Y-Z, Jin X-D, Guan L-X, Yan H-C, Wang P, Gong Z, Li S-J, Cao X, Xing Y-L,
and Gao T-M (2015) Nicotine inhibits microglial proliferation and is neuro-
protective in global ischemia rats. Mol Neurobiol 51:14801488.
Guan ZZ, Zhang X, Ravid R, and Nordberg A (2000) Decreased protein levels of
nicotinic receptor subunits in the hippocampus and temporal cortex of patients
with Alzheimers disease. J Neurochem 74:237243.
Guerra-Álvarez M, Moreno-Ortega AJ, Navarro E, Fernández-Morales JC, Egea J,
López MG, and Cano-Abad MF (2015) Positive allosteric modulation of alpha-7
nicotinic receptors promotes cell death by inducing Ca(21)release from the en-
doplasmic reticulum. J Neurochem 133:309319.
Hajós M, Hurst RS, Hoffmann WE, Krause M, Wall TM, Higdon NR, and Groppi VE
(2005) The selective alpha7 nicotinic acetylcholine receptor agonist PNU-282987
[N-[(3R)-1-Azabicyclo[2.2.2]oct-3-yl]-4-chlorobenzamide hydrochloride] enhances
GABAergic synaptic activity in brain slices and restores auditory gating deficits in
anesthetized rats. J Pharmacol Exp Ther 312:12131222.
Hansen SB and Taylor P (2007) Galanthamine and non-competitive inhibitor binding
to ACh-binding protein: evidence for a binding site on non-alpha-subunit interfaces
of heteromeric neuronal nicotinic receptors. J Mol Biol 369:895901.
Hashimoto K (2015) Tropisetron and its targets in Alzheimers disease. Expert Opin
Ther Targets 19:15.
Hashimoto T, Ichiki T, Watanabe A, Hurt-Camejo E, Michaëlsson E, Ikeda J, Inoue
E, Matsuura H, Tokunou T, Kitamoto S, et al. (2014) Stimulation of a7 nicotinic
acetylcholine receptor by AR-R17779 suppresses atherosclerosis and aortic aneu-
rysm formation in apolipoprotein E-deficient mice. Vascul Pharmacol 61:4955.
Hassaine G, Deluz C, Grasso L, Wyss R, Tol MB, Hovius R, Graff A, Stahlberg H,
Tomizaki T, Desmyter A, et al. (2014) X-ray structure of the mouse serotonin 5-
HT3 receptor. Nature 512:276281.
Hauser TA, Kucinski A, Jordan KG, Gatto GJ, Wersinger SR, Hesse RA, Stachowiak
EK, Stachowiak MK, Papke RL, Lippiello PM, et al. (2009) TC-5619: an alpha7
neuronal nicotinic receptor-selective agonist that demonstrates efficacy in animal
models of the positive and negative symptoms and cognitive dysfunction of
schizophrenia. Biochem Pharmacol 78:803812.
Hibbs RE and Gouaux E (2011) Principles of activation and permeation in an anion-
selective Cys-loop receptor. Nature 474:5460.
Hilf RJC and Dutzler R (2008) X-ray structure of a prokaryotic pentameric ligand-
gated ion channel. Nature 452:375379.
Hilf RJC and Dutzler R (2009) Structure of a potentially open state of a proton-
activated pentameric ligand-gated ion channel. Nature 457:115118.
Hu X-Q and Lovinger DM (2008) The L293 residue in transmembrane domain 2 of
the 5-HT3A receptor is a molecular determinant of allosteric modulation by 5-
hydroxyindole. Neuropharmacology 54:11531165.
Hurst R, Rollema H, and Bertrand D (2013) Nicotinic acetylcholine receptors: from
basic science to therapeutics. Pharmacol Ther 137:2254.
Hurst RS, Hajós M, Raggenbass M, Wall TM, Higdon NR, Lawson JA, Rutherford-
Root KL, Berkenpas MB, Hoffmann WE, Piotrowski DW, et al. (2005) A novel
positive allosteric modulator of the a7 neuronal nicotinic acetylcholine receptor: in
vitro and in vivo characterization. J Neurosci 25:43964405.
Iturriaga-Vásquez P, Alzate-Morales J, Bermudez I, Varas R, and Reyes-Parada M
(2015) Multiple binding sites in the nicotinic acetylcholine receptors: An opportu-
nity for polypharmacolgy. Pharmacol Res 101:917.
Jayakar SS, Dailey WP, Eckenhoff RG, and Cohen JB (2013) Identification of
propofol binding sites in a nicotinic acetylcholine receptor with a photoreactive
propofol analog. JBiolChem288:61786189.
Jones AK, Buckingham SD, and Sattelle DB (2010) Proteins interacting with nico-
tinic acetylcholine receptors: expanding functional and therapeutic horizons.
Trends Pharmacol Sci 31:455462.
Jones IW and Wonnacott S (2004) Precise localization of alpha7 nicotinic acetyl-
choline receptors on glutamatergic axon terminals in the rat ventral tegmental
area. J Neurosci 24:1124411252.
Kabbani N, Nordman JC, Corgiat BA, Veltri DP, Shehu A, Seymour VA, and Adams
DJ (2013) Are nicotinic acetylcholine receptors coupled to G proteins? BioEssays
35:10251034.
Kadir A, Almkvist O, Wall A, Långström B, and Nordberg A (2006) PET imaging of
cortical 11C-nicotine binding correlates with the cognitive function of attention in
Alzheimers disease. Psychopharmacology (Berl) 188:509520.
Kalappa BI, Sun F, Johnson SR, Jin K, and Uteshev VV (2013) A positive allosteric
modulator of a7 nAChRs augments neuroprotective effects of endogenous nicotinic
agonists in cerebral ischaemia. Br J Pharmacol 169:18621878.
Karamihalev S, Prickaerts J, and van Goethem NP (2014) Donepezil and the alpha-7
agonist PHA 568487, but not risperidone, ameliorate spatial memory deficits in a
subchronic MK-801 mouse model of cognitive impairment in schizophrenia. Behav
Brain Res 272:248251.
Kawashima K and Fujii T (2004) Expression of non-neuronal acetylcholine
in lymphocytes and its contribution to the regulation of immune function. Front
Biosci 9:20632085.
King JR, Nordman JC, Bridges SP, Lin M-K, and Kabbani N (2015) Identification
and characterization of a G protein-binding cluster in a7 nicotinic acetylcholine
receptors. J Biol Chem 290:2006020070.
Langley JN (1905) On the reaction of cells and of nerve-endings to certain poisons,
chiefly as regards the reaction of striated muscle to nicotine and to curari. J Physiol
33:374413.
Lape R, Colquhoun D, and Sivilotti LG (2008) On the nature of partial agonism in the
nicotinic receptor superfamily. Nature 454:722727.
a7 Modulation 297
at ASPET Journals on August 18, 2016molpharm.aspetjournals.orgDownloaded from
Le Novère N and Changeux JP (2001) LGICdb: the ligand-gated ion channel data-
base. Nucleic Acids Res 29:294295.
Le Novère N and Changeux JP (1995) Molecular evolution of the nicotinic acetyl-
choline receptor: an example of multigene family in excitable cells. J Mol Evol 40:
155172.
Lee WY and Sine SM (2005) Principal pathway coupling agonist binding to channel
gating in nicotinic receptors. Nature 438:243247.
Lendvai B, Kassai F, Szájli A, and Némethy Z (2013) a7 nicotinic acetylcholine re-
ceptors and their role in cognition. Brain Res Bull 93:8696.
Lester HA, Dibas MI, Dahan DS, Leite JF, and Dougherty DA (2004) Cys-loop re-
ceptors: new twists and turns. Trends Neurosci 27:329336.
Levin ED, Bettegowda C, Blosser J, and Gordon J (1999) AR-R17779, and alpha7 nic-
otinic agonist, improves learning and memory in rats. Behav Pharmacol 10:675680.
Li S-X, Huang S, Bren N, Noridomi K, Dellisanti CD, Sine SM, and Chen L (2011)
Ligand-binding domain of an a7-nicotinic receptor chimera and its complex with
agonist. Nat Neurosci 14:12531259.
Liu Q, Huang Y, Shen J, Steffensen S, and Wu J (2012) Functional a7b2 nicotinic
acetylcholine receptors expressed in hippocampal interneurons exhibit high sen-
sitivity to pathological level of amyloid bpeptides. BMC Neurosci 13:155.
Liu Q, Huang Y, Xue F, Simard A, DeChon J, Li G, Zhang J, Lucero L, Wang M,
Sierks M, et al. (2009) A novel nicotinic acetylcholine receptor subtype in basal
forebrain cholinergic neurons with high sensitivity to amyloid peptides. J Neurosci
29:918929.
Lovinger DM, Sikes S, and Zhou Q (2002) Rapid drug superfusion and kinetic
analysis, in Methods in Alcohol-Related Neuroscience Research (Liu Y et al. eds) pp
159187, CRC Press, Boca Raton, FL.
Ludwig J, Höffle-Maas A, Samochocki M, Luttmann E, Albuquerque EX, Fels G,
and Maelicke A (2010) Localization by site-directed mutagenesis of a galantamine
binding site on a7 nicotinic acetylcholine receptor extracellular domain. J Recept
Signal Transduct Res 30:469483.
Mackin KD, Maus AD, Pereira EF, Albuquerque EX, and Conti-Fine BM (1998)
Human vascular endothelial cells express functional nicotinic acetylcholine re-
ceptors. J Pharmacol Exp Ther 287:435439.
Malysz J, Grønlien JH, Anderson DJ, Håkerud M, Thorin-Hagene K, Ween H,
Wetterstrand C, Briggs CA, Faghih R, Bunnelle WH, et al. (2009) In vitro phar-
macological characterization of a novel allosteric modulator of alpha 7 neuronal
acetylcholine receptor, 4-(5-(4-chlorophenyl)-2-methyl-3-propionyl-1H-pyrrol-1-yl)
benzenesulfonamide (A-867744), exhibiting unique pharmacological profile.
J Pharmacol Exp Ther 330:257267.
Martelli D, McKinley MJ, and McAllen RM (2014) The cholinergic anti-inflammatory
pathway: a critical review. Auton Neurosci 182:6569.
Maus AD, Pereira EF, Karachunski PI, Horton RM, Navaneetham D, Macklin K,
Cortes WS, Albuquerque EX, and Conti-Fine BM (1998) Human and rodent
bronchial epithelial cells express functional nicotinic acetylcholine receptors. Mol
Pharmacol 54:779788.
Menze ET, Esmat A, Tadros MG, Abdel-Naim AB, and Khalifa AE (2015) Genistein
improves 3-NPA-induced memory impairment in ovariectomized rats: impact of its
antioxidant, anti-inflammatory and acetylcholinesterase modulatory properties.
PLoS One 10:e0117223.
Menze ET, Esmat A, Tadros MG, Khalifa AE, and Abdel-Naim AB (2016) Genistein
improves sensorimotor gating: Mechanisms related to its neuroprotective effects on
the striatum. Neuropharmacology 105:3546.
Mike A, Castro NG, and Albuquerque EX (2000) Choline and acetylcholine have
similar kinetic properties of activation and desensitization on the alpha7 nicotinic
receptors in rat hippocampal neurons. Brain Res 882:155168.
Miledi R and Potter LT (1971) Acetylcholine receptors in muscle fibres. Nature 233:
599603.
Millar NS (2008) RIC-3: a nicotinic acetylcholine receptor chaperone. Br J Pharmacol
153 (Suppl 1):S177S183.
Miller PS and Aricescu AR (2014) Crystal structure of a human GABAA receptor.
Nature 512:270275.
Möller-Acuña P, Contreras-Riquelme JS, Rojas-Fuentes C, Nuñez-Vivanco G, Alzate-
Morales J, Iturriaga-Vásquez P, Arias HR, and Reyes-Parada M (2015) Similarities
between the binding sites of SB-206553 at serotonin type 2 and alpha7 acetyl-
choline nicotinic receptors: rationale for its polypharmacological profile. PLoS One
10:e0134444.
Monod J, Wyman J, and Changeux JP (1965) On the nature of allosteric transitions:
A plausible model. J Mol Biol 12:88118.
Moretti M, Zoli M, George AA, Lukas RJ, Pistillo F, Maskos U, Whiteaker P,
and Gotti C (2014) The novel a7b2-nicotinic acetylcholine receptor subtype is
expressed in mouse and human basal forebrain: biochemical and pharmacological
characterization. Mol Pharmacol 86:306317.
Mowrey DD, Liu Q, Bondarenko V, Chen Q, Seyoum E, Xu Y, Wu J, and Tang P
(2013) Insights into distinct modulation of a7 and a7b2 nicotinic acetylcholine
receptors by the volatile anesthetic isoflurane. J Biol Chem 288:3579335800.
Mukhtasimova N, Lee WY, Wang H-L, and Sine SM (2009) Detection and trapping of
intermediate states priming nicotinic receptor channel opening. Nature 459:
451454.
Nemecz A and Taylor P (2011) Creating an a7 nicotinic acetylcholine recognition
domain from the acetylcholine-binding protein: crystallographic and ligand selec-
tivity analyses. J Biol Chem 286:4255542565.
Neri M, Bonassi S, and Russo P (2012) Genetic variations in CHRNA7 or CHRFAM7
and susceptibility to dementia. Curr Drug Targets 13:636643.
Ng HJ, Whittemore ER, Tran MB, Hogenkamp DJ, Broide RS, Johnstone TB, Zheng
L, Stevens KE, and Gee KW (2007) Nootropic a7 nicotinic receptor allosteric
modulator derived from GABAA receptor modulators. Proc Natl Acad Sci USA 104:
80598064.
Ni M, Yang Z-W, Li D-J, Li Q, Zhang S-H, Su D-F, Xie H-H, and Shen F-M (2010) A
potential role of alpha-7 nicotinic acetylcholine receptor in cardiac angiogenesis in
a pressure-overload rat model. J Pharmacol Sci 114:311319.
Nikiforuk A, Kos T, Potasiewicz A, and Popik P (2015) Positive allosteric modulation
of alpha 7 nicotinic acetylcholine receptors enhances recognition memory and
cognitive flexibility in rats. Eur Neuropsychopharmacol 25:13001313.
Nordman JC and Kabbani N (2012) An interaction between a7 nicotinic receptors
and a G-protein pathway complex regulates neurite growth in neural cells. J Cell
Sci 125:55025513.
Nury H, Van Renterghem C, Weng Y, Tran A, Baaden M, Dufresne V, Changeux
J-P, Sonner JM, Delarue M, and Corringer P-J (2011) X-ray structures of general
anaesthetics bound to a pentameric ligand-gated ion channel. Nature 469:
428431.
ODonnell CJ, Rogers BN, Bronk BS, Bryce DK, Coe JW, Cook KK, Duplantier AJ,
Evrard E, Hajós M, Hoffmann WE, et al. (2010) Discovery of 4-(5-methyloxazolo
[4,5-b]pyridin-2-yl)-1,4-diazabicyclo[3.2.2]nonane (CP-810,123), a novel a7 nico-
tinic acetylcholine receptor agonist for the treatment of cognitive disorders in
schizophrenia: synthesis, SAR development, and in vivo efficacy in cognition
models. J Med Chem 53:12221237.
Olincy A, Harris JG, Johnson LL, Pender V, Kongs S, Allensworth D, Ellis J, Zerbe
GO, Leonard S, Stevens KE, et al. (2006) Proof-of-concept trial of an alpha7 nico-
tinic agonist in schizophrenia. Arch Gen Psychiatry 63:630638.
Pałczy
nska MM, Jindrichova M, Gibb AJ, and Millar NS (2012) Activation of a7
nicotinic receptors by orthosteric and allosteric agonists: influence on single-
channel kinetics and conductance. Mol Pharmacol 82:910917.
Palma E, Bertrand S, Binzoni T, and Bertrand D (1996) Neuronal nicotinic alpha 7
receptor expressed in Xenopus oocytes presents five putative binding sites for
methyllycaconitine. J Physiol 491:151161.
Papke RL and Porter Papke JK (2002) Comparative pharmacology of rat and human
alpha7 nAChR conducted with net charge analysis. Br J Pharmacol 137:4961.
Parada E, Egea J, Buendia I, Negredo P, Cunha AC, Cardoso S, Soares MP,
and López MG (2013) The microglial a7-acetylcholine nicotinic receptor is a key
element in promoting neuroprotection by inducing heme oxygenase-1 via nuclear
factor erythroid-2-related factor 2. Antioxid Redox Signal 19:11351148.
Park HJ, Lee PH, Ahn YW, Choi YJ, Lee G, Lee D-Y, Chung ES, and Jin BK (2007)
Neuroprotective effect of nicotine on dopaminergic neurons by anti-inflammatory
action. Eur J Neurosci 26:7989.
Patrick J, Ballivet M, Boas L, Claudio T, Forrest J, Ingraham H, Mason P, Stengelin
S, Ueno S, and Heinemann S (1983) Molecular cloning of the acetylcholine receptor.
Cold Spring Harb Symp Quant Biol 48:7178.
Paulo JA, Brucker WJ, and Hawrot E (2009) Proteomic analysis of an alpha7 nico-
tinic acetylcholine receptor interactome. J Proteome Res 8:18491858.
Pichat P, Bergis OE, Terranova J-P, Urani A, Duarte C, Santucci V, Gueudet C, Voltz
C, Steinberg R, Stemmelin J, et al. (2007) SSR180711, a novel selective alpha7
nicotinic receptor partial agonist: (II) efficacy in experimental models predictive of
activity against cognitive symptoms of schizophrenia. Neuropsychopharmacology
32:1734.
Placzek AN, Grassi F, Papke T, Meyer EM, and Papke RL (2004) A single point
mutation confers properties of the muscle-type nicotinic acetylcholine receptor to
homomeric alpha7 receptors. Mol Pharmacol 66:169177.
Prickaerts J, van Goethem NP, Chesworth R, Shapiro G, Boess FG, Methfessel C,
Reneerkens OAH, Flood DG, Hilt D, Gawryl M, et al. (2012) EVP-6124, a novel and
selective a7 nicotinic acetylcholine receptor partial agonist, improves memory
performance by potentiating the acetylcholine response of a7 nicotinic acetylcho-
line receptors. Neuropharmacology 62:10991110.
Quik M, Zhang D, McGregor M, and Bordia T (2015) Alpha7 nicotinic receptors as
therapeutic targets for Parkinsons disease. Biochem Pharmacol 97:399407.
Rayes D, De Rosa MJ, Sine SM, and Bouzat C (2009) Number and locations of agonist
binding sites required to activate homomeric Cys-loop receptors. J Neurosci 29:
60226032.
Razani-Boroujerdi S, Boyd RT, Dávila-García MI, Nandi JS, Mishra NC, Singh SP,
Pena-Philippides JC, Langley R, and Sopori ML (2007) T cells express alpha7-
nicotinic acetylcholine receptor subunits that require a functional TCR and
leukocyte-specific protein tyrosine kinase for nicotine-induced Ca21response.
J Immunol 179:28892898.
Sahdeo S, Wallace T, Hirakawa R, Knoflach F, Bertrand D, Maag H, Misner D,
Tombaugh GC, Santarelli L, Brameld K, et al. (2014) Characterization of
RO5126946, a Novel a7 nicotinic acetylcholine receptor-positive allosteric modu-
lator. J Pharmacol Exp Ther 350:455468.
Sauguet L, Shahsavar A, Poitevin F, Huon C, Menny A, Nemecz À, Haouz A,
Changeux J-P, Corringer P-J, and Delarue M (2014) Crystal structures of a pen-
tameric ligand-gated ion channel provide a mechanism for activation. Proc Natl
Acad Sci USA 111:966971.
Schaaf CP (2014) Nicotinic acetylcholine receptors in human genetic disease. Genet
Med 16:649656.
Schirmer SU, Eckhardt I, Lau H, Klein J, DeGraaf YC, Lips KS, Pineau C, Gibbins
IL, Kummer W, Meinhardt A, et al. (2011) The cholinergic system in rat testis is of
non-neuronal origin. Reproduction 142:157166.
Séguéla P, Wadiche J, Dineley-Miller K, Dani JA, and Patrick JW (1993) Molecular
cloning, functional properties, and distribution of rat brain alpha 7: a nicotinic
cation channel highly permeable to calcium. J Neurosci 13:596604.
Sharma G and Vijayaraghavan S (2001) Nicotinic cholinergic signaling in hippo-
campal astrocytes involves calcium-induced calcium release from intracellular
stores. Proc Natl Acad Sci USA 98:41484153.
Shen JX and Yakel JL (2012) Functional a7 nicotinic ACh receptors on astrocytes in
rat hippocampal CA1 slices. J Mol Neurosci 48:1421.
Shiina A, Shirayama Y, Niitsu T, Hashimoto T, Yoshida T, Hasegawa T, Haraguchi
T, Kanahara N, Shiraishi T, Fujisaki M, et al. (2010) A randomised, double-blind,
placebo-controlled trial of tropisetron in patients with schizophrenia. Ann Gen
Psychiatry 9:27.
Shytle RD, Mori T, Townsend K, Vendrame M, Sun N, Zeng J, Ehrhart J, Silver AA,
Sanberg PR, and Tan J (2004) Cholinergic modulation of microglial activation by
alpha 7 nicotinic receptors. J Neurochem 89:337343.
298 Corradi and Bouzat
at ASPET Journals on August 18, 2016molpharm.aspetjournals.orgDownloaded from
Sine SM (2012) End-plate acetylcholine receptor: structure, mechanism, pharma-
cology, and disease. Physiol Rev 92:11891234.
Sine SM and Engel AG (2006) Recent advances in Cys-loop receptor structure and
function. Nature 440:448455.
Sinkus ML, Graw S, Freedman R, Ross RG, Lester HA, and Leonard S (2015) The
human CHRNA7 and CHRFAM7A genes: A review of the genetics, regulation, and
function. Neuropharmacology 96 (Pt B):274288.
Sinkus ML, Lee MJ, Gault J, Logel J, Short M, Freedman R, Christian SL, Lyon J,
and Leonard S (2009) A 2-base pair deletion polymorphism in the partial dupli-
cation of the alpha7 nicotinic acetylcholine gene (CHRFAM7A) on chromosome
15q14 is associated with schizophrenia. Brain Res 1291:111.
Spurny R, Debaveye S, Farinha A, Veys K, Vos AM, Gossas T, Atack J, Bertrand S,
Bertrand D, Danielson UH, et al. (2015) Molecular blueprint of allosteric binding
sites in a homologue of the agonist-binding domain of the a7 nicotinic acetylcholine
receptor. Proc Natl Acad Sci USA 112:E2543E2552.
Stuckenholz V, Bacher M, Balzer-Geldsetzer M, Alvarez-Fischer D, Oertel WH, Dodel
RC, and Noelker C (2013) The a7 nAChR agonist PNU-282987 reduces in-
flammation and MPTP-induced nigral dopaminergic cell loss in mice. J Parkinsons
Dis 3:161172.
Sun F, Jin K, and Uteshev VV (2013) A type-II positive allosteric modulator of a7
nAChRs reduces brain injury and improves neurological function after focal cere-
bral ischemia in rats. PLoS One 8:e73581 10.1371/journal.pone.0073581.
Sydserff S, Sutton EJ, Song D, Quirk MC, Maciag C, Li C, Jonak G, Gurley D, Gordon
JC, Christian EP, et al. (2009) Selective a7 nicotinic receptor activation by
AZD0328 enhances cortical dopamine release and improves learning and atten-
tional processes. Biochem Pharmacol 78:880888.
Szabo AK, Pesti K, Mike A, and Vizi ES (2014) Mode of action of the positive mod-
ulator PNU-120596 on a7 nicotinic acetylcholine receptors. Neuropharmacology
81:4254.
Taly A and Changeux J-P (2008) Functional organization and conformational dy-
namics of the nicotinic receptor: a plausible structural interpretation of myasthenic
mutations. Ann N Y Acad Sci 1132:4252.
Targowska-Duda KM, Wnorowski A, Budzynska B, Jozwiak K, Biala G, and Arias
HR (2016) The positive allosteric modulator of a7 nicotinic acetylcholine receptors,
3-furan-2-yl-N-p-tolyl-acrylamide, enhances memory processes and stimulates
ERK1/2 phosphorylation in mice. Behav Brain Res 302:142151.
Tasneem A, Iyer LM, Jakobsson E, and Aravind L (2005) Identification of the pro-
karyotic ligand-gated ion channels and their implications for the mechanisms and
origins of animal Cys-loop ion channels. Genome Biol 6:R4.
Terry AV, Jr, Callahan PM, and Hernandez CM (2015) Nicotinic ligands as multi-
functional agents for the treatment of neuropsychiatric disorders. Biochem Phar-
macol 97:388398.
Thomsen MS and Mikkelsen JD (2012) Type I and II positive allosteric modulators
differentially modulate agonist-induced up-regulation of a7 nicotinic acetylcholine
receptors. J Neurochem 123:7383.
Thomsen MS, Zwart R, Ursu D, Jensen MM, Pinborg LH, Gilmour G, Wu J, Sher E,
and Mikkelsen JD (2015) a7 and b2 nicotinic acetylcholine receptor subunits form
heteromeric receptor complexes that are expressed in the human cortex and dis-
play distinct pharmacological properties. PLoS One 10:e0130572 10.1371/journal.
pone.0130572.
Timmermann DB, Grønlien JH, Kohlhaas KL, Nielsen EØ, Dam E, Jørgensen TD,
Ahring PK, Peters D, Holst D, Christensen JK, et al. (2007) An allosteric modulator
of the a7 nicotinic acetylcholine receptor possessing cognition-enhancing properties
in vivo. J Pharmacol Exp Ther 323:294307.
Tsuneki H, Klink R, Léna C, Korn H, and Changeux JP (2000) Calcium mobilization
elicited by two types of nicotinic acetylcholine receptors in mouse substantia nigra
pars compacta. Eur J Neurosci 12:24752485.
Uteshev VV (2014) The therapeutic promise of positive allosteric modulation of nic-
otinic receptors. Eur J Pharmacol 727:181185.
Uteshev V (2016) Are positive allosteric modulators of a7 nAChRs clinically safe?
J Neurochem 136:217219.
Van Kampen M, Selbach K, Schneider R, Schiegel E, Boess F, and Schreiber R (2004)
AR-R 17779 improves social recognition in rats by activation of nicotinic alpha7
receptors. Psychopharmacology (Berl) 172:375383.
van Westerloo DJ, Giebelen IA, Florquin S, Bruno MJ, Larosa GJ, Ulloa L, Tracey KJ,
and van der Poll T (2006) The vagus nerve and nicotinic receptors modulate exper-
imental pancreatitis severity in mice. Gastroenterology 130:18221830.
Vukelic M, Qing X, Redecha P, Koo G, and Salmon JE (2013) Cholinergic receptors
modulate immune complex-induced inflammation in vitro and in vivo. J Immunol
191:18001807.
Wallace TL and Bertrand D (2013) Alpha7 neuronal nicotinic receptors as a drug
target in schizophrenia. Expert Opin Ther Targets 17:139155.
Wallace TL, Callahan PM, Tehim A, Bertrand D, Tombaugh G, Wang S, Xie W, Rowe
WB, Ong V, Graham E, et al. (2011) RG3487, a novel nicotinic a7 receptor partial
agonist, improves cognition and sensorimotor gating in rodents. J Pharmacol Exp
Ther 336:242253.
Wallace TL and Porter RHP (2011) Targeting the nicotinic alpha7 acetylcholine re-
ceptor to enhance cognition in disease. Biochem Pharmacol 82:891903.
Wang Y, Xiao C, Indersmitten T, Freedman R, Leonard S, and Lester HA (2014) The
duplicated a7 subunits assemble and form functional nicotinic receptors with the
full-length a7. J Biol Chem 289:2645126463.
Wessler I and Kirkpatrick CJ (2008) Acetylcholine beyond neurons: the non-neuronal
cholinergic system in humans. Br J Pharmacol 154:15581571.
Williams DK, Peng C, Kimbrell MR, and Papke RL (2012) Intrinsically low open
probability of a7 nicotinic acetylcholine receptors can be overcome by positive al-
losteric modulation and serum factors leading to the generation of excitotoxic
currents at physiological temperatures. Mol Pharmacol 82:746759.
Williams DK, Stokes C, Horenstein NA, and Papke RL (2011a) The effective opening
of nicotinic acetylcholine receptors with single agonist binding sites. J Gen Physiol
137:369384.
Williams DK, Wang J, and Papke RL (2011b) Investigation of the molecular mechanism
of the a7 nicotinic acetylcholine receptor positive allosteric modulator PNU-120596
provides evidence for two distinct desensitized states. Mol Pharmacol 80:10131032.
Williams DK, Wang J, and Papke RL (2011c) Positive allosteric modulators as an
approach to nicotinic acetylcholine receptor-targeted therapeutics: advantages and
limitations. Biochem Pharmacol 82:915930.
Williams ME, Burton B, Urrutia A, Shcherbatko A, Chavez-Noriega LE, Cohen CJ,
and Aiyar J (2005) Ric-3 promotes functional expression of the nicotinic acetyl-
choline receptor alpha7 subunit in mammalian cells. J Biol Chem 280:12571263.
Wonnacott S and Barik J (2007) Nicotinic ACh Receptors. Tocris Rev 28, http://www.
komabiotech.co.kr/pdf/nicotinic_ach_receptors_review.pdf.
Yan H, Pan N, Xue F, Zheng Y, Li C, Chang Y, Xu Z, Yang H, and Zhang J (2015) The
coupling interface and pore domain codetermine the single-channel activity of the
a7 nicotinic receptor. Neuropharmacology 95:448458.
Yang J-S, Seo SW, Jang S, Jung GY, and Kim S (2012) Rational engineering of
enzyme allosteric regulation through sequence evolution analysis. PLOS Comput
Biol 8:e1002612 10.1371/journal.pcbi.1002612.
Young GT, Zwart R, Walker AS, Sher E, and Millar NS (2008) Potentiation of a7
nicotinic acetylcholine receptors via an allosteric transmembrane site. Proc Natl
Acad Sci USA 105:1468614691.
Zdanowski R, Krzy_
zowska M, Ujazdowska D, Lewicka A, and Lewicki S (2015) Role of
a7 nicotinic receptor in the immune system and intracellular signaling pathways.
Cent Eur J Immunol 40:373379.
Zhang J, Xue F, Liu Y, Yang H, and Wang X (2013) The structural mechanism of the
Cys-loop receptor desensitization. Mol Neurobiol 48:97108.
Zhou Q, Verdoorn TA, and Lovinger DM (1998) Alcohols potentiate the function of
5-HT3 receptor-channels on NCB-20 neuroblastoma cells by favouring and sta-
bilizing the open channel state. JPhysiol507:335352.
Zia S, Ndoye A, Nguyen VT, and Grando SA (1997) Nicotine enhances expression of
the alpha 3, alpha 4, alpha 5, and alpha 7 nicotinic receptors modulating calcium
metabolism and regulating adhesion and motility of respiratory epithelial cells.
Res Commun Mol Pathol Pharmacol 97:243262.
Zimmermann I, Marabelli A, Bertozzi C, Sivilotti LG, and Dutzler R (2012) Inhibition
of the prokaryotic pentameric ligand-gated ion channel ELIC by divalent cations.
PLoS Biol 10:e1001429.
Zoli M, Pistillo F, and Gotti C (2015) Diversity of native nicotinic receptor subtypes in
mammalian brain. Neuropharmacology 96 (Pt B):302311.
Address correspondence to: Dr. Cecilia Bouzat, Instituto de Investigaciones
Bioquímicas de Bahía Blanca, CONICET/Universidad Nacional del Sur,
Camino La Carrindanga Km 7, Bahía Blanca, Argentina. E-mail: inbouzat@
criba.edu.ar
a7 Modulation 299
at ASPET Journals on August 18, 2016molpharm.aspetjournals.orgDownloaded from
... These anti-inflammatory pathways are independent of calcium ion influx during activation of microglial α7 nAChRs. Therefore, microglial α7 nAChRs have been proposed to be metabotropic receptors [60,73,74]. ...
Article
Full-text available
Neuroinflammation contributes to the pathophysiology of major depressive disorder (MDD) by inducing neuronal excitability via dysregulation of microglial brain-derived neurotrophic factor (BDNF), Na-K-Cl cotransporter-1 (NKCC1), and K-Cl cotransporter-2 (KCC2) due to activation of BDNF-tropomyosin receptor kinase B (TrkB) signaling. Allosteric modulation of α7 nAChRs has not been investigated on BDNF, KCC2, and NKCC1 during LPS-induced depressive-like behavior. Therefore, we examined the effects of PNU120596, an α7 nAChR positive allosteric modulator, on the expression of BDNF, KCC2, and NKCC1 in the hippocampus and prefrontal cortex using Western blot analysis, immunofluorescence assay, and real-time polymerase chain reaction. The effects of ANA12, a TrkB receptor antagonist, on LPS-induced cognitive deficit and depressive-like behaviors were determined using the Y-maze, tail suspension test (TST), and forced swim test (FST). Pharmacological interactions between PNU120596 and ANA12 were also examined. Experiments were conducted in male C57BL/6J mice. LPS administration (1 mg/kg) resulted in increased expression of BDNF and the NKCC1/KCC2 ratio and decreased expression of KCC2 in the hippocampus and prefrontal cortex. PNU120596 pretreatment (4 mg/kg) attenuated the LPS-induced increase in the expression of BDNF and NKCC1/KCC2 ratio and the reduction in KCC2 expression in these brain regions. In addition, ANA12 (0.25 or 0.50 mg/kg) reduced the LPS-induced cognitive deficit and depressive-like behaviors measured by a reduced spontaneous alternation in the Y-maze and increased immobility duration in TST and FST. Coadministration of PNU120596 (1 mg/kg) and ANA12 (0.25 mg/kg) prevented the LPS-induced cognitive deficit and depressive-like behaviors. Overall, PNU120596 prevented the LPS-induced depressive-like behavior by likely decreasing neuronal excitability via targeting microglial α7 nAChR in the hippocampus and prefrontal cortex.
... nAChRs are pentameric integral membrane proteins belonging to the Cys-loop superfamily of ligand-gated ion channels [35][36][37][38]. Each of its five subunits possesses a large extracellular region that harbors the agonist-binding site, a transmembrane region with extensive contacts with surrounding lipids through evolutionarily conserved structural motifs [34,[39][40][41] and an intracellular region containing modulatory sites and determinants of channel conductance [42,43]. The transmembrane domain consists of four segments (TM1-TM4), with TM2 forming the walls of the ion channel pore and TM1, TM3, and TM4 being more externally located [34,44,45]. ...
Article
Full-text available
Professor Carlos Gutiérrez-Merino, a prominent scientist working in the complex realm of biological membranes, has made significant theoretical and experimental contributions to the field. Contemporaneous with the development of the fluid-mosaic model of Singer and Nicolson, the Förster resonance energy transfer (FRET) approach has become an invaluable tool for studying molecular interactions in membranes, providing structural insights on a scale of 1–10 nm and remaining important alongside evolving perspectives on membrane structures. In the last few decades, Gutiérrez-Merino’s work has covered multiple facets in the field of FRET, with his contributions producing significant advances in quantitative membrane biology. His more recent experimental work expanded the ground concepts of FRET to high-resolution cell imaging. Commencing in the late 1980s, a series of collaborations between Gutiérrez-Merino and the authors involved research visits and joint investigations focused on the nicotinic acetylcholine receptor and its relation to membrane lipids, fostering a lasting friendship.
Article
PDZ domains are modular domains that conventionally bind to C terminal or internal motifs of target proteins to control cellular functions through the regulation of protein complex assemblies. Almost all reported structures of PDZ-target protein complexes rely on fragments or peptides as target proteins. No intact target protein complexed with PDZ was structurally characterized. In this study, we used NMR spectroscopy and other biochemistry and biophysics tools to uncover insights into structural coupling between the PDZ domain of protein interacting with C-kinase 1 (PICK1) and α7 nicotinic acetylcholine receptors (α7 nAChR). Notably, the intracellular domains of both α7 nAChR and PICK1 PDZ exhibit a high degree of plasticity in their coupling. Specifically, the MA helix of α7 nAChR interacts with residues lining the canonical binding site of the PICK1 PDZ, while flexible loops also engage in protein–protein interactions. Both hydrophobic and electrostatic interactions mediate the coupling. Overall, the resulting structure of the α7 nAChR-PICK1 complex reveals an unconventional PDZ binding mode, significantly expanding the repertoire of functionally important PDZ interactions.
Article
Introduction: The landscape of epilepsy treatment has undergone a significant transformation with the emergence of cannabidiol as a potential therapeutic agent. Epidiolex, a pharmaceutical formulation of highly purified CBD, garnered significant attention not just for its therapeutic potential but also for being the first cannabis-derived medication to obtain approval from regulatory bodies. Area covered: In this narrative review the authors explore the intricate landscape of CBD as an antiseizure medication, deepening into its pharmacological mechanisms and clinical trials involving various epileptic encephalopathies. This exploration serves as a comprehensive guide, shedding light on a compound that holds promise for individuals contending with the significant challenges of drug-resistant epilepsy. Expert opinion: Rigorous studies highlight cannabidiol's efficacy, safety profile, and potential cognitive benefits, warranting further exploration for its approval in various drug-resistant epilepsy forms. As a promising therapeutic option, cannabidiol not only demonstrates efficacy in seizure control but also holds the potential for broader enhancements in the quality of life, especially for patients with epileptic encephalopathies.
Article
Full-text available
This review summarizes our research on nicotinic acetylcholine receptors in human chromaffin cells. Limited research has been conducted in this field on human tissue, primarily due to the difficulties associated with obtaining human cells. Receptor subtypes were characterized here using molecular biology and electrophysiological patch-clamp techniques. However, the most significant aspect of this study refers to the cross-talk between the two main subtypes identified in these cells, the α7- and α3β4* subtypes, aiming to avoid their desensitization. The article also reviews other aspects, including the regulation of their expression, function or physical interaction by choline, Ca2+, and tyrosine and serine/threonine phosphatases. Additionally, the influence of sex on their expression is also discussed.
Article
Full-text available
To determine whether 3-furan-2-yl-N-p-tolyl-acrylamide (PAM-2), a positive allosteric modulator of α7 nicotinic acetylcholine receptors (nAChRs), improves memory processes, passive avoidance tests were conducted in male mice after acute and chronic treatments. To determine the neuronal mechanisms underlying the promnesic activity elicited by PAM-2, the effect of this ligand on α7 nAChR up-regulation and ERK1/2 phosphorylation was assessed in the hippocampus and prefrontal cortex. The results indicate that: (1) PAM-2 improves memory acquisition/consolidation after acute treatment (Day 2) and memory consolidation after chronic treatment (Day 22). Although no effect was observed on α7 nAChR up-regulation, the chronic, but not acute, PAM-2 treatment increases ERK1/2 kinase phosphorylation, (2) the promnesic activity of PAM-2 was inhibited by methyllycaconitine, a selective α7-antagonist, confirming the role of α7 nAChRs, (3) a synergistic (acute) effect was observed between inactive doses of PAM-2 (0.1 mg/kg) and DMXBA (0.3 mg/kg), a selective α7-agonist, and (4) PAM-2 reversed the memory impairment elicited by scopolamine, a muscarinic antagonist. The results demonstrate that PAM-2 presents promnesic activity mediated by α7 nAChRs, and is able to trigger ERK1/2 phosphorylation only after chronic treatment.
Article
Full-text available
Acetylcholine has been well known as one of the most exemplary neurotransmitters. In humans, this versatile molecule and its synthesizing enzyme, choline acetyltransferase, have been found in various non-neural tissues such as the epithelium, endothelium, mesothelium muscle, blood cells and immune cells. The non-neuronal acetylcholine is accompanied by the expression of acetylcholinesterase and nicotinic/muscarinic acetylcholine receptors. Increasing evidence of the non-neuronal acetylcholine system found throughout the last few years has indicated this neurotransmitter as one of the major cellular signaling molecules (associated e.g. with kinases and transcription factors activity). This system is responsible for maintenance and optimization of the cellular function, such as proliferation, differentiation, adhesion, migration, intercellular contact and apoptosis. Additionally, it controls proper activity of immune cells and affects differentiation, antigen presentation or cytokine production (both pro- and anti-inflammatory). The present article reviews recent findings about the non-neuronal cholinergic system in the field of immune system and intracellular signaling pathways.
Chapter
Ligand-gated ion channels are a family of proteins specialized for the very rapid generation of transmembrane electrical current in response to binding of a neurotransmitter or other ligand. The ligand-gated ion channel subtypes that respond to neurotransmitters are found throughout the nervous system and come in several different varieties specifically activated by certain neurotransmitters. These include the subfamilies of ligand-gated receptor channels similar to the nicotinic acetylcholine receptors (nAChRs), the ionotropic glutamate receptors, and the ionotropic ATP receptors (see Lovinger, 1997 for review). The functional hallmarks of these proteins are their ability to bind neurotransmitter and to open an ion channel that is an intrinsic part of the receptor protein complex. Normally, these proteins reside at synapses where concentrations of neurotransmitter can reach near-millimolar levels within microseconds and dissipate nearly as rapidly. Thus, the ligand-gated ion channels normally operate on a very fast time scale dictated by the rapid transient rises in synaptic neurotransmitter concentrations. By necessity, the kinetic state transitions within the receptor channels must occur very rapidly. This presents special challenges for the study of ligand-gated ion channel function because the investigator must take into account the normal pattern of neurotransmitter release, as well as the rapid state transitions of the channels when designing such experiments.
Article
Activation of nicotinic acetylcholine receptors (nAChRs) is associated with the binding of agonists such as acetylcholine to an extracellular site that is located at the interface between two adjacent receptor subunits. More recently, there has been considerable interest in compounds, such as positive and negative allosteric modulators (PAMs and NAMs), that are able to modulate nAChR function by binding to distinct allosteric sites. Here we examined a series of compounds differing only in methyl substitution of a single aromatic ring. This series of compounds includes a previously described ?7-selective allosteric agonist, cis-cis-4-p-tolyl-3a,4,5,9b-tetrahydro-3H-cyclopenta[c]quinoline-8-sulfonamide (4MP-TQS), together with all other possible combinations of methyl substitution at a phenyl ring (18 additional compounds). Studies conducted with this series of compounds have revealed five distinct pharmacological effects on ?7 nAChRs. These five effects can be summarized as: 1) nondesensitizing activation (allosteric agonists), 2) potentiation associated with minimal effects on receptor desensitization (type I PAMs), 3) potentiation associated with reduced desensitization (type II PAMs), 4) noncompetitive antagonism (NAMs), and 5) compounds that have no effect on orthosteric agonist responses but block allosteric modulation (silent allosteric modulators (SAMs)). Several lines of experimental evidence are consistent with all of these compounds acting at a common, transmembrane allosteric site. Notably, all of these chemically similar compounds that have been classified as nondesensitizing allosteric agonists or as nondesensitizing (type II) PAMs are cis-cis-diastereoisomers, whereas all of the NAMs, SAMs, and type I PAMs are cis-trans-diastereoisomers. Our data illustrate the remarkable pharmacological diversity of allosteric modulators acting on nAChRs.
Article
Enhancement of α7 nicotinic receptor (nAChR) function by positive allosteric modulators (PAMs) is a promising therapeutic strategy to improve cognitive deficits. PAMs have been classified only on the basis of their macroscopic effects as type I, which only enhance agonist-induced currents, and type II, which also decrease desensitization and reactivate desensitized nAChRs. To decipher the molecular basis underlying these distinct activities, we explored the effects on single-α7 channel currents of representative members of each type and of less characterized compounds. Our results reveal that all PAMs enhance open-channel lifetime and produce episodes of successive openings, thus indicating that both types affect α7 kinetics. Different PAM types show different sensitivity to temperature, suggesting different mechanisms of potentiation. By using a mutant α7 receptor that is insensitive to the prototype type II PAM (PNU-120596), we show that some though not all type I PAMs share the structural determinants of potentiation. Overall, our study provides novel information on α7 potentiation, which is key to the ongoing development of therapeutic compounds.
Article
Nicotine exerts its behavioral and additive actions through a family of brain nicotinic acetylcholine receptors (nAChRs). Enhancing α7-type nAChR signaling improves symptoms in Alzheimer's disease and schizophrenia. The pharmaceutical study of α7 receptors is hampered because these receptors do not form their functional pentameric structure in cell lines, and mechanisms that underlie α7 receptor assembly in neurons are not understood. Here, a genomic screening strategy solves this long-standing puzzle and identifies NACHO, a transmembrane protein of neuronal endoplasmic reticulum that mediates assembly of α7 receptors. NACHO promotes α7 protein folding, maturation through the Golgi complex, and expression at the cell surface. Knockdown of NACHO in cultured hippocampal neurons or knockout of NACHO in mice selectively and completely disrupts α7 receptor assembly and abolishes α7 channel function. This work identifies NACHO as an essential, client-specific chaperone for nAChRs and has implications for physiology and disease associated with these widely distributed neurotransmitter receptors.
Article
Huntington's disease (HD) is a neurodegenerative disorder, characterized by selective atrophy in the striatum, particularly the medium spiny GABAergic efferent neurons. This results in striatal sensorimotor gating deficits. Systemic administration of 3-nitropropionic acid (3-NPA) produces selective lesions mimicking those of HD. Males were found to be more susceptible to 3-NPA-induced neurotoxicity than females, suggesting neuroprotective effects of estrogens. Phytoestrogens, including genistein, are good estrogenic alternatives that keep their beneficial effects on non-reproductive organs and lack the potential hazardous side effects. The current study was designed to investigate the potential beneficial effects of genistein in 3-NPA-induced HD in ovariectomized rats. Results showed that 3-NPA (20 mg/kg) administration caused significant disruption of the rats' locomotor activity and prepulse inhibition. In addition, it decreased striatal ATP levels and increased oxidative stress, inflammatory and apoptotic markers with striatal focal hemorrhage and gliosis. Pretreatment with 17β-estradiol (2.5 mg/kg) or genistein (20 mg/kg) led to a significant improvement of behavioral parameters, increased ATP production, decreased oxidative stress, attenuated inflammation and apoptosis. Therefore, this study suggests potential neuroprotective effects of genistein in ovariectomized rats challenged with 3-NPA.
Article
Lymphocytes express most components of the cholinergic system including acetylcholine (ACh), muscarinic and nicotinic ACh receptors (mAChRs and nAChRs, respectively), choline acetyltransferase (ChAT), high affinity choline transporter and acetylcholinesterase. ACh and mAChR agonists elicit intracellular Ca2+ signaling, up-regulation of c-fos expression and nitric oxide synthesis within T and B cells probably via M3 and M5 mAChRs. Stimulation of nAChRs with ACh or nicotine causes a rapid and transient Ca2+ signaling in T and B cells, probably via alpha7 nAChR subunit-mediated pathways. Phytohemagglutinin- or antigen-induced T cell activation via cell surface molecules (e.g., T cell receptor/CD3 complexes) enhances lymphocytic cholinergic transmission by up-regulating ChAT and M5 mAChR expression. It is thus likely that a local lymphocytic cholinergic system is involved in regulating immune function. This idea is supported by the findings that lymphocytic cholinergic activity is altered in animal models exhibiting immunological abnormalities. In addition, it appears likely that during interactions mediated by cell surface molecules T cells communicate via ACh with thymic epithelial cells and vascular endothelial cells, which also express ChAT and nAChRs or mAChRs. This interaction leads to T cell selection and maturation in the thymus and local vascular smooth muscle relaxation. Collectively, these data provide a compelling picture in which lymphocytes constitute a cholinergic system that is independent of cholinergic nerves, and which is involved in the regulation of immune function and local circulation.