Functional expression and axonal transport of α7 nAChRs by peptidergic nociceptors of rat dorsal...

15
ORIGINAL ARTICLE Functional expression and axonal transport of a7 nAChRs by peptidergic nociceptors of rat dorsal root ganglion Irina Shelukhina Renate Paddenberg Wolfgang Kummer Victor Tsetlin Received: 11 November 2013 / Accepted: 19 March 2014 Ó Springer-Verlag Berlin Heidelberg 2014 Abstract In recent pain studies on animal models, a7 nicotinic acetylcholine receptor (nAChR) agonists dem- onstrated analgesic, anti-hyperalgesic and anti-inflamma- tory effects, apparently acting through some peripheral receptors. Assuming possible involvement of a7 nAChRs on nociceptive sensory neurons, we investigated the mor- phological and neurochemical features of the a7 nAChR- expressing subpopulation of dorsal root ganglion (DRG) neurons and their ability to transport a7 nAChR axonally. In addition, a7 receptor activity and its putative role in pain signal neurotransmitter release were studied. Medium- sized a7 nAChR-expressing neurons prevailed, although the range covered all cell sizes. These cells accounted for one-fifth of total medium and large DRG neurons and \ 5 % of small ones. 83.2 % of a7 nAChR-expressing DRG neurons were peptidergic nociceptors (CGRP-im- munopositive), one half of which had non-myelinated C-fibers and the other half had myelinated Ad- and likely Aa/b-fibers, whereas 15.2 % were non-peptidergic C-fiber nociceptors binding isolectin B4. All non-peptidergic and a third of peptidergic a7 nAChR-bearing nociceptors expressed TRPV1, a capsaicin-sensitive noxious stimulus transducer. Nerve crush experiments demonstrated that CGRPergic DRG nociceptors axonally transported a7 nAChRs both to the spinal cord and periphery. a7 nAChRs in DRG neurons were functional as their specific agonist PNU282987 evoked calcium rise enhanced by a7-selective positive allosteric modulator PNU120596. However, a7 nAChRs do not modulate neurotransmitter CGRP and glutamate release from DRG neurons since nicotinic ligands affected neither their basal nor provoked levels, showing the necessity of further studies to elucidate the true role of a7 nAChRs in those neurons. Keywords Sensory ganglia Nicotinic acetylcholine receptor Nociception Nociceptor Calcitonin gene- related peptide Introduction a7 Nicotinic acetylcholine receptors (nAChRs) are ligand- gated homopentameric cation channels formed by five identical a7 subunits. Less frequently they combine a heteropentameric a7b2 receptor subtype (Cuevas et al. 2000; Wu and Lukas 2011; Murray et al. 2012). Homo- pentameric a7 nAChR is characterized by high calcium permeability, very fast and complete desensitization at high agonist concentrations, and sensitivity to selective agonists such as PNU282987 and to competitive antagonists such as long-chain a-neurotoxins, e.g. a-bungarotoxin (Bgt) (Tsetlin et al. 2009; Wu and Lukas 2011; Gundisch and Eibl 2011). a7 nAChRs are widespread in the nervous system, the majority have a presynaptic localization and modulate neurotransmitter release, but some are postsyn- aptic and mediate fast synaptic transmission or activate downstream signaling (Gotti et al. 2009; Berg and Conroy 2002; Marchi and Grilli 2010). Activation of a7 nAChRs in CNS improves learning, memory and attention deficiency (Thomsen et al. 2010) and relieves pain (Damaj et al. 2000; Abdin et al. 2006; Medhurst et al. 2008). In addition, pain animal model studies have shown analgesic, I. Shelukhina (&) V. Tsetlin Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, Miklukho-Maklaya str., 16/10, 117997 Moscow, Russia e-mail: [email protected] R. Paddenberg W. Kummer Institute for Anatomy and Cell Biology, Justus-Liebig- University, 35385 Giessen, Germany 123 Brain Struct Funct DOI 10.1007/s00429-014-0762-4

Transcript of Functional expression and axonal transport of α7 nAChRs by peptidergic nociceptors of rat dorsal...

Page 1: Functional expression and axonal transport of α7 nAChRs by peptidergic nociceptors of rat dorsal root ganglion

ORIGINAL ARTICLE

Functional expression and axonal transport of a7 nAChRsby peptidergic nociceptors of rat dorsal root ganglion

Irina Shelukhina • Renate Paddenberg •

Wolfgang Kummer • Victor Tsetlin

Received: 11 November 2013 / Accepted: 19 March 2014

� Springer-Verlag Berlin Heidelberg 2014

Abstract In recent pain studies on animal models, a7

nicotinic acetylcholine receptor (nAChR) agonists dem-

onstrated analgesic, anti-hyperalgesic and anti-inflamma-

tory effects, apparently acting through some peripheral

receptors. Assuming possible involvement of a7 nAChRs

on nociceptive sensory neurons, we investigated the mor-

phological and neurochemical features of the a7 nAChR-

expressing subpopulation of dorsal root ganglion (DRG)

neurons and their ability to transport a7 nAChR axonally.

In addition, a7 receptor activity and its putative role in pain

signal neurotransmitter release were studied. Medium-

sized a7 nAChR-expressing neurons prevailed, although

the range covered all cell sizes. These cells accounted for

one-fifth of total medium and large DRG neurons and

\5 % of small ones. 83.2 % of a7 nAChR-expressing

DRG neurons were peptidergic nociceptors (CGRP-im-

munopositive), one half of which had non-myelinated

C-fibers and the other half had myelinated Ad- and likely

Aa/b-fibers, whereas 15.2 % were non-peptidergic C-fiber

nociceptors binding isolectin B4. All non-peptidergic and a

third of peptidergic a7 nAChR-bearing nociceptors

expressed TRPV1, a capsaicin-sensitive noxious stimulus

transducer. Nerve crush experiments demonstrated that

CGRPergic DRG nociceptors axonally transported a7

nAChRs both to the spinal cord and periphery. a7 nAChRs

in DRG neurons were functional as their specific agonist

PNU282987 evoked calcium rise enhanced by a7-selective

positive allosteric modulator PNU120596. However, a7

nAChRs do not modulate neurotransmitter CGRP and

glutamate release from DRG neurons since nicotinic

ligands affected neither their basal nor provoked levels,

showing the necessity of further studies to elucidate the

true role of a7 nAChRs in those neurons.

Keywords Sensory ganglia � Nicotinic acetylcholine

receptor � Nociception � Nociceptor � Calcitonin gene-

related peptide

Introduction

a7 Nicotinic acetylcholine receptors (nAChRs) are ligand-

gated homopentameric cation channels formed by five

identical a7 subunits. Less frequently they combine a

heteropentameric a7b2 receptor subtype (Cuevas et al.

2000; Wu and Lukas 2011; Murray et al. 2012). Homo-

pentameric a7 nAChR is characterized by high calcium

permeability, very fast and complete desensitization at high

agonist concentrations, and sensitivity to selective agonists

such as PNU282987 and to competitive antagonists such as

long-chain a-neurotoxins, e.g. a-bungarotoxin (Bgt)

(Tsetlin et al. 2009; Wu and Lukas 2011; Gundisch and

Eibl 2011). a7 nAChRs are widespread in the nervous

system, the majority have a presynaptic localization and

modulate neurotransmitter release, but some are postsyn-

aptic and mediate fast synaptic transmission or activate

downstream signaling (Gotti et al. 2009; Berg and Conroy

2002; Marchi and Grilli 2010). Activation of a7 nAChRs in

CNS improves learning, memory and attention deficiency

(Thomsen et al. 2010) and relieves pain (Damaj et al. 2000;

Abdin et al. 2006; Medhurst et al. 2008). In addition,

pain animal model studies have shown analgesic,

I. Shelukhina (&) � V. Tsetlin

Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry

RAS, Miklukho-Maklaya str., 16/10, 117997 Moscow, Russia

e-mail: [email protected]

R. Paddenberg � W. Kummer

Institute for Anatomy and Cell Biology, Justus-Liebig-

University, 35385 Giessen, Germany

123

Brain Struct Funct

DOI 10.1007/s00429-014-0762-4

Page 2: Functional expression and axonal transport of α7 nAChRs by peptidergic nociceptors of rat dorsal root ganglion

anti-hyperalgesic and anti-inflammatory effects of a7

agonists acting through unidentified peripheral sites (Gurun

et al. 2009; Pacini et al. 2010; Wang et al. 2005; Feuerbach

et al. 2009; Rowley et al. 2010). Partially these effects

might be mediated by a7 nAChRs on immune cells through

cholinergic anti-inflammatory reflex (Huston 2012; Wang

et al. 2003; Nizri and Brenner 2011). Another prominent

peripheral target for a7 agonists is sensory axons of dorsal

root ganglion (DRG). Although the presence of different

nAChRs in DRG was shown by autoradiography, RT-PCR,

histochemical and electrophysiological studies (Sucher

et al. 1990; Boyd et al. 1991; Genzen et al. 2001; Haber-

berger et al. 2004; Shelukhina et al. 2009; Rau et al. 2005;

Ninkovic and Hunt 1983), it is not yet clear what types of

sensory modalities are perceived by DRG neurons

expressing a7 nAChRs and what direct function these

receptors served for.

DRG neurons are a diverse population including non-

nociceptors responding to non-damaging, low-intensity

stimuli (touch and proprioception) and nociceptors reacting

to high-intensity, potentially damaging stimuli (pain sen-

sation) (Lawson 2002). Two distinct differentiation path-

ways lead to the formation of peptidergic nociceptors

expressing calcitonin gene-related peptide (CGRP) and

substance P, and non-peptidergic nociceptors binding iso-

lectin B4 (IB4) (Bennett et al. 1996; Molliver et al. 1997;

Woolf and Ma 2007). The most extensively studied trans-

ducer of noxious stimuli is TRPV1, a non-selective cation

channel highly permeable to Ca2? and sensitive to tem-

peratures above 43 �C, acidification and capsaicin (Tomi-

naga et al. 1998; Caterina et al. 1997; Brenneis et al. 2013;

Wetsel 2011).

The present study was focused on the first level of the

pain pathway—DRG and sensory axons. Based on histo-

chemical classification we characterized the a7 nAChR-

expressing subpopulation of DRG neurons, specifically,

their cell size distribution and sensory modalities. We also

examined their ability to transport a7 nAChR axonally to

the spinal cord and periphery. In addition, the receptor

activity as a ligand-gated calcium channel and its role in

neurotransmitter CGRP and glutamate release were studied.

Materials and methods

Animals

Animal care and animal experiments were performed fol-

lowing the current version of the German Law on the

Protection of Animals as well as the NIH ‘‘principles of

laboratory animal care’’. Adult (3–5 months old) Wistar

rats of either sex were used. They were kept under a 12-h

light–dark cycle with free access to chow and water. Rats

were deeply anesthetized by inhalation of isoflurane

(Baxter Deutschland GmbH, Unterschleissheim, Ger-

many), DRGs and spinal cord from segmental levels

T1–T12 and L1–L5 were excised immediately or after

transcardial perfusion with 250 ml fixative solution [4 %

paraformaldehyde in 0.1 M phosphate buffer (PB, pH

7.4)]. For histochemistry the tissues were shock frozen in

melting isopentane or paraformaldehyde-fixed by immer-

sion in the fixative solution for 2 h at room temperature,

repeated washing with phosphate buffered saline (PBS) for

6–8 h and overnight incubation in 18 % w/v sucrose in PB

at 4 �C. Freshly dissected tissues from animals not per-

fused with fixative solution were used in functional studies.

Histochemistry

The histochemical procedure was based on the protocol

published earlier (Shelukhina et al. 2009) with some

modifications. Tissues were sectioned at a thickness of

10 lm with a cryostat. Sections from shock-frozen tissue

were fixed with isopropanol for 10 min at 4 �C or, alter-

natively, with 4 % paraformaldehyde for 10 min at 21 �C,

rinsed with PBS and distilled water and air dried for 1 h.

Sections from paraformaldehyde-fixed tissue were dried

without any pretreatment. Unspecific protein binding sites

were saturated for 1 h with PBS containing 100 ml/l nor-

mal horse serum, 10 g/l BSA and 5 ml/l Tween 20 and

were further pre-incubated for 1–2 h in buffer A (1 g/l

BSA and 150 mmol/l NaCl in PBS). Blocking controls

were run simultaneously by adding 100-fold excess of

unlabeled a-cobratoxin (CTX) or a-neurotoxin II (NTII) to

buffer A (Table 1). Then, a mixture of primary antibodies

and/or Alexa Fluor 488-conjugated aBgt (Alexa-aBgt;

Invitrogen, Molecular Probes, OR, USA) was added to the

slides to reach final concentrations as specified in Table 1.

Biotinylated IB4 (1:100, Vector Laboratories, USA) mixed

with Alexa-aBgt was applied in the same way. After

overnight incubation the sections were washed with PBS

and processed with appropriate combinations of secondary

reagents (Table 1). Cell nuclei were visualized by incu-

bation with 1 lg/ml 40,6-diamidino-20-phenylindole-dihy-

drochloride (DAPI; Sigma-Aldrich, Germany) for 20 min.

Further, the sections were washed with PBS, fixed with

4 % paraformaldehyde for 10 min, rinsed with PBS again

and coverslipped in carbonate-buffered glycerol at pH 8.6.

The slides were evaluated by epifluorescence microscopy

(Axioplan2, Zeiss, Germany) using appropriate filter com-

binations. Preabsorption of rabbit anti-CGRP antibodies

(Table 1) with the corresponding peptide (Acris Antibodies

GmbH, Germany) (1 lg of peptide in 50 ll of antibody final

dilution) or omission of any antibody (Table 1) abolished the

immunoreactivity. Substitution of goat anti-CGRP antibodies

for the rabbit anti-CGRP-antibodies gave the same typical

Brain Struct Funct

123

Page 3: Functional expression and axonal transport of α7 nAChRs by peptidergic nociceptors of rat dorsal root ganglion

punctate staining of the perinuclear region (Zhang et al. 1994;

Guo et al. 1999) and the same percentage of co-localization

with Alexa-aBgt.

Analysis of cell size distribution and percentage of his-

tochemically specified neuron populations was done on

neuronal section profiles containing a nucleus visualized by

DAPI labeling. Mean cell diameter was measured as the

mean of the shortest and longest axes of the neuronal section

profile. Data obtained from lumbar and thoracic DRGs were

pooled since there were no significant differences in their

size distributions (Kolmogorov–Smirnov test, p [ 0.2).

Nerve crush experiment

DRG together with ventral, dorsal roots and spinal nerve

and a piece of sciatic nerve were dissected. Both the roots

and the nerves were crushed with forceps. The preparations

were incubated for 12 h at 37 �C in Hepes-Ringer buffer

(5.6 mM KCl, 136.4 mM NaCl, 1 mM MgCl2, 2.2 mM

CaCl2, 11 mM glucose, 10 mM HEPES, pH 7.4) bubbled

with carbogen (95 % O2/5 % CO2). Then the preparations

were shock frozen and sectioned longitudinally for histo-

chemical procedures.

Primary cell culture

The culturing procedure was performed as described earlier

(Nassenstein et al. 2010; Gold 2012) with some

modifications. Isolated DRGs were incubated in an enzyme

mixture [2 mg/ml collagenase type 1 and 2 mg/ml dispase

II (Sigma-Aldrich, Germany)] in calcium-, magnesium-free

Hanks’ balanced salt solution (Invitrogen, USA) for

15 min at 37 �C and neurons were dissociated by tritur-

ation with a glass Pasteur pipette. This step was repeated

three times, and then cells were washed by centrifugation

(three times at 1,000g for 2 min) and suspended in L15

medium (Invitrogen, USA) containing 10 % fetal bovine

serum (FBS). The cell suspension was transferred onto

poly-D-lysine/laminin (Sigma-Aldrich, Germany)-coated

coverslips or tissue culture 24-well plates (cells obtained

from two ganglia/well). After the suspended neurons had

adhered for 2 h, the neuron-attached coverslips or plates

were flooded with the L-15 medium (10 % FBS) and used

after 20–24 h in culture.

Calcium imaging

The intracellular calcium concentration ([Ca2?]i) mea-

surements were performed on primary cultures of DRG

neurons. Measurements were done in oxygenated Locke’s

buffer (pH 7.4) containing (in mM): NaHCO3 14.3,

NaH2PO4 1.2, KCl 5.6, NaCl 136, MgCl2 1.2, CaCl2 2.2, D-

glucose 10 at constant temperature 34–36 �C. The cells

were loaded for 30 min with 4 lM Fura-2 AM (Invitrogen,

USA) in L-15 medium containing 20 % FBS, placed in a

recording chamber (2 ml bath volume) and washed for

Table 1 Toxins, primary antibodies and secondary reagents used in this work

Toxin Conjugate Host Concentration Source

aBgt Alexa Fluor 488 Bungarus multicinctus 12.5 nM Invitrogen, USA

CTX Naja kaouthia 1.25 lM Own (Kukhtina et al. 2000)

NTII Naja oxiana 1.25 lM Own (Utkin et al. 2001)

Primary antibody, antigen Host Dilution Source

CGRP Goat 1:4,000 Biotrend, Germany

CGRP Rabbit 1:3,200 Peninsula Laboratories, USA

MBP Sheep 1:2,000 Chemicon, USA

NF200 Rabbit 1:600 Sigma-Aldrich, Germany

NF-L Mouse 1:600 Invitrogen, USA

PGP 9.5 Rabbit 1:5,000 Biotrend, Germany

TH Rabbit 1:400 Biotrend, Germany

TRPV1 Rabbit 1:2,000 Chemicon, USA

Reagent Conjugate Host Dilution Source

Anti-goat IgG Texas Red Donkey 1:400 Dianova, Germany

Anti-rabbit IgG Biotin Donkey 1:200 Amersham, UK

Anti-sheep IgG Biotin Donkey 1:400 Amersham, UK

Anti-mouse IgG Biotin Goat 1:200 Jackson Immunoresearch, USA

Streptavidin AMCA 1:200 Dianova, Germany

Brain Struct Funct

123

Page 4: Functional expression and axonal transport of α7 nAChRs by peptidergic nociceptors of rat dorsal root ganglion

20 min by infusion of Locke’s buffer. Fura-2 was excited

at 340 and 380 nm wavelengths (k), and fluorescence was

collected at k[ 420 nm. Each cell was tracked indepen-

dently, and the fluorescence intensity ratio of 340/380 nm

at the beginning of the experiment was set to 100 % and

the relative changes were recorded. In each experiment, the

cells were exposed to nicotinic agonist epibatidine

(10-5 M) or PNU282987 (10-6 M) followed by capsaicin

(0.5 lM) and KCl (50 mM) applications. In some experi-

ments, cells were pre-incubated with nicotinic positive

allosteric modulator PNU120596 (10-6 M) or antagonists

CTX (10-6–10-7 M, Table 1) and mecamylamine

(10-4 M) before agonist application. All ligands except

CTX (Table 1) were purchased from Sigma-Aldrich,

Germany.

Neurotransmitter release

CGRP

For CGRP release studies, primary DRG neuronal culture

and whole ganglia were used. Whole DRGs were placed

into plate wells and repeatedly incubated at 35–37 �C with

the oxygenated Locke’s buffer, pH 7.4, for 15 min. Cell

cultures underwent the same procedures after aspiration of

growth medium and washing with oxygenated Locke’s

buffer, but incubation intervals were decreased to 10 min.

In order to exclude CGRP released due to plating and

initial handling of the preparations, we discarded the first

four incubation solutions and collected the fifth for ana-

lyzing basal CGRP release. Then the specimens were

exposed to successive incubation with the buffer alone or

buffer containing nicotinic drugs. Similarly, next incuba-

tion with 500 nM capsaicin was performed to evoke CGRP

release in the presence or absence of nicotinic drugs. The

supernatants were collected at 10 min intervals for neuro-

nal cultures and at 15 min intervals for ganglia, and ana-

lyzed for CGRP content by radioimmunoassay (RIA,

Phoenix Pharmaceuticals, USA) and by enzyme immuno-

assay [CGRP (rat) EIA kit, Bertin Pharma, France]

according to the manufacturer’s protocol. At the end of the

experiment, DRG cell cultures were exposed to 2 N acetic

acid for 10 min to determine the remaining intracellular

peptide content. Aliquots of this incubation were diluted

ten times prior to RIA of CGRP content.

The reliability of EIA was checked using CGRP

knockout and wild-type mouse DRGs. Extracts from DRGs

were made according to the protocol given by Bracci-

Laudieroa et al. (2002). Briefly, ten DRGs were placed in

0.25 ml of 1 M acetic acid and boiled for 5 min. Subse-

quently, the DRGs were disrupted in a ball mill. After

centrifugation (20 min, 960g, 4 �C) the supernatants were

collected, lyophilized and stored at -80 �C. Immediately

before starting the CGRP EIA, the pellets were solubilised

in 100 ll of EIA buffer. Samples were analyzed at a 1:100

dilution.

Glutamate

Freshly dissected spinal cords (T1–T12, L1–L5) were

sectioned at a thickness of 300 lm with a vibratome, then

slices were manually divided into dorsal and ventral parts.

Dorsal parts of spinal cord slices were placed into plate

wells and underwent the same protocol as for CGRP

release from whole ganglia with minor modifications.

Instead of capsaicin, 50 mM KCl solution was utilized to

evoke neurotransmitter release, and after stimulation the

slices were exposed for 15 min to oxygenated Locke’s

buffer alone to re-establish basal release. Glutamate con-

tent in supernatants was measured by high-performance

liquid chromatography (HPLC) of its phthaldialdehyde

(OPA, Sigma-Aldrich, Germany) derivative. The chro-

matograph system included binary HPLC pump Waters

1525 and Waters 2487 absorbance detector (Waters Corp.,

USA). The analytical column used was a reverse-phase

Phenomenex Nucleosil C18 (250 9 4.6 mm, 5 lm particle

size). Chromatographic separation was carried out at room

temperature in linear gradient of methanol (0–40 % for

15 min, 40–95 % for 1 min, 95–0 % for 1 min) in

12.5 mM phosphate buffer, pH 6.8, at a flow rate of 0.7 ml/

min and the detection was performed at 340 nm. The level

of glutamate was calculated by comparing the peak areas

with those of standards [glutamic acid (Sigma-Aldrich,

Germany), 125 pmol–2 nmol]. Using this method, the

minimal detectable amount of glutamate was 125 pmol.

Statistical analysis

Statistical analyses of normally distributed data were made

by one-way ANOVA for multiple comparisons or by Stu-

dent’s t test, when only two sets of data were compared.

For analysis of other data, Kruskal–Wallis one-way

ANOVA on ranks with Dunn’s post test or Kolmogorov–

Smirnov test was performed. p \ 0.05 was considered

significant.

Results

Specificity of labeling of DRG neuronal a7 nAChR-

expressing subpopulation

At the first step we showed that could reliably visualize a7

nAChR expressed by rat lumbar and thoracic DRG neurons

using a histochemical protocol with Alexa-aBgt, validated

on a7 nAChR knockout mouse tissues (Shelukhina et al.

Brain Struct Funct

123

Page 5: Functional expression and axonal transport of α7 nAChRs by peptidergic nociceptors of rat dorsal root ganglion

2009). Positive neurons exhibited labeling of their cell

membrane as well as granular perinuclear labeling (Fig. 1a,

b) similar to that obtained for mouse DRGs (Shelukhina

et al. 2009). Double labeling with Alexa-aBgt and an

antibody against a neuronal cytoplasmic protein [protein

gene product 9.5 (PGP 9.5) (Thompson et al. 1983)]

showed that all a7 nAChR-positive cells were neurons (112

cells, n = 4) (Fig. 1c) and did not reveal any a7 nAChR-

positive glial satellite cells. The specificity of a7 nAChR

histochemical labeling was confirmed by competition of

Alexa-aBgt with a-CTX (Fig. 1f), capable of blocking

both muscle-type and a7 nAChRs, and by a lack of com-

petition for a-NTII specific for muscle-type nAChR

(Fig. 1e).

Morphological and neurochemical classification of a7

nAChR-expressing DRG neurons

DRG neurons are grossly divided into two major classes:

(1) Neurons with small to medium, neurofilament-poor

somata and thin non-myelinated C-fibers and (2) neurons

with medium to large, neurofilament-rich somata and thinly

(Ad) or thickly (Aa/b) myelinated A-fibers, respectively

(Lawson 2002). DRG neuronal soma size and fiber type are

known to correlate with its sensory modality. Generally,

the number of nociceptors decreases from small to large

DRG neurons (C [ Ad[ Aa/b), and the proportion of

non-nociceptors increases in parallel (C \ Ad\ Aa/b)

(Djouhri and Lawson 2004). On this basis, we investigated

cell size and neurofilament expression of a7 nAChR-

expressing neurons.

Alexa-aBgt-binding neurons formed an individual sub-

population with a significant *5 lm shift of their soma

diameter median to larger values relative to the total pop-

ulation of DRG neurons (Fig. 2a, b). Medium-sized a7

nAChR-expressing neurons prevailed, although the range

covered all cell sizes except the smallest group (15–20 lm)

(Fig. 2b). Alexa-aBgt-binding cells accounted for one-fifth

of all medium-sized and large DRG neurons, and\5 % of

small somata (Fig. 2c). Double labeling showed that 46 %

of a7 nAChR-expressing cells were neurofilament-immu-

nopositive (287/624 cells, n = 6, Fig. 3A(a), B). Conse-

quently, one half of the a7 nAChR-expressing population

consisted of small- to medium-sized, neurofilament-poor

DRG neurons with presumably thin, non-myelinated

C-fibers, and the other of medium-sized to large neurofil-

ament-rich cells with thick, myelinated Ad- and Aa/b-

axons.

To obtain more clues as to their potential involvement in

nociception, we applied two neurochemical markers widely

used to define two major classes of nociceptors: peptidergic

(CGRP-immunopositive) and non-peptidergic (IB4-binding)

Fig. 1 Detection of a7 nAChRs in rat DRG cryostat slices with

Alexa-aBgt (green). Alexa-aBgt-binding DRG neurons exhibit (a, b,

d) granular perinuclear labeling (arrowhead) and labeling of the cell

membrane (arrow), and (c) nerve fiber labeling. b Simultaneous

staining with antibody to cytoplasm marker PGP 9.5 (red) revealed

neuronal section profiles. c Neurofilament- (red) and a7 nAChR-

containing (green) nerve fibers localize next to each other, but are

separate. The specificity of a7 nAChR detection was confirmed by

(f) inhibition of Alexa-aBgt binding with a 100-fold excess of a-

CTX, capable of blocking both muscle-type and a7 nAChRs, and by

(e) a lack of inhibition with a-NTII specific for muscle-type nAChR.

Bars 40 lm

Brain Struct Funct

123

Page 6: Functional expression and axonal transport of α7 nAChRs by peptidergic nociceptors of rat dorsal root ganglion

cells (Lawson et al. 2002; Woolf and Ma 2007; Fang et al.

2006; Djouhri and Lawson 2004). In accordance with

previous studies (Lawson et al. 2002) where CGRP was

shown to be expressed in small to large nociceptive neu-

rons with C-, Ad- and Aa/b-fibers, in our material CGRP-

positive cells were 12.5–57 lm in diameter (Fig. 4). IB4-

binding neurons were smaller (12.5–42 lm, Fig. 4)

including small C-fiber nociceptors exclusively (Fang et al.

2006). Double-labeling experiments demonstrated that the

vast majority of a7 nAChR-expressing DRG neurons were

peptidergic, whereas only a small fraction was non-pepti-

dergic nociceptors, as 83 % of Alexa-aBgt-labeled neurons

co-expressed CGRP [459/552 cells, n = 6, Fig. 3A(b), B)]

and only 15 % (127/838 cells, n = 6, Fig. 3A(c), B) bound

IB4.

Next, we investigated co-expression of TRPV1, a

transducer of some noxious stimuli (Tominaga et al. 1998;

Caterina et al. 1997; Wetsel 2011; Brenneis et al. 2013),

with a7 nAChR in DRG. This receptor is expressed in C-

and Ad-fiber nociceptors (Mitchell et al. 2010; Brenneis

et al. 2013; Kobayashi et al. 2005): in 49.2–59 % of small

to medium peptidergic and in 50–67 % of small non-pep-

tidergic neurons of adult rat DRGs (Guo et al. 1999; Price

and Flores 2007). In our conditions, 64.8 % (362/559 cells,

15–42 lm in diameter, n = 4) of peptidergic and 61.2 %

(336/549 cells, 14–40 lm, n = 4) of non-peptidergic DRG

neurons contained TRPV1 (Fig. 4). 28 % of Alexa-aBgt-

binding cells (347/1242 cells, n = 10) were TRPV1-im-

munolabeled [Fig. 3A(d), B]. Triple staining showed that,

among a7 nAChR-expressing neurons, almost all (92 %,

105/114 cells, n = 5) non-peptidergic (binding IB4) and a

third (29.7 %, 77/259 cells, n = 4) of peptidergic (CGRP-

positive) neurons were TRPV1 positive (Fig. 3C).

In sum, the presented histochemical data showed that a7

nAChR-expressing DRG neurons were primarily small to

large peptidergic nociceptive neurons, one half of which

had C-fibers and the other had A-fibers, which most likely

included both Ad- and Aa/b-fiber types. A small part of

Alexa-aBgt-binding cells was non-peptidergic IB4-binding

C-fiber nociceptors. Almost all non-peptidergic and a third

of peptidergic a7 nAChR-containing neurons expressed a

receptor responsive to noxious stimuli, i.e., TRPV1.

Axonal localization and transport of a7 nAChR

Alexa-aBgt-labeled varicose nerve fibers were observed in

DRG sections without additional intervention to block

axonal transport (Fig. 1c). There are two possible origins

for these axons: (1) DRG neurons and (2) postganglionic

noradrenergic sympathetic neurons which are known to

innervate DRGs (Kummer 1994) and to express a7 nAC-

hRs (Cuevas et al. 2000; Si and Lee 2003). Since these

A

B

C

Fig. 2 Size distribution of neurons in rat DRGs. The plots represent

the size distribution of (a) total (402 cells, n = 5) and b Alexa-aBgt-

labeled neurons (309 cells, n = 7) present in the ganglia; c relative

frequencies of labeled neurons within each size class. Size distribu-

tions of total and aBgt-binding populations were significantly

different according to Kolmogorov–Smirnov test, p \ 0.05. The

relative frequency (rf) was calculated with a formula rfi = (li/

ti) 9 8.5 %, where rfi is the relative frequency of the size group i, li is

the frequency of the size group i within labeled neurons, ti is the

frequency of the size group i within total neurons and 8.5 % is the

average percentage of labeled among total cells in DRGs (218/2553

cells, n = 5)

Brain Struct Funct

123

Page 7: Functional expression and axonal transport of α7 nAChRs by peptidergic nociceptors of rat dorsal root ganglion

Alexa-aBgt-positive fibers were not immunoreactive (data

not shown) to tyrosine hydroxylase (TH) and the rate-

limiting enzyme of noradrenaline synthesis, and since

nerve fibers containing TH were thinner and generally

localized in the DRG capsule (Kummer 1994), these in-

traganglionic a7 nAChR-positive axons are unlikely of

sympathetic origin. They were immunonegative to both

heavy (NF200, Fig. 1c) and light (NF-L, data not shown)

components of neurofilament as well, leading us to assume

that most of them were non-myelinated, sensory C-fibers.

To increase the local level of intra-axonal a7 nAChR

and to demonstrate its directed transport in DRG neurons, a

complex nerve crush experiment was performed. It inclu-

ded dorsal root and both sciatic and spinal nerve crushing

to test transport in central and peripheral branches of axons

of DRG neurons, respectively. Since the sciatic and spinal

nerves do not consist of afferent fibers exclusively and also

contain efferent fibers, we performed crushing of ventral

root to monitor axonal transport in efferent motoneurons

separately.

A notable anterograde accumulation of Alexa-aBgt-

binding receptors was revealed at the peripheral (i.e.,

ganglionic) side of a dorsal root crush and at the central

(i.e., ganglionic) side of spinal and sciatic nerve crushes,

where a weak retrograde accumulation was demonstrated

as well (Fig. 5a–c, f). However, we did not find any aBgt-

binding sites in crushed ventral roots (Fig. 5d), in contrast

to clearly observed proximal accumulation of CGRP

expressed by efferent motoneurons (Gibson et al. 1984)

(Fig. 5e). This indicated that the axonal anterograde

transport in motoneurons functioned normally (Kashihara

et al. 1989), but did not contribute to a7 nAChR accu-

mulation observed in sciatic and spinal nerves. In addition

to extensive co-localisation of a7 nAChRs and CGRP in

Fig. 3 Double (a, b) and triple (c) labeling of rat DRG cryostat

sections with Alexa-aBgt (Green) and antibodies (red) to NF200 (a),

CGRP (b) and TRPV1 (d) as well as with biotinylated IB4 [(c), red].

Histograms (B, C) demonstrate percentage of double- and triple-

labeled cells among a7 nAChR-expressing (Alexa-aBgt-binding)

neurons. At least 4 (up to 10) independent experiments were carried

out for each pair (three) of substances. Bars 40 lm

Fig. 4 Comparison of mean diameters of CGRP-, IB4-, TRPV1- or

double-positive rat DRG neurons with Alexa-aBgt-binding ones.

Neuron diameter profiles were compared by Kruskal–Wallis one-way

ANOVA on ranks, Dunn’s post test, p \ 0.05, 309–726 cells per

condition, n = 4–7

Brain Struct Funct

123

Page 8: Functional expression and axonal transport of α7 nAChRs by peptidergic nociceptors of rat dorsal root ganglion

cell bodies of DRG neurons (Fig. 3) we showed their joint

anterograde transport by some afferent axons in dorsal root

and in spinal and sciatic nerves (Fig. 5f). We did not reveal

if these axons were myelinated because immunoreactivity

to histochemical markers NF200 and myelin basic protein

(MBP) significantly decreased near the crush point. It was

earlier demonstrated that afferent neurons react to axotomy

by down-regulating neurofilament expression (Fornaro

et al. 2008). Collectively, the obtained results demonstrated

a strong anterograde and weak retrograde transport of a7

nAChRs in both central and peripheral processes of DRG

neurons, particularly, CGRPergic.

Calcium responses to a7 nAChR activation

Calcium entry through a7 nAChR activates a variety of

biological processes in neurons including, but not restricted

to, neurotransmitter release, cell signaling, and gene

expression (Shen and Yakel 2009; Uteshev 2012; Dajas-

Bailador and Wonnacott 2004). The scheme of our

experiment included from one to three applications of

nicotinic drugs followed by capsaicin administration to

distinguish C- and Ad-fiber nociceptive neurons (Brenneis

et al. 2013; Mitchell et al. 2010; Ringkamp et al. 2001) and

ended by addition of potassium chloride at high concen-

tration to confirm the excitability of the investigated cell.

To activate exclusively a7 nAChRs, their highly potent

selective agonist PNU282987 (10-6 M) (Bodnar et al.

2005) was used. It induced a small and fast-decaying cal-

cium rise in 5.8 % of cultured DRG neurons (31/537 cells,

n = 13) (Fig. 6). This response was amplified by pre-

treatment with the a7 nAChR-specific positive allosteric

modulator PNU120596 (10-6 M, Fig. 6), which cannot

initiate nicotinic channel opening, but decreases its

desensitization rate and increases agonist-evoked calcium

flux (Hurst et al. 2005). Under these conditions, 12 % of

Fig. 5 Accumulation a7

nAChR (Alexa-aBgt binding;

green) after blockade of axonal

transport by nerve crush.

a Dorsal root, b, f spinal nerve,

c sciatic nerve and d, e ventral

root. The sites of crushes are

indicated with arrows;

P peripheral, C central side of

the crush. f CGRP (red) is co-

transported with a7 nAChRs

(green) centrally (ganglionic

side) to a crush of a spinal nerve

(yellow, arrowheads), but in

ventral root (e) CGRP (red)

transport was revealed in the

absence of (d) a7 nAChR

(green) accumulation. Bars

40 lm

Brain Struct Funct

123

Page 9: Functional expression and axonal transport of α7 nAChRs by peptidergic nociceptors of rat dorsal root ganglion

DRG neurons (30/249 cells, n = 6) showed an a7 nAChR-

mediated calcium response, with most of them were being

capsaicin-sensitive C- and Ad-fiber nociceptors (23/30

cells, n = 6), Fig. 6a).

Further, we used non-selective nicotinic agonist epi-

batidine to produce calcium responses through both ho-

mopentameric a7 nAChR and heteropentameric nAChR

subtypes and to compare their form, amplitude and number

with those evoked by the a7 nAChR-specific agonist.

Epibatidine (10-5 M) induced a rapid calcium rise in 33 %

(280/851 cells, n = 18) of DRG neurons, half of which

were nociceptive capsaicin-sensitive (55 %, 154/280 cells,

n = 18) (Fig. 6). This response was due to activation of

nAChRs as it was abolished by preincubation with the

neuronal nicotinic receptor antagonist mecamylamine

(10-4 M) (Fig. 6a, 0/204 cells, n = 4). In the presence of

a7-specific antagonist CTX (10-6 M) epibatidine provoked

[Ca2?]i increase in 30.6 % cells (139/454 cells, n = 9)

demonstrating that a7 nAChR could be rarely found in

isolation and is mainly co-expressed with other nAChR

subtypes in DRG neurons (Fig. 6a). The amplitude and

character of the epibatidine-evoked responses varied sig-

nificantly indicating involvement of different nAChR

types, along with amplification of calcium signal through a

number of mechanisms (Dajas-Bailador and Wonnacott

2004; Ween et al. 2010) (Fig. 6b–d). PNU282987 induced

a small and fast-decaying calcium response in comparison

with most epibatidine-evoked ones (Fig. 6b–d). It can be

explained by the typical fast and complete desensitization

of a7 nAChR rather than by different potency of two

agonists applied, since the pretreatment with the positive

allosteric modulator increased the amplitude of a7 nAChR-

mediated responses considerably, but they remained fast-

decaying. All these data show that along with other nAChR

subtypes which are not in the focus of this work, adult rat

DRG neurons express typical calcium-permeable func-

tional a7 nAChR.

Neurotransmitter release

CGRP

Before neurotransmitter release studies, we confirmed the

specificity of a commercially available enzyme immuno-

assay. We could detect 101.3 ± 0.5 pg of CGRP/DRG in

Fig. 6 Intracellular calcium response of rat DRG neurons to nicotinic

ligands: agonists epibatidine (EPI, 10-5 M) and PNU282987 (PNU,

10-6 M), antagonists a-CTX (10-6 M) and mecamylamine (MEC,

10-4 M), positive allosteric modulator PNU120596 (PAM, 10-6 M),

and to capsaicin (CPS, 0.5 lM). a Percentage of reactive neurons,

b calcium response curves, c amplitude and d extent of calcium rise

Brain Struct Funct

123

Page 10: Functional expression and axonal transport of α7 nAChRs by peptidergic nociceptors of rat dorsal root ganglion

wild-type mouse, while measured concentration of CGRP

in knockout mouse tissue was at background level.

The study of putative a7 nAChR role in neurotrans-

mitter CGRP and glutamate release from DRG neurons

included three test systems: primary DRG neuronal cell

culture, whole ganglia and dorsal part of spinal cord slices

where these neurons terminate. Basal CGRP release from

DRG cell culture within 10 min was 69 ± 9.4 pg/ml

(Fig. 7a, n = 5). Stimulation by capsaicin at 0.5 lM

concentration caused a sevenfold increase in CGRP release

(490 ± 10.9 pg/ml) (Fig. 7a). When expressed as the per-

cent of intracellular CGRP total content from the beginning

to the end of the experiment, these values correspond to

3.6 % in the basal condition and 25.2 % in the capsaicin-

stimulated condition. Addition of non-specific nicotinic

agonist epibatidine (10-5 M) and a7-selective agonist PNU

282987 (10-6 M) or competitive antagonist CTX (10-6 M)

affected neither basal nor capsaicin-provoked CGRP

release (Fig. 7a). The experiment was repeated with some

modifications on freshly isolated DRGs to exclude any

influence of neuronal cell culture preparation procedures,

e.g. enzymatic treatment and mechanical trituration. After

dissection DRGs were incubated with oxygenated Locke’s

buffer, pH 7.4, for 1 h. Basal CGRP release was then

97 ± 11 pg/ml for 15 min (Fig. 7b, n = 4). Application of

capsaicin (0.5 lM) for the same period elevated CGRP

release to 316 ± 23 pg/ml. Nicotinic agonists such as

nicotine (10-4 M), epibatidine (10-5 M) and PNU282987

(10-6 M) did not provoke CGRP release from DRGs and

did not modulate capsaicin-stimulated release. The positive

allosteric modulator PNU120596 (10-6 M) did not mani-

fest a nicotinic agonist action in this test.

Glutamate

As a7 nAChR is known to modulate glutamatergic trans-

mission in different parts of CNS (McGehee et al. 1995;

Gotti et al. 2006; Gray et al. 1996; Jiang and Role 2008;

Wonnacott et al. 2006; Genzen and McGehee 2003), we

checked if nicotinic agonists act on basal or evoked release

of glutamate from DRG neuron terminals, and if an excess

of CGRP could modulate this process. Application of

50 mM KCl onto dorsal part of spinal cord slices led to a

2.1-fold increase of glutamate release as compared to the

basal level (Fig. 8, n = 4). Within the next 15 min wash-

out period we observed a significant decline in glutamate

release. However, we did not reveal statistically significant

changes of basal or KCl-provoked glutamate release under

application of nicotinic agonists epibatidine (10-5 M) and

PNU282987 (10-6 M). In addition we tested the reaction to

these drugs in the presence of a7 nAChR-specific positive

allosteric modulator PNU120596 (10-6 M) and competi-

tive antagonist CTX (10-6 M) as well as under excess of

CGRP (10-6 M). Neither positive nor negative modulation

of glutamate release was observed (Fig. 8).

Discussion

Here we showed that functionally active a7 nAChRs are

expressed and axonally transported by a subpopulation of

DRG neurons, mostly composed of C-, Ad- and Aa/b-fiber

* * * * *

A

B

Fig. 7 CGRP release from (a) rat DRG neuronal cell culture and

(b) whole ganglia. a Cells were exposed to four consecutive

incubation periods: (1) with buffer (basal release), (2) with nicotinic

ligands [agonists epibatidine (10-5 M) and PNU282987 (10-6 M)

and antagonist CTX (10-6 M)], (3) with capsaicin (0.5 lM) in the

presence or absence of nicotinic drugs and (4) with 2 N acetic acid to

measure the remaining intracellular CGRP content (asterisk—histo-

gram represents 10 % of the remaining CGRP content), n = 5

(mean ± SD). b Whole ganglia underwent the same procedure

without exposure to acid and with additional nicotinic ligands

[agonist nicotine (10-4 M) and positive allosteric modulator (PAM)

PNU120596 (10-6 M)], n = 4 (mean ± SD). Paired t test revealed a

significant (p \ 0.05) difference between basal and capsaicin-stimu-

lated releases in all groups, whereas nicotinic ligands modulate

neither basal nor capsaicin-stimulated CGRP release significantly

(one-way ANOVA)

Brain Struct Funct

123

Page 11: Functional expression and axonal transport of α7 nAChRs by peptidergic nociceptors of rat dorsal root ganglion

peptidergic (CGRPergic) nociceptors. A part of these

neurons responded to noxious stimulus, i.e., capsaicin, and

expressed its receptor TRPV1. a7 nAChRs on DRG neu-

rons are calcium-permeable and can be activated and

positively allosterically modulated by their selective

ligands. Whereas a7 nAChRs localize almost exclusively

in CGRPergic cells, nicotinic ligands do not affect the

neuropeptide CGRP release from DRG neurons in vitro.

All our findings deal with neurons, since these are single

type of cells which, as we found, express a7 nAChRs in

DRGs, although there is an increasing number of studies

showing expression and functional role of these receptors

in CNS astrocytes (Shen and Yakel 2012; Liu et al. 2012)

and microglia (Shytle et al. 2004; Parada et al. 2013;

Thomsen and Mikkelsen 2012). Here in DRG, we dem-

onstrated plasmatic membrane localization of neuronal a7

nAChRs, in addition to their intracellular pool, that could

include both endoplasmatic and mitochondrial receptors

(Gergalova et al. 2012; Drisdel et al. 2004).

a7 nAChR-expressing cells compose an individual

population comprising one-fifth of total medium and large

DRG neurons and\5 % of small ones. This non-ubiquitous

localization might suggest a special functional role for a7

receptors. By applying a combination of well-known neu-

rochemical markers (Lawson et al. 2002; Woolf and Ma

2007; Fang et al. 2006; Djouhri and Lawson 2004), we

found that great majority of a7 nAChR-expressing DRG

neurons were nociceptive: 83.2 % peptidergic (CGRPergic)

and 15.2 % non-peptidergic. Based on their cell size

distribution and neurofilament content, we suggested that

a7 nAChR-bearing peptidergic nociceptors might include,

in addition to common small to medium rather slowly

conducting C- and Ad-fiber cells, rare medium to large

thickly myelinated fast-conducting Aa/b-fiber type (Law-

son 2002; Lawson et al. 2002).

a7 nAChRs localized on DRG neuronal cell body might

play a functional role, but in CNS they are preferentially

situated at preterminal or presynaptic sites (Gotti et al.

2009; Berg and Conroy 2002; Marchi and Grilli 2010).

This requires axonal transport from the site of protein

synthesis, i.e., the soma, towards terminals. Our crush

experiments on explanted ganglia with dorsal and ventral

roots and nerves indeed revealed an anterograde and a

weak retrograde transport of a7 nAChRs in the central and

peripheral branches of DRG neuron axons. It is consistent

with previous reports on axonal transport of aBgt-binding

sites in rat (Millington et al. 1985; Ninkovic and Hunt

1983) and chicken (Roth et al. 2000) sciatic nerve. Since in

our experiments a significant number of a7 nAChR-con-

taining axons were CGRP-immunoreactive, and a7 nAChR

transport was missing from ventral horn motoneurons, we

conclude that the majority of a7 nAChR transporting

neurons belong to the class of peptidergic nociceptors.

Being localized on nociceptive neurons, a7 nAChRs

along with other receptors, such as transient receptor poten-

tial (TRP) channels, acid-sensing sodium ion channels

(ASIC1-3), the ATP-gated P2X3 receptor, etc. (Woolf and

Ma 2007; Julius and Basbaum 2001), might make up an

Fig. 8 Glutamate release from dorsal part of rat spinal cord slices

(data normalized to basal release, mean ± SD, n = 4). Slices were

exposed to four consecutive incubation periods: (1) with buffer (basal

release), (2) with nicotinic agonists epibatidine (10-5 M) or

PNU282987 (10-6 M) and their mixture with a7 nAChR-specific

positive allosteric modulator (PAM) PNU120596 (10-6 M) or

antagonist CTX (10-6 M) or CGRP (10-6 M), (3) with 50 mM KCl

in the presence or absence of nicotinic drugs and (4) with buffer to re-

establish basal release. Paired t test revealed a significant (p \ 0.05)

difference between basal and KCl-stimulated glutamate releases in all

groups, whereas nicotinic ligands modulate neither basal nor capsa-

icin-stimulated CGRP release significantly (one-way ANOVA)

Brain Struct Funct

123

Page 12: Functional expression and axonal transport of α7 nAChRs by peptidergic nociceptors of rat dorsal root ganglion

ensemble capable of detecting a broad spectrum of external

and internal stimuli. This idea is supported by findings that

non-selective nicotinic agonists excite DRG neurons in vitro

and in vivo (Steen and Reeh 1993; Bernardini et al. 2001;

Carr and Proske 1996; Rau et al. 2005; Genzen et al. 2001).

We revealed that specific agonist of a7 nAChR (PNU

282987) indeed evoked a small and fast-decaying calcium

rise in DRG neurons, which could be enhanced by a7

nAChR-selective positive allosteric modulator PNU120596.

Most of activated cells were capsaicin-sensitive C- and Ad-

fiber nociceptors and their number corresponded to per-

centage of a7 nAChR-expressing DRG neurons in histo-

chemical experiments, i.e., 12 versus 8.5 %, respectively. In

comparison with PNU 282987, a non-selective nicotinic

agonist epibatidine provoked a stronger [Ca2?]i increase in

greater number of cells (33 %), only a few of which could be

blocked by a7 nAChR-specific antagonist CTX. This finding

indicates the presence of other nAChR subtypes on the same

DRG neuron. Assortment of nAChRs varied, since the

amplitude and form of epibatidine-evoked calcium rises were

different, in accord with previous reports on non-uniform

electrophysiological responses of DRG neurons to nicotinic

drugs (Genzen et al. 2001; Rau et al. 2005).

High calcium permeability of a7 nAChR is the cause of

its efficacy as a modulator of neurotransmitter release in

CNS (Gray et al. 1996; Jiang and Role 2008; Gotti et al.

2006; Wonnacott et al. 2006; McGehee et al. 1995). It

might play the same role on DRG neurons, since cholin-

ergic interneurons presynaptically contact sensory termi-

nals in the spinal cord (Ribeiro-da-Silva and Cuello 1990)

and activation of nAChRs enhances both excitatory (Gen-

zen and McGehee 2003) and inhibitory (Liu et al. 2011)

synaptic transmission in dorsal horn. The release of glu-

tamate, the main excitatory neurotransmitter of both noci-

ceptive and non-nociceptive DRG neurons (Woolf and Ma

2007), was also analyzed in the present work. We did not

observe a significant change of basal or evoked glutamate

release from dorsal part of adult rat spinal cord in response

to nicotinic agonists. CGRP excess also did not influence

this process, although its inhibitory effect on nAChR is

well known (Di Angelantonio et al. 2003). The discrepancy

between our finding and the previously demonstrated

increase in excitatory transmission by a7 nAChR in neo-

natal rat dorsal horn (Genzen and McGehee 2003) might be

explained by significantly higher level of its expression

during earlier stages of ontogenesis, as documented for

numerous organs (Gotti and Clementi 2004; Putz et al.

2008; Zoli et al. 1995).

Peptidergic nociceptors, the main class of a7 nAChR-

expressing neurons, use neuropeptide CGRP as both a pain

signal neurotransmitter releasing in the spinal cord (Woolf

and Ma 2007) and as a pro-inflammatory agent on the

periphery (Richardson and Vasko 2002). Since a7 nAChR

is involved in release of every brain neurotransmitter (Gotti

et al. 2006), as well as in peripheral anti-inflammatory

reactions (Marrero et al. 2011; Olofsson et al. 2012; Huston

2012), and because nicotine could stimulate a CGRP

release from different organs (Franco-Cereceda et al. 1989;

Hua et al. 1994; Lou et al. 1992), we supposed that a7

receptor might modulate this process. However, in our

study nicotinic ligands including a7 nAChR-specific one

influenced neither basal nor capsaicin-evoked CGRP

release from DRG neurons. Although earlier nicotine-pro-

voked CGRP release from various tissues was explained by

its direct action through DRG neurons (Franco-Cereceda

et al. 1992), later publications have shown that nicotine

requires sympathetic efferent terminals activation to

mediate its effect upon primary afferent CGRP release

(Hua et al. 1995; Kawasaki et al. 2011). Our observations

also support the hypothesis about indirect stimulation of

CGRP release from sensory neurons by nicotinic drugs.

Elucidation of a7 nAChR functional role is in the focus

of intensive studies and appears to involve a cross talk with

other receptors (Marchi and Grilli 2010). In DRGs, we

demonstrated co-localization of a7 nAChR and TRPV1,

the capsaicin-sensitive transducer of heat, chemical (H?)

and mechanical noxious stimuli (Brenneis et al. 2013;

Wetsel 2011). All C-fiber non-peptidergic and a third of C-

and Ad-fiber peptidergic a7 nAChR-bearing nociceptors

expressed TRPV1, as follows from histochemical labeling.

In primary DRG cell culture, most neurons activated by a7-

specific agonist were capsaicin-sensitive, probably, due to

higher portion of small size cells surviving after culturing.

It is likely that a7 nAChRs co-expressed with TRPV1 in

DRG neurons might modulate its functioning, particularly,

contribute to nicotine modification of capsaicin-induced

TRPV1-mediated currents into those with faster desensiti-

zation and smaller amplitude (Fucile et al. 2005), although

application of nicotinic ligands did not alter capsaicin-

evoked CGRP release according to our study.

In summary, functional a7 nAChRs are expressed and

axonally transported to the spinal cord and periphery by

nociceptive DRG neurons, mainly by medium-sized

CGRPergic. These receptors do not modulate neurotrans-

mitter CGRP and glutamate release directly since nicotinic

ligands affected neither their basal nor provoked levels,

showing the necessity of further studies of the a7 nAChR

function in DRG neurons.

Acknowledgments This work was supported by the LOEWE Pro-

gram of the State of Hessen, Research Focus Non-neuronal cholinergic

system, by grants of Russian Foundation for Basic Research No. 13-04-

40377-N KOMFI and MCB RAS program. We thank Martin Boden-

benner, Tamara Papadakis, Silke Wiegand and Anna Goldenberg for

technical help, Dr. Christina Nassenstein for helpful discussion and

Prof. E. Weihe (Philipps-University Marburg) for providing CGRP

knockout mice. The authors declare no conflict of interests.

Brain Struct Funct

123

Page 13: Functional expression and axonal transport of α7 nAChRs by peptidergic nociceptors of rat dorsal root ganglion

References

Abdin MJ, Morioka N, Morita K, Kitayama T, Kitayama S,

Nakashima T, Dohi T (2006) Analgesic action of nicotine on

tibial nerve transection (TNT)-induced mechanical allodynia

through enhancement of the glycinergic inhibitory system in

spinal cord. Life Sci 80(1):9–16. doi:10.1016/j.lfs.2006.08.011

Bennett DL, Averill S, Clary DO, Priestley JV, McMahon SB (1996)

Postnatal changes in the expression of the trkA high-affinity

NGF receptor in primary sensory neurons. Eur J Neurosci

8(10):2204–2208

Berg DK, Conroy WG (2002) Nicotinic alpha 7 receptors: synaptic

options and downstream signaling in neurons. J Neurobiol

53(4):512–523. doi:10.1002/neu.10116

Bernardini N, Sauer SK, Haberberger R, Fischer MJ, Reeh PW (2001)

Excitatory nicotinic and desensitizing muscarinic (M2) effects

on C-nociceptors in isolated rat skin. J Neurosci

21(9):3295–3302 (pii: 21/9/3295)

Bodnar AL, Cortes-Burgos LA, Cook KK, Dinh DM, Groppi VE,

Hajos M, Higdon NR, Hoffmann WE, Hurst RS, Myers JK,

Rogers BN, Wall TM, Wolfe ML, Wong E (2005) Discovery and

structure-activity relationship of quinuclidine benzamides as

agonists of alpha7 nicotinic acetylcholine receptors. J Med Chem

48(4):905–908. doi:10.1021/jm049363q

Boyd RT, Jacob MH, McEachern AE, Caron S, Berg DK (1991)

Nicotinic acetylcholine receptor mRNA in dorsal root ganglion

neurons. J Neurobiol 22(1):1–14. doi:10.1002/neu.480220102

Bracci-Laudiero L, Aloe L, Buanne P, Finn A, Stenfors C, Vigneti E,

Theodorsson E, Lundeberg T (2002) NGF modulates CGRP

synthesis in human B-lymphocytes: a possible anti-inflammatory

action of NGF? J Neuroimmunol 123(1–2):58–65 (pii:

S0165572801004751)

Brenneis C, Kistner K, Puopolo M, Segal D, Roberson D, Sisignano

M, Labocha S, Ferreiros N, Strominger A, Cobos EJ, Ghasemlou

N, Geisslinger G, Reeh PW, Bean BP, Woolf CJ (2013)

Phenotyping the function of TRPV1-expressing sensory neurons

by targeted axonal silencing. J Neurosci 33(1):315–326. doi:10.

1523/JNEUROSCI.2804-12.2013

Carr RW, Proske U (1996) Action of cholinesters on sensory nerve

endings in skin and muscle. Clin Exp Pharmacol Physiol

23(5):355–362

Caterina MJ, Schumacher MA, Tominaga M, Rosen TA, Levine JD,

Julius D (1997) The capsaicin receptor: a heat-activated ion

channel in the pain pathway. Nature 389(6653):816–824. doi:10.

1038/39807

Cuevas J, Roth AL, Berg DK (2000) Two distinct classes of functional

7-containing nicotinic receptor on rat superior cervical ganglion

neurons. J Physiol 525(Pt 3):735–746 (pii: PHY_0374)

Dajas-Bailador F, Wonnacott S (2004) Nicotinic acetylcholine

receptors and the regulation of neuronal signalling. Trends

Pharmacol Sci 25(6):317–324. doi:10.1016/j.tips.2004.04.006

Damaj MI, Meyer EM, Martin BR (2000) The antinociceptive effects

of alpha7 nicotinic agonists in an acute pain model. Neurophar-

macology 39(13):2785–2791 (pii: S0028390800001398)

Di Angelantonio S, Giniatullin R, Costa V, Sokolova E, Nistri A

(2003) Modulation of neuronal nicotinic receptor function by the

neuropeptides CGRP and substance P on autonomic nerve cells.

Br J Pharmacol 139(6):1061–1073. doi:10.1038/sj.bjp.0705337

Djouhri L, Lawson SN (2004) Abeta-fiber nociceptive primary

afferent neurons: a review of incidence and properties in relation

to other afferent A-fiber neurons in mammals. Brain Res Brain

Res Rev 46(2):131–145. doi:10.1016/j.brainresrev.2004.07.015

Drisdel RC, Manzana E, Green WN (2004) The role of palmitoylation

in functional expression of nicotinic alpha7 receptors. J Neurosci

24(46):10502–10510. doi:10.1523/JNEUROSCI.3315-04.2004

Fang X, Djouhri L, McMullan S, Berry C, Waxman SG, Okuse K,

Lawson SN (2006) Intense isolectin-B4 binding in rat dorsal root

ganglion neurons distinguishes C-fiber nociceptors with broad

action potentials and high Nav1.9 expression. J Neurosci

26(27):7281–7292. doi:10.1523/JNEUROSCI.1072-06.2006

Feuerbach D, Lingenhoehl K, Olpe HR, Vassout A, Gentsch C,

Chaperon F, Nozulak J, Enz A, Bilbe G, McAllister K, Hoyer D

(2009) The selective nicotinic acetylcholine receptor alpha7

agonist JN403 is active in animal models of cognition, sensory

gating, epilepsy and pain. Neuropharmacology 56(1):254–263.

doi:10.1016/j.neuropharm.2008.08.025

Fornaro M, Lee JM, Raimondo S, Nicolino S, Geuna S, Giacobini-

Robecchi M (2008) Neuronal intermediate filament expression in

rat dorsal root ganglia sensory neurons: an in vivo and in vitro

study. Neuroscience 153(4):1153–1163. doi:10.1016/j.neu

roscience.2008.02.080

Franco-Cereceda A, Saria A, Lundberg JM (1989) Differential release

of calcitonin gene-related peptide and neuropeptide Y from the

isolated heart by capsaicin, ischaemia, nicotine, bradykinin and

ouabain. Acta Physiol Scand 135(2):173–187

Franco-Cereceda A, Rydh M, Dalsgaard CJ (1992) Nicotine- and

capsaicin-, but not potassium-evoked CGP-release from cultured

guinea-pig spinal ganglia is inhibited by Ruthenium red.

Neurosci Lett 137(1):72–74

Fucile S, Sucapane A, Eusebi F (2005) Ca2? permeability of nicotinic

acetylcholine receptors from rat dorsal root ganglion neurones.

J Physiol 565(Pt 1):219–228. doi:10.1113/jphysiol.2005.084871

Genzen JR, McGehee DS (2003) Short- and long-term enhancement

of excitatory transmission in the spinal cord dorsal horn by

nicotinic acetylcholine receptors. Proc Natl Acad Sci USA

100(11):6807–6812. doi:10.1073/pnas.1131709100

Genzen JR, Van Cleve W, McGehee DS (2001) Dorsal root ganglion

neurons express multiple nicotinic acetylcholine receptor sub-

types. J Neurophysiol 86(4):1773–1782

Gergalova G, Lykhmus O, Kalashnyk O, Koval L, Chernyshov V,

Kryukova E, Tsetlin V, Komisarenko S, Skok M (2012)

Mitochondria express alpha7 nicotinic acetylcholine receptors

to regulate Ca2? accumulation and cytochrome c release: study

on isolated mitochondria. PLoS ONE 7(2):e31361. doi:10.1371/

journal.pone.0031361

Gibson SJ, Polak JM, Bloom SR, Sabate IM, Mulderry PM, Ghatei

MA, McGregor GP, Morrison JF, Kelly JS, Evans RM et al

(1984) Calcitonin gene-related peptide immunoreactivity in the

spinal cord of man and of eight other species. J Neurosci

4(12):3101–3111

Gold MS (2012) Whole-cell recording in isolated primary sensory

neurons. Methods Mol Biol 851:73–97. doi:10.1007/978-1-

61779-561-9_5

Gotti C, Clementi F (2004) Neuronal nicotinic receptors: from

structure to pathology. Prog Neurobiol 74(6):363–396. doi:10.

1016/j.pneurobio.2004.09.006

Gotti C, Zoli M, Clementi F (2006) Brain nicotinic acetylcholine

receptors: native subtypes and their relevance. Trends Pharmacol

Sci 27(9):482–491. doi:10.1016/j.tips.2006.07.004

Gotti C, Clementi F, Fornari A, Gaimarri A, Guiducci S, Manfredi I,

Moretti M, Pedrazzi P, Pucci L, Zoli M (2009) Structural and

functional diversity of native brain neuronal nicotinic receptors.

Biochem Pharmacol 78(7):703–711. doi:10.1016/j.bcp.2009.05.024

Gray R, Rajan AS, Radcliffe KA, Yakehiro M, Dani JA (1996)

Hippocampal synaptic transmission enhanced by low concentra-

tions of nicotine. Nature 383(6602):713–716. doi:10.1038/

383713a0

Gundisch D, Eibl C (2011) Nicotinic acetylcholine receptor ligands, a

patent review (2006–2011). Expert Opin Ther Pat

21(12):1867–1896. doi:10.1517/13543776.2011.637919

Brain Struct Funct

123

Page 14: Functional expression and axonal transport of α7 nAChRs by peptidergic nociceptors of rat dorsal root ganglion

Guo A, Vulchanova L, Wang J, Li X, Elde R (1999) Immunocyto-

chemical localization of the vanilloid receptor 1 (VR1):

relationship to neuropeptides, the P2X3 purinoceptor and IB4

binding sites. Eur J Neurosci 11(3):946–958

Gurun MS, Parker R, Eisenach JC, Vincler M (2009) The effect of

peripherally administered CDP-choline in an acute inflammatory

pain model: the role of alpha7 nicotinic acetylcholine receptor.

Anesth Analg 108(5):1680–1687. doi:10.1213/ane.

0b013e31819dcd08

Haberberger RV, Bernardini N, Kress M, Hartmann P, Lips KS,

Kummer W (2004) Nicotinic acetylcholine receptor subtypes in

nociceptive dorsal root ganglion neurons of the adult rat. Auton

Neurosci 113(1–2):32–42. doi:10.1016/j.autneu.2004.05.008

Hua XY, Jinno S, Back SM, Tam EK, Yaksh TL (1994) Multiple

mechanisms for the effects of capsaicin, bradykinin and nicotine

on CGRP release from tracheal afferent nerves: role of

prostaglandins, sympathetic nerves and mast cells. Neurophar-

macology 33(10):1147–1154

Hua XY, Wong S, Jinno S, Yaksh TL (1995) Pharmacology of

calcitonin gene related peptide release from sensory terminals in

the rat trachea. Can J Physiol Pharmacol 73(7):999–1006

Hurst RS, Hajos M, Raggenbass M, Wall TM, Higdon NR, Lawson

JA, Rutherford-Root KL, Berkenpas MB, Hoffmann WE,

Piotrowski DW, Groppi VE, Allaman G, Ogier R, Bertrand S,

Bertrand D, Arneric SP (2005) A novel positive allosteric

modulator of the alpha7 neuronal nicotinic acetylcholine recep-

tor: in vitro and in vivo characterization. J Neurosci

25(17):4396–4405. doi:10.1523/JNEUROSCI.5269-04.2005

Huston JM (2012) The vagus nerve and the inflammatory reflex:

wandering on a new treatment paradigm for systemic inflam-

mation and sepsis. Surg Infect (Larchmt) 13(4):187–193. doi:10.

1089/sur.2012.126

Jiang L, Role LW (2008) Facilitation of cortico-amygdala synapses

by nicotine: activity-dependent modulation of glutamatergic

transmission. J Neurophysiol 99(4):1988–1999. doi:10.1152/jn.

00933.2007

Julius D, Basbaum AI (2001) Molecular mechanisms of nociception.

Nature 413(6852):203–210. doi:10.1038/35093019

Kashihara Y, Sakaguchi M, Kuno M (1989) Axonal transport and

distribution of endogenous calcitonin gene-related peptide in rat

peripheral nerve. J Neurosci 9(11):3796–3802

Kawasaki H, Takatori S, Zamami Y, Koyama T, Goda M, Hirai K,

Tangsucharit P, Jin X, Hobara N, Kitamura Y (2011) Paracrine

control of mesenteric perivascular axo-axonal interaction. Acta

Physiol (Oxf) 203(1):3–11. doi:10.1111/j.1748-1716.2010.

02197.x

Kobayashi K, Fukuoka T, Obata K, Yamanaka H, Dai Y, Tokunaga

A, Noguchi K (2005) Distinct expression of TRPM8, TRPA1,

and TRPV1 mRNAs in rat primary afferent neurons with adelta/

c-fibers and colocalization with trk receptors. J Comp Neurol

493(4):596–606. doi:10.1002/cne.20794

Kukhtina VV, Weise K, Osipov AV, Starkov VG, Titov MI, Esipov

SE, Ovchinnikova TV, Tsetlin VI, Utkin IuN (2000) MALDI-

mass spectrometry for identification of new proteins in snake

venoms. Bioorg Khim 26(11):803–807

Kummer W (1994) Sensory ganglia as a target of autonomic and

sensory nerve fibres in the guinea-pig. Neuroscience 59(3):

739–754

Lawson SN (2002) Phenotype and function of somatic primary

afferent nociceptive neurones with C-Adelta- or Aalpha/beta-

fibres. Exp Physiol 87(2):239–244 (pii: EPH_2350)

Lawson SN, Crepps B, Perl ER (2002) Calcitonin gene-related

peptide immunoreactivity and afferent receptive properties of

dorsal root ganglion neurones in guinea-pigs. J Physiol 540(Pt

3):989–1002 (pii: PHY_13086)

Liu T, Fujita T, Kumamoto E (2011) Acetylcholine and norepineph-

rine mediate GABAergic but not glycinergic transmission

enhancement by melittin in adult rat substantia gelatinosa

neurons. J Neurophysiol 106(1):233–246. doi:10.1152/jn.

00838.2010

Liu Y, Hu J, Wu J, Zhu C, Hui Y, Han Y, Huang Z, Ellsworth K, Fan

W (2012) alpha7 nicotinic acetylcholine receptor-mediated

neuroprotection against dopaminergic neuron loss in an MPTP

mouse model via inhibition of astrocyte activation. J Neuroin-

flamm 9:98. doi:10.1186/1742-2094-9-98

Lou YP, Franco-Cereceda A, Lundberg JM (1992) Different ion

channel mechanisms between low concentrations of capsaicin

and high concentrations of capsaicin and nicotine regarding

peptide release from pulmonary afferents. Acta Physiol Scand

146(1):119–127

Marchi M, Grilli M (2010) Presynaptic nicotinic receptors modulating

neurotransmitter release in the central nervous system: func-

tional interactions with other coexisting receptors. Prog Neuro-

biol 92(2):105–111. doi:10.1016/j.pneurobio.2010.06.004

Marrero MB, Bencherif M, Lippiello PM, Lucas R (2011) Applica-

tion of alpha7 nicotinic acetylcholine receptor agonists in

inflammatory diseases: an overview. Pharm Res

28(2):413–416. doi:10.1007/s11095-010-0283-7

McGehee DS, Heath MJ, Gelber S, Devay P, Role LW (1995)

Nicotine enhancement of fast excitatory synaptic transmission in

CNS by presynaptic receptors. Science 269(5231):1692–1696

Medhurst SJ, Hatcher JP, Hille CJ, Bingham S, Clayton NM,

Billinton A, Chessell IP (2008) Activation of the alpha7-

nicotinic acetylcholine receptor reverses complete freund adju-

vant-induced mechanical hyperalgesia in the rat via a central site

of action. J Pain 9(7):580–587. doi:10.1016/j.jpain.2008.01.336

Millington WR, Aizenman E, Bierkamper GG, Zarbin MA, Kuhar MJ

(1985) Axonal transport of alpha-bungarotoxin binding sites in

rat sciatic nerve. Brain Res 340(2):269–276 (pii:

0006-8993(85)90923-0)

Mitchell K, Bates BD, Keller JM, Lopez M, Scholl L, Navarro J,

Madian N, Haspel G, Nemenov MI, Iadarola MJ (2010) Ablation

of rat TRPV1-expressing Adelta/C-fibers with resiniferatoxin:

analysis of withdrawal behaviors, recovery of function and

molecular correlates. Mol Pain 6:94. doi:10.1186/1744-8069-6-94

Molliver DC, Wright DE, Leitner ML, Parsadanian AS, Doster K,

Wen D, Yan Q, Snider WD (1997) IB4-binding DRG neurons

switch from NGF to GDNF dependence in early postnatal life.

Neuron 19(4):849–861 (pii: S0896-6273(00)80966-6)

Murray TA, Bertrand D, Papke RL, George AA, Pantoja R,

Srinivasan R, Liu Q, Wu J, Whiteaker P, Lester HA, Lukas RJ

(2012) alpha7beta2 nicotinic acetylcholine receptors assemble,

function, and are activated primarily via their alpha7-alpha7

interfaces. Mol Pharmacol 81(2):175–188. doi:10.1124/mol.111.

074088

Nassenstein C, Taylor-Clark TE, Myers AC, Ru F, Nandigama R,

Bettner W, Undem BJ (2010) Phenotypic distinctions between

neural crest and placodal derived vagal C-fibres in mouse lungs.

J Physiol 588(Pt 23):4769–4783. doi:10.1113/jphysiol.2010.

195339

Ninkovic M, Hunt SP (1983) Alpha-bungarotoxin binding sites on

sensory neurones and their axonal transport in sensory afferents.

Brain Res 272(1):57–69 (pii: 0006-8993(83)90364-5)

Nizri E, Brenner T (2011) Modulation of inflammatory pathways by

the immune cholinergic system. Amino Acids. doi:10.1007/

s00726-011-1192-8

Olofsson PS, Rosas-Ballina M, Levine YA, Tracey KJ (2012)

Rethinking inflammation: neural circuits in the regulation of

immunity. Immunol Rev 248(1):188–204. doi:10.1111/j.1600-

065X.2012.01138.x

Brain Struct Funct

123

Page 15: Functional expression and axonal transport of α7 nAChRs by peptidergic nociceptors of rat dorsal root ganglion

Pacini A, Di Cesare Mannelli L, Bonaccini L, Ronzoni S, Bartolini A,

Ghelardini C (2010) Protective effect of alpha7 nAChR:

behavioural and morphological features on neuropathy. Pain

150(3):542–549. doi:10.1016/j.pain.2010.06.014

Parada E, Egea J, Buendia I, Negredo P, Cunha AC, Cardoso S,

Soares MP, Lopez MG (2013) The microglial alpha7 acetylcho-

line nicotinic receptor is a key element in promoting neuropro-

tection by inducing HO-1 via Nrf2. Antioxid Redox Signal.

doi:10.1089/ars.2012.4671

Price TJ, Flores CM (2007) Critical evaluation of the colocalization

between calcitonin gene-related peptide, substance P, transient

receptor potential vanilloid subfamily type 1 immunoreactivities,

and isolectin B4 binding in primary afferent neurons of the rat

and mouse. J Pain 8(3):263–272. doi:10.1016/j.jpain.2006.09.

005

Putz G, Kristufek D, Orr-Urtreger A, Changeux JP, Huck S, Scholze P

(2008) Nicotinic acetylcholine receptor-subunit mRNAs in the

mouse superior cervical ganglion are regulated by development

but not by deletion of distinct subunit genes. J Neurosci Res

86(5):972–981. doi:10.1002/jnr.21559

Rau KK, Johnson RD, Cooper BY (2005) Nicotinic AChR in

subclassified capsaicin-sensitive and -insensitive nociceptors of

the rat DRG. J Neurophysiol 93(3):1358–1371. doi:10.1152/jn.

00591.2004

Ribeiro-da-Silva A, Cuello AC (1990) Choline acetyltransferase-

immunoreactive profiles are presynaptic to primary sensory

fibers in the rat superficial dorsal horn. J Comp Neurol

295(3):370–384. doi:10.1002/cne.902950303

Richardson JD, Vasko MR (2002) Cellular mechanisms of neurogenic

inflammation. J Pharmacol Exp Ther 302(3):839–845. doi:10.

1124/jpet.102.032797

Ringkamp M, Peng YB, Wu G, Hartke TV, Campbell JN, Meyer RA

(2001) Capsaicin responses in heat-sensitive and heat-insensitive

A-fiber nociceptors. J Neurosci 21(12):4460–4468

Roth AL, Shoop RD, Berg DK (2000) Targeting alpha7-containing

nicotinic receptors on neurons to distal locations. Eur J

Pharmacol 393(1–3):105–112 (pii: S0014-2999(00)00092-3)

Rowley TJ, McKinstry A, Greenidge E, Smith W, Flood P (2010)

Antinociceptive and anti-inflammatory effects of choline in a

mouse model of postoperative pain. Br J Anaesth

105(2):201–207. doi:10.1093/bja/aeq113

Shelukhina IV, Kryukova EV, Lips KS, Tsetlin VI, Kummer W

(2009) Presence of alpha7 nicotinic acetylcholine receptors on

dorsal root ganglion neurons proved using knockout mice and

selective alpha-neurotoxins in histochemistry. J Neurochem

109(4):1087–1095. doi:10.1111/j.1471-4159.2009.06033.x

Shen JX, Yakel JL (2009) Nicotinic acetylcholine receptor-mediated

calcium signaling in the nervous system. Acta Pharmacol Sin

30(6):673–680. doi:10.1038/aps.2009.64

Shen JX, Yakel JL (2012) Functional alpha7 nicotinic ACh receptors

on astrocytes in rat hippocampal CA1 slices. J Mol Neurosci

48(1):14–21. doi:10.1007/s12031-012-9719-3

Shytle RD, Mori T, Townsend K, Vendrame M, Sun N, Zeng J,

Ehrhart J, Silver AA, Sanberg PR, Tan J (2004) Cholinergic

modulation of microglial activation by alpha 7 nicotinic

receptors. J Neurochem 89(2):337–343. doi:10.1046/j.1471-

4159.2004.02347.x

Si ML, Lee TJ (2003) Pb2? inhibition of sympathetic alpha

7-nicotinic acetylcholine receptor-mediated nitrergic neurogenic

dilation in porcine basilar arteries. J Pharmacol Exp Ther

305(3):1124–1131. doi:10.1124/jpet.102.046854

Steen KH, Reeh PW (1993) Actions of cholinergic agonists and

antagonists on sensory nerve endings in rat skin, in vitro.

J Neurophysiol 70(1):397–405

Sucher NJ, Cheng TP, Lipton SA (1990) Neural nicotinic acetylcho-

line responses in sensory neurons from postnatal rat. Brain Res

533(2):248–254 (pii: 0006-8993(90)91346-I)

Thompson RJ, Doran JF, Jackson P, Dhillon AP, Rode J (1983) PGP

9.5—a new marker for vertebrate neurons and neuroendocrine

cells. Brain Res 278(1–2):224–228 (pii: 0006-8993(83)90241-X)

Thomsen MS, Mikkelsen JD (2012) The alpha7 nicotinic acetylcho-

line receptor ligands methyllycaconitine, NS6740 and GTS-21

reduce lipopolysaccharide-induced TNF-alpha release from

microglia. J Neuroimmunol 251(1–2):65–72. doi:10.1016/j.

jneuroim.2012.07.006

Thomsen MS, Hansen HH, Timmerman DB, Mikkelsen JD (2010)

Cognitive improvement by activation of alpha7 nicotinic

acetylcholine receptors: from animal models to human patho-

physiology. Curr Pharm Des 16(3):323–343

Tominaga M, Caterina MJ, Malmberg AB, Rosen TA, Gilbert H,

Skinner K, Raumann BE, Basbaum AI, Julius D (1998) The

cloned capsaicin receptor integrates multiple pain-producing

stimuli. Neuron 21(3):531–543 (pii: S0896-6273(00)80564-4)

Tsetlin V, Utkin Y, Kasheverov I (2009) Polypeptide and peptide

toxins, magnifying lenses for binding sites in nicotinic acetyl-

choline receptors. Biochem Pharmacol 78(7):720–731. doi:10.

1016/j.bcp.2009.05.032

Uteshev VV (2012) alpha7 nicotinic ACh receptors as a ligand-gated

source of Ca(2?) ions: the search for a Ca(2?) optimum. Adv

Exp Med Biol 740:603–638. doi:10.1007/978-94-007-2888-2_27

Utkin YN, Kukhtina VV, Kryukova EV, Chiodini F, Bertrand D,

Methfessel C, Tsetlin VI (2001) ‘‘Weak toxin’’ from Naja

kaouthia is a nontoxic antagonist of alpha 7 and muscle-type

nicotinic acetylcholine receptors. J Biol Chem

276(19):15810–15815. doi:10.1074/jbc.M100788200

Wang H, Yu M, Ochani M, Amella CA, Tanovic M, Susarla S, Li JH,

Yang H, Ulloa L, Al-Abed Y, Czura CJ, Tracey KJ (2003) Nicotinic

acetylcholine receptor alpha7 subunit is an essential regulator of

inflammation. Nature 421(6921):384–388. doi:10.1038/nature01339

Wang Y, Su DM, Wang RH, Liu Y, Wang H (2005) Antinociceptive

effects of choline against acute and inflammatory pain. Neuro-

science 132(1):49–56. doi:10.1016/j.neuroscience.2004.12.026

Ween H, Thorin-Hagene K, Andersen E, Gronlien JH, Lee CH,

Gopalakrishnan M, Malysz J (2010) Alpha3* and alpha 7

nAChR-mediated Ca2? transient generation in IMR-32 neuro-

blastoma cells. Neurochem Int 57(3):269–277. doi:10.1016/j.

neuint.2010.06.005

Wetsel WC (2011) Sensing hot and cold with TRP channels. Int J

Hyperth 27(4):388–398. doi:10.3109/02656736.2011.554337

Wonnacott S, Barik J, Dickinson J, Jones IW (2006) Nicotinic

receptors modulate transmitter cross talk in the CNS: nicotinic

modulation of transmitters. J Mol Neurosci 30(1–2):137–140.

doi:10.1385/JMN:30:1:137

Woolf CJ, Ma Q (2007) Nociceptors—noxious stimulus detectors.

Neuron 55(3):353–364. doi:10.1016/j.neuron.2007.07.016

Wu J, Lukas RJ (2011) Naturally-expressed nicotinic acetylcholine

receptor subtypes. Biochem Pharmacol 82(8):800–807. doi:10.

1016/j.bcp.2011.07.067

Zhang X, Bao L, Xu ZQ, Kopp J, Arvidsson U, Elde R, Hokfelt T

(1994) Localization of neuropeptide Y Y1 receptors in the rat

nervous system with special reference to somatic receptors on

small dorsal root ganglion neurons. Proc Natl Acad Sci USA

91(24):11738–11742

Zoli M, Le Novere N, Hill JA Jr, Changeux JP (1995) Developmental

regulation of nicotinic ACh receptor subunit mRNAs in the rat central

and peripheral nervous systems. J Neurosci 15(3 Pt 1):1912–1939

Brain Struct Funct

123