You are on page 1of 11

See

discussions, stats, and author profiles for this publication at: https://www.researchgate.net/publication/320013159

Molecular basis of dental sensitivity: The


odontoblasts are multisensory cells and
express multifunctional...

Article in Annals of Anatomy - Anatomischer Anzeiger · September 2017


DOI: 10.1016/j.aanat.2017.09.006

CITATIONS READS

0 76

6 authors, including:

Luis Junquera José A Vega


University of Oviedo University of Oviedo
195 PUBLICATIONS 1,734 CITATIONS 376 PUBLICATIONS 5,159 CITATIONS

SEE PROFILE SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Osteonecrosis of the Jaw View project

Los implantes MG-Osseous: Estudio multicéntrico retrospectivo View project

All content following this page was uploaded by José A Vega on 06 October 2017.

The user has requested enhancement of the downloaded file.


Annals of Anatomy 215 (2018) 20–29

Contents lists available at ScienceDirect

Annals of Anatomy
journal homepage: www.elsevier.com/locate/aanat

Molecular basis of dental sensitivity: The odontoblasts are


multisensory cells and express multifunctional ion channels夽
A. Solé-Magdalena a,1 , M. Martínez-Alonso a,1 , C.A. Coronado b , L.M. Junquera c,d , J. Cobo c,e ,
J.A. Vega a,b,∗
a
Departamento de Morfología y Biología Celular Universidad de Oviedo, Spain
b
Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Temuco, Chile
c
Departamento de Especialidades Médico-Quirúrgicas, Universidad de Oviedo, Spain
d
Servicio de Cirugía Maxilofacial, Hospital Universitario Central de Asturias, Oviedo, Spain
e
Instituto Asturiano de Odontología, Oviedo, Spain

a r t i c l e i n f o a b s t r a c t

Article history: Odontoblasts are the dental pulp cells responsible for the formation of dentin. In addition, accumulating
Received 17 April 2017 data strongly suggest that they can also function as sensory cells that mediate the early steps of mechan-
Received in revised form 22 August 2017 ical, thermic, and chemical dental sensitivity. This assumption is based on the expression of different
Accepted 10 September 2017
families of ion channels involved in various modalities of sensitivity and the release of putative neuro-
transmitters in response to odontoblast stimulation which are able to act on pulp sensory nerve fibers.
Keywords:
This review updates the current knowledge on the expression of transient-potential receptor ion chan-
Odontoblasts
nels and acid-sensing ion channels in odontoblasts, nerve fibers innervating them and trigeminal sensory
Dentin sensitivity
Transient-receptor potential ion channels
neurons, as well as in pulp cells. Moreover, the innervation of the odontoblasts and the interrelationship
Acid-sensing ion channels been odontoblasts and nerve fibers mediated by neurotransmitters was also revisited. These data might
ATP provide the basis for novel therapeutic approaches for the treatment of dentin sensibility and/or dental
ATP receptors pain.
© 2017 Published by Elsevier GmbH.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 21
2. Innervation of the odontoblasts . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 21
3. Dentin sensitivity and ion channels . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 22
4. The superfamily of transient receptor potential ion channels (TRP) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 22
5. The superfamily of acid-sensing ion channels . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 22
6. Putative mechanosensors in odontoblasts and trigeminal afferent fibers and neurons . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 24
7. Putative thermosensors in odontoblasts and trigeminal afferent fibers/neurons . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 24
8. Putative chemosensors in odontoblasts and trigeminal afferent fibers and neurons . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 24
9. Other mechano- and thermos-sensitive proteins in odontoblasts . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 24
10. The primary cilium of the odontoblasts and its relation to TRP and ASIC ion channels . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 25
11. Intercellular signal transmission between odontoblasts and nerves . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 25
12. Future perspectives: TRPs, ASICs, and the ATP signaling system as targets for the treatment of dentin sensitivity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 26
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 26

夽 This paper belongs to the special issue Dentomaxillary.


∗ Corresponding author at: Departamento de Morfología y Biología Celular, Facultad de Medicina y Ciencias de la Salud, C/Julián Clavería, 6, 33006 Oviedo, Spain.
E-mail addresses: javega@uniovi.es, javega@hotmail.es (J.A. Vega).
1
These authors contributed equally to this paper.

https://doi.org/10.1016/j.aanat.2017.09.006
0940-9602/© 2017 Published by Elsevier GmbH.
A. Solé-Magdalena et al. / Annals of Anatomy 215 (2018) 20–29 21

1. Introduction presence of ion channels, primarily belonging to the TRP and ASICs
superfamilies, in these cells. The blockade of these channels would
Odontoblasts are specialized cells of the dental pulp originating provide a novel therapeutic intervention for the treatment of dentin
from the neural crest (Mayor and Theveneau, 2013) which migrate sensitivity and tooth pain (see El Karim et al., 2011; Alexander et al.,
beneath the oral epithelium and finally differentiate into cells that 2013; Kweon and Suh, 2013; Holzer and Izzo, 2014; Kaneko and
will organize and regulate the synthesis of the mineralized dentin Szallasi, 2014; Nilius and Szallasi, 2014; Sousa-Valente et al., 2014;
matrix (Arana-Chavez and Massa, 2004; Bleicher, 2014; Kawashima Baron and Lingueglia, 2015).
and Okiji, 2016). During maturation, odontoblasts exhibit change in
gene expression, acquiring a typical morphology with a body and a
2. Innervation of the odontoblasts
long process (Simon et al., 2009; Byers and Westenbroek, 2011). The
cell bodies of the odontoblasts are located at the dentin-pulp inter-
Nerves entering the dental pulp consist of sensory and post-
face complex where they are organized in a palisade between the
ganglionic sympathetic fibres arising from the trigeminal and the
mineralized tissues (enamel and dentin) and the living tissue (the
superior sympathetic ganglia neurons, respectively. Within the
pulp) of the tooth. In fact, odontoblasts are connected at their api-
dental pulp they innervate the blood vessels, the pulp cells, and the
cal pole (i.e. the zones connecting the bodies with the processes) by
odontoblasts through fibers evolved from the so-called subodon-
numerous junctional complexes (tight junctions and desmosome-
toblastic plexus (Raschkow’s plexus; Fig. 2). Odontoblasts, both at
like) forming a selective barrier controlling the relationship and
the dentin-pulp border and within the dentinal tubules, are inner-
trafficking between dentin and pulp and vice versa under physio-
vated by a dense network of myelinated A␦ and unmyelinated C
logical and pathological conditions. The odontoblast processes are
sensory nerve fibers. However, the definitive structure of the con-
contained in dentinal tubules bathing in the dentinal fluid (Fig. 1;
tacts between the odontoblasts and the subodontoblastic plexus
see Murray et al., 2003; Bleicher, 2014).
remains to be elucidated (see Allard et al., 2006). The A␦ fibers are
In addition to the formation of the dentin, the odontoblasts pre-
principally located at the pulp-dentin border and reach the basal
sumably mediate the early stage of sensory processes, playing a key
odontoblast layer, while the C fibers enter the dentin tubules (Byers
role in mechanical, thermal, and chemical sensation, thus, in dental
and Närhi, 1999; Struys et al., 2007). Byers (1984) observed that
sensitivity and pain (Gillam, 1995; Maurin et al., 2003). Because of
more than 40% of the dentinal tubules contain nerve fibres and
their location in the tooth, odontoblasts are the first targets of exter-
many of them more than one. An elegant and detailed study car-
nal stimuli (chemicals changes in dentinal fluid, thermal variations
ried out by Cardá and Peydró (2006) in human teeth demonstrated
or mechanical forces), and can therefore act as sensory cells for the
that about 30–70% of odontoblast processes are in contact with
detection of these sensory signals. Dentin sensitivity is due to the
nerve endings that enter the innermost segments of the dentinal
direct exposure of dentin to mechanical, chemical and/or thermal
tubules, no further than 200 ␮m from the pulp. They also observed
stimuli. In this way, the odontoblasts express mechanosensitive,
that each dentinal tubule contains a single nerve fiber which varies
chemosensitive and thermosensitive transient receptor potential
in relationship to the odontoblast process from simple adjacency
(TRP; Magloire et al., 2010) and acid-sensing (ASIC; Solé-Magdalena
to encircling. Nevertheless, no synapse-like structures have been
et al., 2011) ion channels, which are regarded as necessary to ini-
observed (Byers et al., 1987; Ibuki et al., 1996; Cardá and Peydró,
tiate those sensory processes (Delmas and Coste, 2013; Nilius and
2006).
Szallasi, 2014; Ranade et al., 2015; Sharif-Naeini, 2015). Further-
The pulp is a highly vascular tissue with a dense capillary plexus
more, it has been demonstrated that gating ion channels present in
under the odontoblast layer that is innervated by postganglionic
odontoblasts induce release of ATP, which is the primary transmit-
sympathetic nerve fibers (Yoshida and Ohshima, 1996).
ter between odontoblasts and nerve fibers (Egbuniwe et al., 2014;
The pattern of innervation of odontoblasts is directly regu-
Liu et al., 2015; Lee et al., 2017) thus initiating the transmission of
lated by local nerve attractive and nerve repulsive molecules (such
a given sensory modality to the central nervous system.
as nerve growth factor, glial-cell line derived neutrophic factor,
This review is an update of the molecular basis of mechano-
sema7A, or reelin) released from the pulp cells and the odonto-
, chemical- and thermo-sensitivity in odontoblasts, based on the

Fig. 2. Double immunofluorecence showing the innervation (green fluorescence)


of human odontoblasts (red fluorescence). The odontoblasts were immunolabelled
Fig. 1. (A) Schematic representation of the odontoblasts and their relations to for detection of vimentin which was found in both the cell bodies and processes.
the dentin and nerve fibers. Pictures show dontoblasts immunolabelled with anti- Spots of green fluorescence (arrows) indicate axons immunolabelled for detection
vimentin (B), anti-TRPV4 (C) and anti-PGP 9.5 antibodies (D). Vimentin and PGP of neurofilaments proteins (NFP). NFP-positive nerve profiles contact odontoblast
9.5 are an intermediate cytoskeletal protein and a cytosolic protein, respectively, cell bodies and also enter the deep segment of dentinal canals. Objective 60×/1.25
present in the odontoblasts and their processes. TRPV4 is an ion channel detected oil; pinhole airy 1, XY resolution 156 nm and Z resolution 334 nm. (For interpretation
in the odontoblasts and related to different modalities of sensitivity. d: dentin; O: of the references to colour in this figure legend, the reader is referred to the web
odontoblasts; p: pulp. version of this article.)
22 A. Solé-Magdalena et al. / Annals of Anatomy 215 (2018) 20–29

blasts themselves; these molecules direct the nerve terminals to with the distinct types of sensibility. In fact, ion channels originally
appropriate sites (Mitsiadis and Luukko, 1995; Luukko et al., 1997; associated with the transduction of one particular form of energy
Maurin et al., 2004, 2005). For example, nerve growth factor and are also activated by different stimuli. In addition, some ion chan-
its high affinity signaling receptor TrkA regulate the development, nels associated with a specific type of stimulus are expressed in
density and maintenance of both nociceptive and postganglionic sensory cells functionally defined as specific for other sensitivities.
nerve fibers (Kirstein and Fariñas, 2002). Consistently, the den- In other words, a specific ion channel can be expressed in more than
tal pulp of TrkA-deficient mice lack nerve fibers in tooth pulp, a sensory cell type, and each cell type may express more than one
including sympathetic fibers, calcitonin gene-related peptide and type of ion channel. Thus, the capacity exhibited by the different
substance P nociceptive fibers (Matsuo et al., 2001; Ichikawa et al., functional types of sensory cells, to preferentially detect a specific
2004). stimulus is the result of a characteristic combinatorial expression of
different ion channels (Liedtke, 2007; Belmonte and Viana, 2008;
Ezak et al., 2010); and this is the case for the trigeminal neurons
3. Dentin sensitivity and ion channels (Vandewauw et al., 2013) and the odontoblasts (Magloire et al.,
2010).
The dentin sensitivity is caused by its exposition to mechanical, Several classes of ion channels have been identified in odon-
chemical or thermal stimuli. When enamel is removed or injured, toblasts which are involved in nociception and other types of
and the dentin tubules opened, the liquid content of the dentinal sensitivity, such as L-type Ca2+ channels, mechanosensitive K+
tubules, the odontoblasts, the trigeminal nerve entering the tooth, channels and voltage-gated Na+ channels (Guo and Davidson, 1998;
and probably also the dental pulp cells, are exposed to the sensi- Magloire et al., 2003, 2009, 2010; Allard et al., 2000, 2006; Solé-
tivizing agents. Thus, dentin sensitivity results from the activation Magdalena et al., 2011; Ichikawa et al., 2012). Here we focused
of dental (i.e., trigeminal) sensory neurons for various mechani- on two superfamilies of ion channels: TRP ion channels and
cal, thermic or chemical stimuli which affect the dentinal fluid, the degenerin/epithelial Na+ channels (DEG/ENa+ C), particularly ASICs.
dentinal tubule content (nerves and odontoblast processes) or the
dental pulp cells. Nevertheless, the molecular mechanisms under-
4. The superfamily of transient receptor potential ion
lying tooth-dentin sensitivity have not been fully elucidated.
channels (TRP)
Three hypotheses are currently considered to explain dentinal
sensitivity. The first hypothesis (nerve theory; Fig. 3a) is based
TRPs are integral membrane proteins that function as ion chan-
on the direct stimulation of dental nerves and the properties
nels. They are non-selective cation channels, a few are highly
of trigeminal primary afferents innervating the tooth, especially
Ca2+ selective and some are permeable for highly hydrated Mg2+ .
the functional expression of mechanosensitive, chemosensitive
The TRP superfamily is subdivided into seven subfamilies: TRPC
or thermosensitive ion channels. The second hypothesis (hydro-
(canonical), TRPV (vanilloid), TRPM (melastatin), TRPP (polycystin),
dynamic theory; Fig. 3b) attributes dentinal sensitivity to nerve
TRPML (mucolipin), TRPA (ankyrin) and TRPN (NOMPC-like); the
stimulation by fluid movement within dentinal tubules: cold stimu-
latter being found only in invertebrates and fishes. At least 28 dif-
lation causes an outward fluid flow while a hot stimulation induces
ferent TRP proteins have been identified in mammals. Structurally,
an inward fluid flow in dentinal tubules (Linsuwanont et al., 2007).
a typical TRP protein contains six putative transmembrane domains
Moreover, the pulp cells and nerves can also detect the tempera-
(S1 to S6) with a pore-forming reentrant loop between S5 and
ture and chemical composition of the fluid (Andrew and Matthews,
S6. Intracellular N- and C-termini are variable in length and con-
2002). The third hypothesis (odontoblastic theory; Fig. 3c) is consis-
sist of a variety of domains (Clapham et al., 2005; Hellmich and
tent with the capability of the odontoblasts to sense diverse stimuli.
Gaudet, 2014; Nilius and Szallasi, 2014). This ion channel superfam-
The presence in these cells of specific ion channels strongly sug-
ily shows a variety of gating mechanisms with modes of activation
gest that they are able to detect mechanical, chemical, and thermal
ranging from ligand binding, voltage and changes in temperature to
signals (Chung and Oh, 2013; Chung et al., 2013).
covalent modifications of nucleophilic residues (see for a review Eid
These three theories are not mutually exclusive and cannot
and Cortright, 2009; Nilius and Owsianik, 2011; Nilius and Szallasi,
be considered separately because of the presence of nerves and
2014).
odontoblast processes within the dentinal tubules, bathing in the
dentinal fluid, and the close apposition of the odontoblasts to the
dentinal or basal nerves terminals. Thus, external stimuli modify- 5. The superfamily of acid-sensing ion channels
ing the dentinal fluid induce responses in the odontoblasts, in the
nerves, and in the odontoblast–nerve complex, and this may rep- ASICs are Na+ -selective cation channels which are voltage-
resent a unique polymodal sensory system responsible for dentin insensitive and amiloride-sensitive and monitor moderate devia-
sensibility. In this context the odontoblasts play a pivotal role in sig- tions from the physiological values of extracellular pH (Waldmann
nal transduction, but how they sense signals and how these signals et al., 1997; Lingueglia, 2007; Lumpkin and Caterina, 2007; Baron
are transmitted to axons are questions which have been resolved and Lingueglia, 2015). Six ASIC proteins encoded by four genes have
in part only recently. been identified: ASIC1a, ASIC1b, ASIC2a, ASIC2b, ASIC3, and ASIC4
Various studies in non-vertebrates and vertebrates have iden- which differ in their kinetics, external pH sensitivity, tissue dis-
tified several ion channels that are responsible, or are required, tribution and pharmacological properties (Krishtal, 2003). The pH
for detecting a range of thermal, chemical or mechanical stimuli, values required for half-maximal activation are 6.2–6.8 for ASIC1a,
and stimuli-transduction ion channels are known for most sensory 5.9–6.2 for ASIC1b, 4.9 for ASIC2a and 6.5–6.7 for ASIC3 (Kress
modalities (Belmonte and Viana, 2008; Damann et al., 2008; Nilius and Waldmann, 2006; Hanukoglu, 2017). In addition, some ASICs
and Szallasi, 2014; Jardín et al., 2017). may work as mechanosensors (or are required for mechanosen-
The identification of ion channels selectively activated by differ- sation) and nociception (Wemmie et al., 2006; Holzer, 2009, 2011;
ent stimuli, supported the concept that the specificity of a sensory Sherwood et al., 2012; Zha, 2013; Holzer and Izzo, 2014; Omerbašić
cell is determined by their expression of a particular ion channel et al., 2015). Structurally, ASICs consist of two transmembrane
conferring its selectivity to respond to a unique stimulus. Never- domains and a large extracellular loop (Sherwood et al., 2012).
theless, it is currently accepted that the ion channels proposed as The members of these two families of ion channels exhibiting
specific transducers are not as neatly and selectively associated mechanosensitivity, thermosensitivity and chemosensitivity are
A. Solé-Magdalena et al. / Annals of Anatomy 215 (2018) 20–29
Fig. 3. Schematic representation of the three main theories explaining the dentin sensitivity. The nerve theory postulates the direct stimulation of dentinal tubules and pulpar nerve terminals; the hydrodynamic theory assumes
stimulation of dental nerves by dentinal fluid; and the odontoblastic theory postulates direct stimulation of odontoblast, and is based on the expression of several ion channels by these cells.

23
24 A. Solé-Magdalena et al. / Annals of Anatomy 215 (2018) 20–29

Table 1 them have been detected in the odontoblasts or trigeminal affer-


Members of the TRP and Deg/ENaC superfamilies exhibiting mechano-, thermo- and
ent neurons. TRPV1, TRPV2, TRPV3, and TRPV4 have incompletely
chemo-sensitivity (based on Belmonte and Viana, 2008).
overlapping functions over a broad thermal range from warm to
Mechanosensing Thermosensing Chemiosensing hot. While TRPA1 and TRPM8 respond to cool and cold, TRPV1 and
TRPC1 TRPV2 are activated by painful levels of heat (>43 ◦ C and >52 ◦ C,
PRPC6 respectively), TRPV3 and TRPV4 respond to non-painful warmth
TRPV1 TRPV1 TRPV1 (33–39 ◦ C), TRPM 8 is activated by non-painful cool temperatures
TRPV2 TRPV2
(<25 ◦ C), and TRPA 1 is activated by painful cold (<18 ◦ C; Palkar
TRPV3
TRPV4 TRPV4 et al., 2015). TRPV1 is also responsive to noxious stimuli and vari-
TRPM3 ous chemical agents (Reid, 2005; Nieto-Posadas et al., 2011; Wetsel,
TRPM4 2011; Vay et al., 2012; Voets, 2012).
TRPM8 TRPM8
Heat- and cold-sensing TRP channels, TRPV1, TRPV2, and TRPV3,
TRPA1 TRPA1
TRPP2 TRPM8 and TRPA1 are functionally expressed in odontoblasts (Son
et al., 2009; El Karim et al., 2011; Kim et al., 2012; Sato et al., 2013),
ASIC 1 ASIC1 ASIC1
but the results vary widely among species. Cultured mouse odonto-
ASIC2 ASIC2 ASIC2
ASIC3 ASIC3 ASIC3 blastic cells express TRPV1, TRPV2, TRPV3, TRPV4, and TRPM3, but
not TRPM8 and TRPA1 (Son et al., 2009), while odontoblasts from
adults rats express TRPA1 (Byers and Westenbroek, 2011) and lack
summarized in Table 1. In the following pages the occurrence of TRPV1 or TRPV2 (Yeon et al., 2009). In partial disagreement with
these ion channels in the odontoblast and the nerve fibres supply- these findings, Tsumura et al. (2012, 2013) have detected functional
ing them will be detailed. Regarding the trigeminal neurons, almost expression of TRPV1, TRPM8 and TRPA1 channels in rat odonto-
all the ion channels involved in all sensory modalities have been blasts. The human odontoblasts contain TRPM8, TRPA1 along with
detected in these cells. In fact, in the murine trigeminal neurons 17 TRPV1 (Okumura et al., 2005; El Karim et al., 2011; Tazawa et al.,
of the 28 TRP channel genes were detectable (TRPA1, TRPC1, TRPC3, 2017). Finally, in human immortalized dental pulp cells derived
TRPC4, TRPC5, TRPM2, TRPM3, TRPM4, TRPM5, TRPM6, TRPM7, from an odontoblast phenotype, Egbuniwe et al. (2014) demon-
TRPM8, TRPV1, TRPV2, TRPV4, TRPML1 and TRPP2; Vandewauw strated expression of mRNA for TRPA1, TRPV1, and TRPV4 but not
et al., 2013), while in the human trigeminal ganglion 10 TRPs TRPM8. Nevertheless, although all these putative thermal sensors
(TRPC1, TRPM2, TRPM3, TRPM7, TRPM8, TRPV1, TRPV2, TRPV3, are present in odontoblasts, it is unlikely that these cells serve as
TRPV4 and TRPML1) and ASIC1-3 have been identified (Flegel et al., thermal sensors (Yeon et al., 2009).
2015; Fu et al., 2016). TRPV1, TRPV2 and TRPA1 are also expressed in dental affer-
ents whereas primary afferent trigeminal neurons express TRPV1,
6. Putative mechanosensors in odontoblasts and TRPV4, TRPA1, TRPM8 and TRPM3 (Story et al., 2003; Park et al.,
trigeminal afferent fibers and neurons 2006; Chung et al., 2013). In the human dental pulp, TRPA1 was
expressed in a large number of axons branching in the peripheral
To understand what mechanoproteins are, it is necessary to have pulp as well as in the cell body of odontoblasts (Kim et al., 2012).
in mind that they are not receptors and that mechanical forces
are not ligands. The force is rather an “anti-ligand” because it can
disrupt intramolecular or intermolecular interactions to trigger a
conformational transmission (see Sukharev and Anishkin, 2004).
Most of the ion channels candidates for mechanosensors belong
to the superfamilies of DEG/ENaC and TRP ion channels (see for
a review Gillespie and Walker, 2001; Lumpkin and Caterina, 2007; 8. Putative chemosensors in odontoblasts and trigeminal
Arnadottir and Chalfie, 2010; Delmas and Coste, 2013; Ranade et al., afferent fibers and neurons
2015). Moreover, members of the two potassium pore (K2P ) chan-
nels and the product of Piezo1 and Piezo2 genes have been also The chemosensory capacity of the sensory systems relies on the
identified as components of mechanically activated cation channels appropriate expression of chemoreceptors, which detect chemical
(Coste et al., 2010; Delmas and Coste, 2013; Ranade et al., 2015). stimuli and transduce sensory information into cellular signals. The
The putative mechanosensors TRPV4, TRPM3, TRPP1 and TRPP2 molecular mechanism of chemosensation in the trigeminal system
are expressed in cultured mouse odontoblasts (Son et al., 2009; Sato has been revised recently by Viana (2010), but without any direct
et al., 2013), whereas the adult rat odontoblasts express TRPM7, reference to the dental sensitivity.
TRPC1, TRPC6, and TRPV4 (Kwon et al., 2014), and human odonto- Most of the authors suggest that chemosensation is determined
blasts display immunoreactivity for TRPV4, ASIC2 and the ␤- and primarily by the chemical activation of nociceptors and thermore-
ϒ-ENaC, but not ␣-ENaC, subunits (Solé-Magdalena et al., 2011). ceptors, and that activation by chemicals involves the direct activity
TRPP1 and TRPP2, which act together as a mechanical receptor, are of an ion channel by chemical stimuli: the so-called ionotrophic
present at the surface of odontoblasts, and appear to be located at transduction (Wood and Docherty, 1997; Lee et al., 2005). The
the base of the primary cilium (Thivichon-Prince et al., 2009). ionotropic channels include some TRPs, ASICs and two-pore K2 P
TRPM3, TRPV4, TRPA1, ASIC3, ENaC-␣ and ENaC-ϒ ion chan- channels in trigeminal neurons (Caterina et al., 1997; Tominaga
nels have been detected in primary afferent dental neurons and Tominaga, 2005; Bautista and Julius, 2008; Viana, 2010; Flegel
(Hermanstyne et al., 2008; Vandewauw et al., 2013). et al., 2015), and at least TRPM5 in the odontoblasts (Khatibi Shahidi
et al., 2015).
7. Putative thermosensors in odontoblasts and trigeminal The data about the presence of primary TRP and ASIC ion
afferent fibers/neurons channels involved in mechanosensitivity and thermosensitivity
in odontoblasts, pulpar nerve fibres and trigeminal neurons are
Six members of the TRP superfamily, TRPA1, TRPM8, TRPV1, summarized in Fig. 4. Moreover, Fig. 5 show pictures of human
TRPV2, TRPV3, and TRPV4, proposed to participate in thermosen- odontoblasts showing immunoreactivity for some ion channels
sation have been detected in sensory nerves, but only some of mentioned in this review.
A. Solé-Magdalena et al. / Annals of Anatomy 215 (2018) 20–29 25

than dental pulp (Bron et al., 2014) and, recently, Khatibi Shahidi
et al. (2015) found occurrence of Piezo2 in murine odontoblasts.

10. The primary cilium of the odontoblasts and its relation


to TRP and ASIC ion channels

Typically, the odontoblasts contain a primary cilium than is


involved in odontogenesis (Thivichon-Prince et al., 2009), but also
in detection of fluid movement in dentinal tissue (elicited by high
pressure, osmotic, chemical or thermal stimuli) that may cause a
cilium deformation thus initiating a signal transduction pathway
(Magloire et al., 2004). The primary cilium is a cell system which
senses and transduces mechanical and chemical stimuli (Pazour
Fig. 4. Schematic representation of the distribution of TRP and ASIC ion channels in and Witman, 2003; Praetorius and Spring, 2005; Muhammad et al.,
odontoblasts, dental nerve fibres and trigeminal ganglia neurons. 2012; Prasad et al., 2014). Both processes involve Ca2+ entry and
require the presence of stretch-activated Ca2+ channels (poly-
9. Other mechano- and thermos-sensitive proteins in cystins) localized in the cilia (Delmas, 2004; Delmas et al., 2004;
odontoblasts Praetorius and Spring, 2005). It is also noteworthy that the mem-
brane of the cilium contains multiple channel proteins, including
In addition to the above mentioned TRP and ASIC ion channels, a variety of Ca2+ -ion permeable channels that play a role in Ca2+ -
members of the two-pore domain potassium (K2p ) channels fam- mediated fluid-flow mechanosensation (Anishkin and Kung, 2013;
ily, and the product of Piezo1 and Piezo2 genes are also considered Delling et al., 2013). Very probably, ASICs, which have been local-
as putative mechanotransducer channels (Lumpkin and Caterina, ized in the cilia of various tissues, are among these ion channels
2007; Coste et al., 2010; Lumpkin et al., 2010; Ranade et al., 2015; (Kikuchi et al., 2008; Kikuchi et al., 2010; Viña et al., 2015), as
Sharif-Naeini, 2015). well TRP channels, since the cilium is a unique Ca2+ compart-
K2P ion channels consist of six subfamilies within the super- ment regulated by a heteromeric TRP channel (Giamarchi et al.,
family of K+ -selective channel subunits (Yost, 2003; Sabbadinia 2006; Delling et al., 2013). On the other hand, the mechanosen-
and Yost, 2009), and two members of this superfamily, TREK1 and sory and chemosensory functions of the cilium are those of ASIC2
TRAAK, are among the few channels for which a direct mechanical (see Sherwood et al., 2012), thus it can be speculated that ASIC2
gating has been demonstrated (Maingret et al., 2000). TREK/TRAAK participates in the ciliary function.
channels are co-expressed with TRP-thermo channels, and can also
work as nociceptors, thermosensors and controlling pain produced 11. Intercellular signal transmission between odontoblasts
by mechanical stimulation (Maingret et al., 1999a, 1999b, 2000; and nerves
Chemin et al., 2005; Alloui et al., 2006; Noël et al., 2009). Inter-
estingly, TREK1 and TREK2 are present in neurons innervating the How is the firing of odontoblasts transmitted to nerve endings
dental pulp (Hermanstyne et al., 2008), and the transcripts of TREK1 supplying them? As previously mentioned, no specific synaptic-like
are expressed in the human odontoblasts (Magloire et al., 2003). structures have been observed between nerve fibers and odonto-
On the other hand, it has been demonstrated that the products blasts even when they are in close proximity (Byers, 1984; Cardá
of Piezo1 and Piezo2 genes are rapidly adapting, mechanically acti- and Peydró, 2006). Nevertheless, release of mediators from stim-
vated ion channels (Ranade et al., 2014; Volkers et al., 2015). Piezo2 ulated odontoblasts into the gap-space between odontoblasts and
has been reported in trigeminal neurons innervating tissues other nerves, followed by signaling to dental afferents, are sine qua non

Fig. 5. Immunohistochemical localization of some TRP and ASIC1 ion channels in human odontoblasts. Positive immunostaining was detected in both the cell bodies and
processes. d: dentin; o: odontoblasts; p: pulp.
26 A. Solé-Magdalena et al. / Annals of Anatomy 215 (2018) 20–29

pulp cells (Alavi et al., 2001; Renton et al., 2003; Wang et al., 2016).
Moreover, myelinated and unmyelinated P2X positive nerve fibers
were detected in the subodontoblastic plexus in close association
with odontoblasts (Cook et al., 1997; Alavi et al., 2001; Sharma and
Pradeep, 2006). In rats, P2XR2 and P2XR3 receptors were found
in both pulp nerves and a subpopulation of trigeminal ganglion
neurons (Matsuka et al., 2001; Jiang and Gu, 2002; Staikopoulos
et al., 2007; Chung et al., 2008; Kuroda et al., 2012). Furthermore,
odontoblast themselves express several P2XRs of different sub-
types (Lee et al., 2017; Shiozaki et al., 2017) thus suggesting that
ATP might regulate the physiology of the odontoblasts throughout
autocrine–paracrine mechanisms, since inhibition of interodonto-
blastic communication can be reached blocking of ATP release (Lee
et al., 2017). Furthermore, P2Y receptor subtypes are present in
pulp cells (Wang et al., 2016), trigeminal neurons (Li et al., 2014;
Kawaguchi et al., 2015) and trigeminal satellite glial cells (Magni
et al., 2015), as well as in odontoblasts (Sato et al., 2015; Wang
Fig. 6. Schematic representation of the molecular mechanisms coupling direct et al., 2016).
odontoblast stimulation or activation of odontoblastic TRP/ASIC ion channels that It has been traditionally accepted that mechanical distortion or
results in ATP outflow. The extracellular ATP binds to specific receptors present in direct stimulation of the odontoblasts lead to ATP release, presum-
nerve fibers, odontoblasts and other pulp cells acting via synaptic-like (grey lines), ably acting via ion channels. The activation of TRPA1 and TRPV4,
autocrine (red line) or paracrine (blue line) mechanisms. (For interpretation of the
references to colour in this figure legend, the reader is referred to the web version
but not TRPV1, in human odontoblast-like cells causes an increase
of this article.) in ATP release than can be abolished by selective TRP antagonists
(Egbuniwe et al., 2014). Also, following mechanical stimuli activ-
ity of TRPV1, TRPV3, TRPV4 and TRPA1, but not TRPM8, there is
conditions supporting the hypothesis that odontoblasts are sensory a release of ATP via pannexina-1 that transmits a signal to P2X3
cells, and, thus, the odontogenic theory of dentin sensitivity. receptors of trigeminal neurons (Shibukawa et al., 2013, 2015; Sato
Among the proposed mediators are nitric oxide (Korkmaz et al., et al., 2015; Fig. 6).
2005), galanin (Suzuki et al., 2002), glutamate (Nishiyama et al.,
2016) and, especially, extracellular purines such as ATP and adeno- 12. Future perspectives: TRPs, ASICs, and the ATP signaling
sine (see for a review Lim and Mitchel, 2012; Lee et al., 2017); system as targets for the treatment of dentin sensitivity
the process of signaling throughout purines is known as puriner-
gic transmission. Thus, purinergic signaling refers to the process The expression of mechanosensing, thermosensing and
by which purines and pyrimidines mediate cellular responses fol- chemosensing ion channels by odontoblasts; the close relation-
lowing stimulation of specific receptors. Every cell has a supply of ships between odontoblasts and nerve fibers; the release of ATP by
cytoplasmic ATP that can be released under physiological condi- the odontoblasts in response to, at least, mechanical stimulation;
tions through fusion of ATP-containing vesicles with the plasma the existence of an enzymatic apparatus for ATP degradation
membrane in a classic release pathway. Also, ATP can passively in dental pulp; and the expression of ATP receptors in dental
efflux out of the cell through permeant membrane pores or chan- afferents, strongly suggests that ATP might act as a signaling
nels (anion channels, connexin and pannexin hemichannels), or via molecule between odontoblasts and neurons in the sensory
a hybrid mechanism throughout Ca2+ -dependent fusion of vesi- transduction sequence for dentinal sensibility (Lim and Mitchell,
cles containing permeant channels with the plasma membrane 2012; Shibukawa et al., 2015). Taken together, these steps in the
(Reigada and Mitchell, 2005). Odontoblasts expressed the gap transmission of signals demonstrate the key role of odontoblasts as
junction protein connexin43, or pannexin-1 which can form trans- sensory cells, lending support to the odontoblastic theory of dentin
membrane hemichannels for ATP release (Liu et al., 2012; Fig. 6). sensitivity (Fig. 6). Consistenty, the disruption of these sequential
Multiple release mechanisms may exist within the same cell events using appropriate pharmacological modulators for TRP and
able to be activated by different stimuli, although ATP-release is fre- ASIC ion channels (El Karim et al., 2011; Alexander et al., 2013) or
quently triggered by mechanical stimuli (Xia et al., 2012). Released prurinergic transmission (Burnstock, 2006, 2009) would provide a
ATP acts over specific receptors and is then rapidly dephosphory- novel therapeutic approach in the treatment of dentin sensitivity.
lated by a series of extracellular enzymes converting ATP directly
into AMP and pyrophosphate (Goding et al., 2003; Zimmermann, References
2006).
The purinergic receptors fall into two main categories termed P1 Alavi, A.M., Dubyak, G.R., Burnstock, G., 2001. Immunohistochemical evidence for
and P2 receptors (Burnstock, 1978). P1 are receptors for adenosine, ATP receptors in human dental pulp. J. Dent. Res. 80, 476–483.
Alexander, S.P., Benson, H.E., Faccenda, E., Pawson, A.J., Sharman, J.L., Catterall,
and P2 receptors for ATP. P1 receptors are G protein-coupled recep- W.A., Spedding, M., Peters, J.A., Harmar, A.J., CGTP Collaborators, 2013. The Con-
tors. P2 receptors are subclassified into ionotropic cation channels cise Guide to PHARMACOLOGY 2013/14: ion channels. Br. J. Pharmacol. 170,
P2X receptors and G-protein-coupled P2Y receptors. To date, seven 1607–1651.
Allard, B., Couble, M.L., Magloire, H., Bleicher, F., 2000. Characterization and gene
P2X receptors (P2 × 1R-7R) and eight P2Y receptors (P2Y1R, P2Y2R,
expression of high conductance calcium-activated potassium channels display-
P2Y4R, P2Y6R, P2Y11R, P2Y12R, P2Y13R and P2Y14R) have been ing mechanosensitivity in human odontoblasts. J. Biol. Chem. 275, 25556–25561.
identified in mammals (Burnstock, 2006, 2006; Coddou et al., 2011; Allard, B., Magloire, H., Couble, M.L., Maurin, J.C., Bleicher, F., 2006. Voltage-gated
sodium channels confer excitability to human odontoblasts: possible role in
von Kugelgen and Harden, 2011).
tooth pain transmission. J. Biol. Chem. 281, 29002–29010.
The P2X receptors, differ in their rates of inactivation and sen- Alloui, A., Zimmermann, K., Mamet, J., Duprat, F., Noël, J., Chemin, J., Guy, N., Blon-
sitivity to ATP, the P2 × 7R requiring the highest concentrations deau, N., Voilley, N., Rubat-Coudert, C., Borsotto, M., Romey, G., Heurteaux, C.,
of ATP for activation and inactivating most slowly (North, 2002). Reeh, P., Eschalier, A., Lazdunski, M., 2006. TREK-1, a K+ channel involved in
polymodal pain perception. EMBO J. 25, 2368–2376.
P2XRs are expressed in the nociceptive trigeminal ganglion cells Andrew, D., Matthews, B., 2002. Properties of single nerve fibres that evoke blood
(Staikopoulos et al., 2007; Kim et al., 2008) as well as in the dental flow changes in cat dental pulp. J. Physiol. 542, 921–928.
A. Solé-Magdalena et al. / Annals of Anatomy 215 (2018) 20–29 27

Anishkin, A., Kung, C., 2013. Stiffened lipid platforms at molecular force foci. Proc. Gillam, D.G., 1995. Mechanisms of stimulus transmission across dentin—a review. J.
Natl. Acad. Sci. U. S. A. 110, 4886–4892. West Soc. Periodontol. Periodontal. Abstr. 43, 53–65.
Arana-Chavez, V.E., Massa, L.F., 2004. Odontoblasts: the cells forming and maintain- Gillespie, P.G., Walker, R.G., 2001. Molecular basis of mechanosensory transduction.
ing dentine. Int. J. Biochem. Cell. Biol. 36, 1367–1373. Nature 413, 194–202.
Arnadottir, J., Chalfie, M., 2010. Eukaryotic mechanosensitive channels. Annu. Rev. Goding, J.W., Grobben, B., Slegers, H., 2003. Physiological and pathophysiological
Biophys. 39, 111–137. functions of the ecto-nucleotide pyrophosphatase/phosphodiesterase family.
Baron, A., Lingueglia, E., 2015. Pharmacology of acid-sensing ion channels — physi- Biochim. Biophys. Acta 1638, 1–19.
ological and therapeutical perspectives. Neuropharmacology 94, 19–35. Guo, L., Davidson, R.M., 1998. Potassium and chloride channels in freshly isolated
Bautista, D., Julius, D., 2008. Fire in the hole: pore dilation of the capsaicin receptor rat odontoblasts. J. Dent. Res. 77, 341–350.
TRPV1. Nat. Neurosci. 11, 528–529. Hanukoglu, I., 2017. ASIC and ENaC type sodium channels: conformational states
Belmonte, C., Viana, F., 2008. Molecular and cellular limits to somatosensory speci- and the structures of the ion selectivity filters. FEBS J. 284, 525–545.
ficity. Mol. Pain 4, 14. Hellmich, U.A., Gaudet, R., 2014. Structural biology of TRP channels. Handb. Exp.
Bleicher, F., 2014. Odontoblast physiology. Exp. Cell Res. 325, 65–71. Pharmacol. 23, 963–990.
Bron, R., Wood, R.J., Brock, J.A., Ivanusic, J.J., 2014. Piezo2 expression in corneal Hermanstyne, T.O., Markowitz, K., Fan, L., Gold, M.S., 2008. Mechanotransducers in
afferent neurons. J. Comp. Neurol. 522, 2967–2979. rat pulpal afferents. J. Dent. Res. 87, 834–838.
Burnstock, G., 1978. A basis for distinguishing two types of purinergic receptor. In: Holzer, P., 2009. Acid-sensitive ion channels and receptors. Handb. Exp. Pharmacol.
Straub, R.W., Bolis, L. (Eds.), Cell Membrane Receptors for Drugs and Hormones: 194, 283–332.
A Multidisciplinary Approach. Raven Press, New York, NY, pp. 107–118. Holzer, P., 2011. Acid sensing by visceral afferent neurones. Acta Physiol. 201, 63–75.
Burnstock, G., 2006. Purinergic P2 receptors as targets for novel analgesics. Pharma- Holzer, P., Izzo, A.A., 2014. The pharmacology of TRP channels. Br. J. Pharmacol. 171,
col. Ther. 110, 433–454. 2469–2473.
Burnstock, G., 2009. Purines and sensory nerves. Handb. Exp. Pharmacol. 194, Ibuki, T., Kido, M.A., Kiyoshima, T., Terada, Y., Tanaka, T., 1996. An ultrastructural
333–392. study of the relationship between sensory trigeminal nerves and odontoblasts
Byers, M.R., 1984. Dental sensory receptors. Int. Rev. Neurobiol. 25, 39–94. in rat dentin/pulp as demonstrated by the anterograde transport of wheat germ
Byers, M.R., Närhi, M.V., 1999. Dental injury models: experimental tools for under- agglutinin–horseradish peroxidase (WGA–HRP). J. Dent. Res. 75, 1963–1970.
standing neuroinflammatory interactions and polymodal nociceptor functions. Ichikawa, H., Matsuo, S., Silos-Santiago, I., Jacquin, M.F., Sugimoto, T., 2004. The
Crit. Rev. Oral Biol. Med. 10, 4–39. development of myelinated nociceptors is dependent upon trks in the trigeminal
Byers, M.R., Närhi, M.V., Dong, W.K., 1987. Sensory innervation of pulp and dentin in ganglion. Acta Histochem. 106, 337–343.
adult dog teeth as demonstrated by autoradiography. Anat. Rec. 218, 207–215. Ichikawa, H., Kim, H.J., Shuprisha, A., Shikano, T., Tsumura, M., Shibukawa, Y., Tazaki,
Byers, M.R., Westenbroek, R.E., 2011. Odontoblasts in developing, mature and ageing M., 2012. Voltage-dependent sodium channels and calcium-activated potassium
rat teeth have multiple phenotypes that variably express all nine voltage-gated channels in human odontoblasts in vitro. J. Endod. 38, 1355–1362.
sodium channels. Arch. Oral Biol. 56, 1199–1220. Jardín, I., López, J.J., Diez, R., Sánchez-Collado, J., Cantonero, C., Albarrán, L., Woodard,
Cardá, C., Peydró, A., 2006. Ultrastructural patterns of human dentinal tubules, odon- G.E., Redondo, P.C., Salido, G.M., Smani, T., Rosado, J.A., 2017. TRPs in pain sen-
toblasts processes and nerve fibres. Tissue Cell 38, 141–150. sation. Front Physiol. 8, 392.
Caterina, M.J., Schumacher, M.A., Tominaga, M., Rosen, T.A., Levine, J.D., Julius, D., Jiang, J., Gu, J., 2002. Expression of adenosine triphosphate P2 × 3 receptors in rat
1997. The capsaicin receptor: a heat-activated ion channel in the pain pathway. molar pulp and trigeminal ganglia. Oral Surg. Oral Med. Oral Pathol. Oral Radiol.
Nature 389, 816–824. Endod. 94, 622–626.
Chemin, J., Patel, A.J., Duprat, F., Lauritzen, I., Lazdunski, M., Honoré, E., 2005. A Kaneko, Y., Szallasi, A., 2014. Transient receptor potential (TRP) channels: a clinical
phospholipid sensor controls mechanogating of the K+ channel TREK-1. EMBO perspective. Br. J. Pharmacol. 171, 2474–2507.
J. 24, 44–53. Kawaguchi, A., Sato, M., Kimura, M., Ichinohe, T., Tazaki, M., Shibukawa, Y., 2015.
Chung, M.K., Güler, A.D., Caterina, M.J., 2008. TRPV1 shows dynamic ionic selectivity Expression and function of purinergic P2Y12 receptors in rat trigeminal ganglion
during agonist stimulation. Nat. Neurosci. 11, 555–564. neurons. Neurosci. Res. 98, 17–27.
Chung, G., Jung, S.J., Oh, S.B., 2013. Cellular and molecular mechanisms of dental Kawashima, N., Okiji, T., 2016. Odontoblasts: specialized hard-tissue-forming cells
nociception. J. Dent. Res. 92, 948–955. in the dentin-pulp complex. Congenit. Anom. (Kyoto) 56, 144–153.
Chung, G., Oh, B.S., 2013. TRP channels in dental pain. Open Pain J. 6 (Suppl. 1), 31–36. Khatibi Shahidi, M., Krivanek, J., Kaukua, N., Ernfors, P., Hladik, L., Kostal, V.,
Clapham, D.E., Julius, D., Montell, C., Schultz, G., 2005. International Union of Pharma- Masich, S., Hampl, A., Chubanov, V., Gudermann, T., Romanov, R.A., Harkany,
cology: XLIX. Nomenclature and structure–function relationships of transient T., Adameyko, I., Fried, K., 2015. Three-dimensional imaging reveals new com-
receptor potential channels. Pharmacol. Rev. 57, 427–450. partments and structural adaptations in odontoblasts. J. Dent. Res. 94, 945–954.
Coddou, C., Yan, Z., Obsil, T., Huidobro-Toro, J.P., Stojilkovic, S.S., 2011. Activation and Kikuchi, S., Ninomiya, T., Kawamata, T., Ogasawara, N., Kojima, T., Tachi, N., Tatsumi,
regulation of purinergic P2X receptor channels. Pharmacol. Rev. 63, 641–683. H., 2010. The acid-sensing ion channel 2 (ASIC2) of ciliated cells in the developing
Cook, S.P., Vulchanova, L., Hargreaves, K.M., Elde, R., McCleskey, E.W., 1997. Dis- rat nasal septum. Arch. Histol. Cytol. 73, 81–89.
tinct ATP receptors on pain-sensing and stretch-sensing neurons. Nature 387, Kikuchi, S., Ninomiya, T., Kawamata, T., Tatsumi, H., 2008. Expression of ASIC2 in
505–508. ciliated cells and stereociliated cells. Cell Tissue Res. 333, 217–224.
Coste, B., Mathur, J., Schmidt, M., Earley, T.J., Ranade, S., Petrus, M.J., Dubin, A.E., Kim, Y.S., Jung, H.K., Kwon, T.K., Kim, C.S., Cho, J.H., Ahn, D.K., Bae, Y.C., 2012. Expres-
Patapoutian, A., 2010. Piezo1 and Piezo2 are essential components of distinct sion of transient receptor potential ankyrin 1 in human dental pulp. J. Endod.
mechanically activated cation channels. Science 330, 55–60. 38, 1087–1092.
Damann, N., Voets, T., Nilius, B., 2008. TRPs in our senses. Curr. Biol. 18, R880–R889. Kim, Y.S., Paik, S.K., Cho, Y.S., Shin, H.S., Bae, J.Y., Moritani, M., Yoshida, A., Ahn, D.K.,
Delling, M., DeCaen, P.G., Doerner, J.F., Febvay, S., Clapham, D.E., 2013. Primary cilia Valtschanoff, J., Hwang, S.J., Moon, C., Bae, Y.C., 2008. Expression of P2 × 3 recep-
are specialized calcium signalling organelles. Nature 504, 311–314. tor in the trigeminal sensory nuclei of the rat. J. Comp. Neurol. 506, 627–639.
Delmas, P., 2004. Polycystins: from mechanosensation to gene regulation. Cell 118, Kirstein, M., Fariñas, I., 2002. Sensing life: regulation of sensory neuron survival by
145–148. neurotrophins. Cell Mol. Life Sci. 59, 1787–1802.
Delmas, P., Coste, B., 2013. Mechano-gated ion channels in sensory systems. Cell Korkmaz, Y., Baumann, M.A., Steinritz, D., Schröder, H., Behrends, S., Addicks, K.,
155, 278–284. Schneider, K., Raab, W.H., Bloch, W., 2005. NO-cGMP signaling molecules in cells
Delmas, P., Padilla, F., Osorio, N., Coste, B., Raoux, M., Crest, M., 2004. Polycystins, of the rat molar dentin-pulp complex. J. Dent. Res. 84, 618–623.
calcium signaling, and human diseases. Biochem. Biophys. Res. Commun. 322, Kress, M., Waldmann, R., 2006. Acid sensing ionic channels. Curr. Top. Membr. 57,
1374–1383. 241–276.
Eid, S.R., Cortright, D.N., 2009. Transient receptor potential channels on sensory Krishtal, O., 2003. The ASICs: signaling molecules? Modulators? Trends Neurosci.
nerves. Handb. Exp. Pharmacol. 194, 261–281. 26, 477–483.
Egbuniwe, O., Grover, S., Duggal, A.K., Mavroudis, A., Yazdi, M., Renton, T., Di Silvio, L., Kuroda, H., Shibukawa, Y., Soya, M., Masamura, A., Kasahara, M., Tazaki, M., Ichi-
Grant, A.D., 2014. TRPA1 and TRPV4 activation in human odontoblasts stimulates nohe, T., 2012. Expression of P2X1 and P2X4 receptors in rat trigeminal ganglion
ATP release. J. Dent. Res. 93, 911–917. neurons. Neuroreport 23, 752–756.
El Karim, I.A., Linden, G.J., Curtis, T.M., About, I., McGahon, M.K., Irwin, C.R., Lundy, Kweon, H.J., Suh, B.C., 2013. Acid-sensing ion channels (ASICs): therapeutic targets
F.T., 2011. Human odontoblasts express functional thermo-sensitive TRP chan- for neurological diseases and their regulation. BMB Rep. 46, 295–304.
nels: implications for dentin sensitivity. Pain 152, 2211–2223. Kwon, M., Baek, S.H., Park, C.K., Chung, G., Oh, S.B., 2014. Single-cell RT-PCR and
Ezak, M.J., Hong, E., Chaparro-Garcia, A., Ferkey, D.M., 2010. Caenorhabditis elegans immunocytochemical detection of mechanosensitive transient receptor poten-
TRPV channels function in a modality-specific pathway to regulate response to tial channels in acutely isolated rat odontoblasts. Arch. Oral Biol. 59, 1266–1271.
aberrant sensory signaling. Genetics 185, 233–244. Lee, B.M., Jo, H., Park, G., Kim, Y.H., Park, C.K., Jung, S.J., Chung, G., Oh, S.B., 2017.
Flegel, C., Schöbel, N., Altmüller, J., Becker, C., Tannapfel, A., Hatt, H., Gisselmann, Extracellular ATP induces calcium signaling in odontoblasts. J. Dent. Res. 96,
G., 2015. RNA-Seq analysis of human trigeminal and dorsal root ganglia with a 200–207.
focus on chemoreceptors. PLoS One 10, e0128951. Lee, Y., Lee, C.H., Oh, U., 2005. Painful channels in sensory neurons. Mol. Cells 20,
Fu, H., Fang, P., Zhou, H.Y., Zhou, J., Yu, X.W., Ni, M., Zheng, J.Y., Jin, Y., Chen, J.G., 315–324.
Wang, F., Hu, Z.L., 2016. Acid-sensing ion channels in trigeminal ganglion neu- Li, N., Lu, Z.Y., Yu, L.H., Burnstock, G., Deng, X.M., Ma, B., 2014. Inhibition of G
rons innervating the orofacial region contribute to orofacial inflammatory pain. protein-coupled P2Y2 receptor induced analgesia in a rat model of trigeminal
Clin. Exp. Pharmacol. Physiol. 43, 193–202. neuropathic pain. Mol. Pain 10, 21.
Giamarchi, A., Padilla, F., Coste, B., Raoux, M., Crest, M., Honoré, E., Delmas, P., 2006. Liedtke, W.B., 2007. TRPV channels’ function in osmo- and mechanotransduction. In:
The versatile nature of the calcium-permeable cation channel TRPP2. EMBO Rep. Liedtke, W.B., Heller, S. (Eds.), TRP Ion Channel Function in Sensory Transduction
7, 787–793. and Cellular Signaling Cascades. CRC Press, Boca Raton, FL.
28 A. Solé-Magdalena et al. / Annals of Anatomy 215 (2018) 20–29

Lim, J.C., Mitchell, C.H., 2012. Inflammation, pain, and pressure—purinergic signaling Palkar, R., Lippoldt, E.K., McKemy, D.D., 2015. The molecular and cellular basis of
in oral tissues. J. Dent. Res. 91, 1103–1109. thermosensation in mammals. Curr. Opin. Neurobiol. 34, 14–19.
Lingueglia, E., 2007. Acid-sensing ion channels in sensory perception. J. Biol. Chem. Park, C.K., Kim, M.S., Fang, Z., Li, H.Y., Jung, S.J., Choi, S.Y., Lee, S.J., Park, K., Kim,
282, 17325–17329. J.S., Oh, S.B., 2006. Functional expression of thermo-transient receptor potential
Linsuwanont, P., Palamara, J.E.A., Messer, H.H., 2007. An investigation of thermal channels in dental primary afferent neurons: implication for tooth pain. J. Biol.
stimulation in intact teeth. Arch. Oral Biol. 52, 218–227. Chem. 281, 17304–17311.
Liu, X., Yu, L., Wang, Q., Pelletier, J., Fausther, M., Sévigny, J., Malmström, H.S., Dirksen, Pazour, G.J., Witman, G.B., 2003. The vertebrate primary cilium is a sensory organelle.
R.T., Ren, Y.F., 2012. Expression of ecto-ATPase NTPDase2 in human dental pulp. Curr. Opin. Cell Biol. 15, 105–110.
J. Dent. Res. 91, 261–267. Praetorius, H.A., Spring, K.R., 2005. A physiological view of the primary cilium. Annu.
Liu, X., Wang, C., Fujita, T., Malmstrom, H.S., Nedergaard, M., Ren, Y.F., Dirksen, R.T., Rev. Physiol. 67, 515–529.
2015. External dentin stimulation induces ATP release in human teeth. J. Dent. Prasad, R.M., Jin, X., Nauli, S.M., 2014. Sensing a sensor: identifiying the mechanosen-
Res. 94, 1259–1266. sory function of primary cilia. Biosensors 2014 (4), 47–62.
Lumpkin, E.A., Caterina, M.J., 2007. Mechanisms of sensory transduction in the skin. Ranade, S.S., Syeda, R., Patapoutian, A., 2015. Mechanically activated ion channels.
Nature 445, 858–865. Neuron 87, 1162–1179.
Lumpkin, E.A., Marshall, K.L., Nelson, A.M., 2010. The cell biology of touch. J. Cell Biol. Ranade, S.S., Woo, S.H., Dubin, A.E., Moshourab, R.A., Wetzel, C., Petrus, M., Mathur,
191, 237–248. J., Bégay, V., Coste, B., Mainquist, J., Wilson, A.J., Francisco, A.G., Reddy, K., Qiu,
Luukko, K., Suvanto, P., Saarma, M., Thesleff, I., 1997. Expression of GDNF and its Z., Wood, J.N., Lewin, G.R., Patapoutian, A., 2014. Piezo2 is the major transducer
receptors in developing tooth is developmentally regulated and suggests mul- of mechanical forces for touch sensation in mice. Nature 516, 121–125.
tiple roles in innervation and organogenesis. Dev. Dyn. 210, 463–471. Reid, G., 2005. ThermoTRP channels and cold sensing: what are they really up to.
Magloire, H., Couble, M.L., Romeas, A., Bleicher, F., 2004. Odontoblast primary cilia: Pflugers Arch. 451, 250–263.
facts nd hypotheses. Cell Biol. Int. 28, 93–99. Reigada, D., Mitchell, C.H., 2005. Release of ATP from RPE cells involves both CFTR
Magloire, H., Couble, M.L., Thivichon-Prince, B., Maurin, J.C., Bleicher, F., 2009. Odon- and vesicular transport. Am. J. Physiol. Cell Physiol. 288, C132–C140.
toblast: a mechano-sensory cell. J. Exp. Zool. B: Mol. Dev. Evol. 312B, 416–424. Renton, T., Yiangou, Y., Baecker, P.A., Ford, A.P., Anand, P., 2003. Capsaicin receptor
Magloire, H., Lesage, F., Couble, M.L., Lazdunski, M., Bleicher, F., 2003. Expression VR1 and ATP purinoceptor P2 × 3 in painful and nonpainful human tooth pulp.
and localization of TREK-1 K+ channels in human odontoblasts. J. Dent. Res. 82, J. Orofac. Pain 17, 245–250.
542–545. Sabbadinia, M., Yost, C., 2009. Molecular biology of background K channels: insights
Magloire, H., Maurin, J.C., Couble, M.L., Shibukawa, Y., Tsumura, M., Thivichon-Prince, from K2P knockout mice. J. Mol. Biol. 385, 1331–1344.
B., Bleicher, F., 2010. Topical review. Dental pain and odontoblasts: facts and Sato, M., Furuya, T., Kimura, M., Kojima, Y., Tazaki, M., Sato, T., Shibukawa, Y., 2015.
hypotheses. J. Orofac. Pain 24, 335–349. Intercellular odontoblast communication via ATP mediated by pannexin-1 chan-
Magni, G., Merli, D., Verderio, C., Abbracchio, M.P., Ceruti, S., 2015. P2Y2 recep- nel and phospholipase C-coupled receptor activation. Front Physiol. 6, 326.
tor antagonists as anti-allodynic agents in acute and sub-chronic trigeminal Sato, M., Sobhan, U., Tsumura, U., Kuroda, H., Soya, M., Masamura, A., Nishiyama, A.,
sensitization: role of satellite glial cells. Glia 63, 1256–1269. Katakura, A., Ichinohe, T., Tazaki, M., Shibukawa, Y., 2013. Hypotonic-induced
Maingret, F., Fosset, M., Lesage, F., Lazdunski, M., Honoré, E., 1999a. TRAAK is a stretching of plasma membrane activates transient receptor potential vanilloid
mammalian neuronal mechano-gated K+ channel. J. Biol. Chem. 274, 1381–1387. channels and sodium–calcium exchangers in mouse odontoblasts. J. Endod. 39,
Maingret, F., Lauritzen, I., Patel, A.J., Heurteaux, C., Reyes, R., Lesage, F., Lazdunski, 779–787.
M., Honoré, E., 2000. TREK-1 is a heat-activated background K(+) channel. EMBO Sharif-Naeini, R., 2015. Contribution of mechanosensitive ion channels to
J. 19, 2483–2491. somatosensation. Prog. Mol. Biol. Transl. Sci. 131, 53–71.
Maingret, F., Patel, A.J., Lesage, F., Lazdunski, M., Honoré, E., 1999b. Mechano- or Sharma, C.G., Pradeep, A.R., 2006. Gingival crevicular fluid osteopontin levels in
acid stimulation, two interactive modes of activation of the TREK-1 potassium periodontal health and disease. J. Periodontol. 77, 1674–1680.
channel. J. Biol. Chem. 274, 26691–26696. Sherwood, T.W., Frey, E.N., Askwith, C.C., 2012. Structure and activity of the acid-
Matsuka, Y., Neubert, J.K., Maidment, N.T., Spigelman, I., 2001. Concurrent release sensing ion channels. Am. J. Physiol. Cell Physiol. 303, C699–C710.
of ATP and substance P within guinea pig trigeminal ganglia in vivo. Brain Res. Shibukawa, Y., Sato, M., Kimura, M., Sobhan, U., Shimada, M., Nishiyama, A.,
915, 248–255. Kawaguchi, A., Soya, M., Kuroda, H., Katakura, A., Ichinohe, T., Tazaki, M.,
Matsuo, S., Ichikawa, H., Henderson, T.A., Silos-Santiago, I., Barbacid, M., Arends, J.J., 2015. Odontoblasts as sensory receptors: transient receptor potential channels,
Jacquin, M.F., 2001. trkA modulation of developing somatosensory neurons in pannexin-1, and ionotropic ATP receptors mediate intercellular odontoblast-
oro-facial tissues: tooth pulp fibers are absent in trkA knockout mice. Neuro- neuron signal transduction. Pflugers Arch. 467, 843–863.
science 105, 747–760. Shibukawa, Y., Sato, M., Tsumura, M., Kuroda, H., Sobhan, U., Tazaki, M., 2013. Odon-
Maurin, J.C., Couble, M.L., Didier-Bazes, M., Brisson, C., Magloire, H., Bleicher, H., toblast as sensory receptor cell: TRP channels, pannexin 1 and P2 × 3 receptor
2004. Expression and localization of reelin in human odontoblasts. Matrix Biol. coupling mediate sensory transduction in dentin. In: Proceedings of the Tooth,
23, 277–285. Morphogenesis and Differentiation Meeting, La Londe Les Maures, France, May
Maurin, J.C., Couble, M.L., Thivichon-Prince, B., Magloire, H., 2003. Odontoblast: a key 21–26.
cell involved in the perception of dentinal pain. Med. Sci. (Paris) 29, 293–299, Shiozaki, Y., Sato, M., Kimura, M., Sato, T., Tazaki, M., Shibukawa, Y., 2017. Ionotropic
article in French. P2X ATP receptor channels mediate purinergic signaling in mouse odontoblasts.
Maurin, J.C., Delorme, G., Machuca-Gayet, I., Couble, M.L., Magloire, H., Jurdic, P., Front Physiol. 8, 3.
Bleicher, F., 2005. Odontoblast expression of semaphorin 7A during innervation Simon, S., Smith, A.J., Lumley, P.J., Berdal, A., Smith, G., Finney, S., Cooper, P.R.,
of human dentin. Matrix Biol. 24, 232–238. 2009. Molecular characterization of young and mature odontoblasts. Bone 45,
Mayor, R., Theveneau, E., 2013. The neural crest. Development 140, 2247–2251. 693–703.
Mitsiadis, T., Luukko, K., 1995. Neurotrophins in odontogenesis. Int. J. Dev. Biol. 39, Solé-Magdalena, A., Revuelta, E.G., Menénez-Díaz, I., Calavia, M.G., Cobo, T., García-
195–202. Suárez, O., Pérez-Piñera, P., De Carlos, F., Cobo, J., Vega, J.A., 2011. Human
Muhammad, H., Rais, Y., Miosge, N., Ornan, E.M., 2012. The primary cilium as a odontoblasts express transient receptor protein and acid-sensing ion channel
dual sensor of mechanochemical signals in chondrocytes. Cell Mol. Life Sci. 69, mechanosensor proteins. Microsc. Res. Tech. 74, 457–463.
2101–2107. Son, A.R., Yang, Y.M., Hong, J.H., Lee, S.I., Shibukawa, Y., Shin, D.M., 2009. Odontoblast
Murray, P.E., About, I., Lumley, P.J., Franquin, J.C., Windsor, L.J., Smith, A.J., 2003. TRP channels and thermo/mechanical transmission. J. Dent. Res. 88, 1014–1019.
Odontoblast morphology and dental repair. J. Dent. 31, 75–82. Sousa-Valente, J., Andreou, A.P., Urban, L., Nagy, L., 2014. Transient receptor potential
Nieto-Posadas, A., Jara-Oseguera, A., Rosenbaum, T., 2011. TRP channel gating phys- ion channels in primary sensory neurons as targets for novel analgesics. Br. J.
iology. Curr. Top. Med. Chem. 11, 2131–2150. Pharmacol. 171, 2508–2527.
Nilius, B., Owsianik, G., 2011. The transient receptor potential family of ion channels. Staikopoulos, V., Sessle, B.J., Furness, J.B., Jennings, E.A., 2007. Localization of P2 × 2
Genome Biol. 12, 218. and P2 × 3 receptors in rat trigeminal ganglion neurons. Neuroscience 144,
Nilius, B., Szallasi, A., 2014. Transient receptor potential channels as drug targets: 208–216.
from the science of basic research to the art of medicine. Pharmacol. Rev. 66, Story, G.M., Peier, A.M., Reeve, A.J., Eid, S.R., Mosbacher, J., Hricik, T.R., Earley, T.J.,
676–814. Hergarden, A.C., Andersson, D.A., Hwang, S.W., McIntyre, P., Jegla, T., Bevan, S.,
Nishiyama, A., Sato, M., Kimura, M., Katakura, A., Tazaki, M., Shibukawa, Y., 2016. Patapoutian, A., 2003. ANKTM1, a TRP-like channel expressed in nociceptive
Intercellular signal communication among odontoblasts and trigeminal gan- neurons, is activated by cold temperatures. Cell 112, 819–829.
glion neurons via glutamate. Cell Calcium 60, 341–355. Struys, T., De Peralta, T., Vandenabeele, F., Raab, W.H.M., Politis, C., Schepers, S.,
Noël, J., Zimmermann, K., Busserolles, J., Deval, E., Alloui, A., Diochot, S., Guy, N., Vrienlinck, L., Lambrichts, I., 2007. Neuro-odontonlast relationships. Chin. J.
Borsotto, M., Reeh, P., Eschalier, A., Lazdunski, M., 2009. The mechano-activated Dent. Res. 10, 7–13.
K+ channels TRAAK and TREK-1 control both warm and cold perception. EMBO Sukharev, S., Anishkin, A., 2004. Mechanosensitive channels: what can we learn from
J. 28, 1308–1318. ‘simple’ model systems. Trends Neurosci. 27, 345–351.
North, R.A., 2002. Molecular physiology of P2X receptors. Physiol. Rev. 82, Suzuki, H., Iwanaga, T., Yoshie, H., Li, J., Yamabe, K., Yanaihara, N., Langel, U., Maeda,
1013–1067. T., 2002. Expression of galanin receptor-1 (GALR1) in the rat trigeminal ganglia
Okumura, R., Shima, K., Muramatsu, T., Nakagawa, K., Shimono, M., Suzuki, T., and molar teeth. Neurosci. Res. 42, 197–207.
Magloire, H., Shibukawa, Y., 2005. The odontoblast as a sensory receptor cell? Tazawa, K., Ikeda, H., Kawashima, N., Okiji, T., 2017. Transient receptor potential
The expression of TRPV1 (VR-1) channels. Arch. Histol. Cytol. 68, 251–257. melastatin (TRPM) 8 is expressed in freshly isolated native human odontoblasts.
Omerbašić, D., Schuhmacher, L.N., Bernal Sierra, Y.A., Smith, E.S., Lewin, G.R., Arch. Oral Biol. 75, 55–61.
2015. ASICs and mammalian mechanoreceptor function. Neuropharmacology Thivichon-Prince, B., Couble, M.L., Giamarchi, A., Delmas, P., Franco, B., Romio, L.,
94, 80–86. Struys, T., Lambrichts, I., Ressnikoff, D., Magloire, H., Bleicher, F., 2009. Primary
A. Solé-Magdalena et al. / Annals of Anatomy 215 (2018) 20–29 29

cilia of odontoblasts: possible role in molar morphogenesis. J. Dent. Res. 88, von Kugelgen, I., Harden, T.K., 2011. Molecular pharmacology, physiology, and struc-
910–915. ture of the P2Y receptors. Adv. Pharmacol. 61, 373–415.
Tominaga, M., Tominaga, T., 2005. Structure and function of TRPV1. Pflugers Arch. Waldmann, R., Champigny, G., Bassilana, F., Heurteaux, C., Lazdunski, M., 1997. A
451, 143–150. proton-gated cation channel involved in acid-sensing. Nature 386, 173–177.
Tsumura, M., Sobhan, U., Muramatsu, T., Sato, M., Ichikawa, H., Sahara, Y., Tazaki, M., Wang, W., Yi, X., Ren, Y., Xie, Q., 2016. Effects of adenosine triphosphate on prolifer-
Shibukawa, Y., 2012. TRPV1-mediated calcium signal couples with cannabinoid ation and odontoblastic differentiation of human dental pulp cells. J. Endod. 42,
receptors and sodium–calcium exchangers in rat odontoblasts. Cell Calcium 52, 1483–1489.
124–136. Wemmie, J.A., Price, M.P., Welsh, M.J., 2006. Acid-sensing ion channels: advances,
Tsumura, M., Sobhan, U., Sato, M., Shimada, M., Nishiyama, A., Kawaguchi, A., Soya, questions and therapeutic opportunities. Trends Neurosci. 29, 578–586.
M., Kuroda, H., Tazaki, M., Shibukawa, Y., 2013. Functional expression of TRPM8 Wetsel, W.C., 2011. Sensing hot and cold with TRP channels. Int. J. Hyperthermia 27,
and TRPA1 channels in rat odontoblasts. PLoS One 8, e82233. 388–398.
Vandewauw, I., Owsianik, G., Voets, T., 2013. Systematic and quantitative mRNA Wood, J.N., Docherty, R., 1997. Chemical activators of sensory neurons. Annu. Rev.
expression analysis of TRP channel genes at the single trigeminal and dorsal Physiol. 59, 457–482.
root ganglion level in mouse. BMC Neurosci. 14, 21. Xia, J., Lim, J.C., Lu, W., Beckel, J.M., Macarak, E.J., Laties, A.M., Mitchell, C.H., 2012.
Vay, L., Gu, C., McNaughton, P.A., 2012. The thermo-TRP ion channel family: prop- Neurons respond directly to mechanical deformation with pannexin-mediated
erties and therapeutic implications. Br. J. Pharmacol. 165, 787–801. ATP release and autostimulation of P2 × 7 receptors. J. Physiol. 590, 2285–2304.
Viana, F., 2010. Chemosensory properties of the trigeminal system. ACS Chem. Neu- Yeon, K.Y., Chung, G., Shin, M.S., Jung, S.J., Kim, J.S., Oh, S.B., 2009. Adult rat odonto-
rosci. 2, 38–50. blasts lack noxious thermal sensitivity. J. Dent. Res. 88, 328–332.
Viña, E., Parisi, V., Abbate, F., Cabo, R., Guerrera, M.C., Laurà, R., Quirós, L.M., Pérez- Yoshida, S., Ohshima, H., 1996. Distribution and organization of peripheral capillaries
Varela, J.C., Cobo, T., Germanà, A., Vega, J.A., García-Suárez, O., 2015. Acid-sensing in dental pulp and their relationship to odontoblasts. Anat. Rec. 245, 313–326.
ion channel 2 (ASIC2) is selectively localized in the cilia of the non-sensory Yost, C.S., 2003. Update on tandem pore (2P) domain K+ channels. Curr. Drug Targets
olfactory epithelium of adult zebrafish. Histochem. Cell Biol. 143, 59–68. 4, 347–351.
Voets, T., 2012. Quantifying and modeling the temperature-dependent gating of TRP Zha, X.M., 2013. Acid-sensing ion channels: trafficking and synaptic function. Mol.
channels. Rev. Physiol. Biochem. Pharmacol. 162, 91–119. Brain 6, 1.
Volkers, L., Mechioukhi, Y., Coste, B., 2015. Piezo channels: from structure to func- Zimmermann, H., 2006. Ectonucleotidases in the nervous system. Novartis Found.
tion. Pflugers Arch. 467, 95–99. Symp. 276, 113–128.

View publication stats

You might also like