Download as pdf or txt
Download as pdf or txt
You are on page 1of 6

Clinical Research

Expression of Transient Receptor Potential Ankyrin 1


in Human Dental Pulp
Yun Sook Kim, PhD,* Hoon Kap Jung, DDS,* Tae Kyung Kwon, DDS,† Chin Soo Kim, DDS, PhD,*
Jin Hyun Cho, DDS, PhD,* Dong Kuk Ahn, DDS, PhD,* and Yong Chul Bae, DDS, PhD*

Abstract
Introduction: Transient receptor potential ankyrin 1
(TRPA1) is activated by noxious cold (<17 C) and
contributes to cold and mechanical hypersensitivity after
D ental cold hypersensitivity is one of the most frequently suffered symptoms in many
adult dental patients. Yet, our knowledge of the mechanisms of dental cold
nociception and cold hypersensitivity are just now beginning to be elucidated.
inflammation and nerve injury. Methods: To investigate Recently, a family of nonselective cation channels (transient receptor potential
whether TRPA1 is involved in the mediation of nocicep- [TRP] channels) has been shown to render neurons and other cells that express
tion, including noxious cold and cold hypersensitivity in them on their plasma membrane thermosensitive in a fairly narrow and partially over-
teeth, we examined the expression of TRPA1 and sodium lapping temperature range. Different TRP channels also mediate the action of a variety of
channel Nav1.8 in human dental pulp using fluorescent pungent compounds in plant extracts on sensory neurons that is perceived as ‘‘hot’’ (eg,
and electron microscopic immunocytochemistry. Results: capsaicin via the transient receptor potential vanilloid 1) or ‘‘cold’’ (eg, menthol via the
TRPA1 was expressed in a large number of axons branch- transient receptor potential ankyrin 1 [TRPA1]).
ing extensively in the peripheral pulp and in a few axons TRPA1 is expressed primarily in nociceptive neurons and plays a crucial role in the
within the nerve bundles in the core of the coronal pulp perception of noxious cold (1). It also mediates the hypersensitivity to cold after inflam-
and in the radicular pulp. Under electron microscopy, mation and nerve injury (2–4). Recently, it has been shown that neurons in the rat
TRPA1 immunoreactivity was typically localized near trigeminal ganglion (TG) that innervate the dental pulp express messenger RNA for
the plasma membrane of unmyelinated axons in the TRPA1 and that stimulation with noxious cold (<17 C) causes increased intracellular
peripheral pulp, suggesting that in these axons it may calcium and evoked currents in these neurons (5, 6), implicating TRPA1 in the
act as a functional receptor. The proportion of axons ex- mediation of noxious cold within the dental pulp. Large numbers of TRPA1-positive
pressing TRPA1 in neurofilament 200–positive axons (+) axons also terminate in the dorsal part of the trigeminal sensory nuclei that receive
significantly increased in the painful pulp compared intraoral input (7), suggesting that the oral cavity may be densely innervated by TRPA1+
with the normal pulp. TRPA1 was also densely expressed sensory neurons. In addition, a potential role for TRPA1 in human odontoblasts is now
in the processes and the cell body of odontoblasts. A large beginning to be elucidated (8). However, evidence for a TRPA1-mediated nociception in
number of axons coexpressed TRPA1 and Nav1.8. the dental pulp is still lacking.
Conclusions: These findings support the notion that Voltage-gated sodium channels (Navs) play a crucial role in the generation and
TRPA1 is involved in the perception of noxious cold and propagation of action potentials in neurons. Among the 9 subtypes of Navs in humans,
cold hypersensitivity in human dental pulp and that the tetrodotoxin-resistant Nav1.8 is expressed mainly in small-sized nociceptive
TRPA1-mediated nociception is primarily mediated by neurons in the TG (9, 10) and dorsal root ganglia (DRG) (11, 12). Nav1.8-null
axons and odontoblasts in the peripheral pulp. (J Endod mice show a negligible behavioral response to noxious cold (13, 14), and their DRG
2012;38:1087–1092) neurons are also unexcitable in the cold (15), suggesting that Nav1.8 is important
for the generation and propagation of action potentials at low temperatures. Recently,
Key Words it has also been reported that rat TG neurons that innervate the dental pulp express
Cold hypersensitivity, dental pulp, immunohistochem- messenger RNA for both TRPA1 and Nav1.8 and respond to temperatures below
istry, inflammation, Nav1.8, nociception, TRPA1 17 C (5, 6). However, whether Nav1.8 is involved in the transduction of
TRPA1-mediated nociception in human pulp is currently unknown. To address these
issues, we examined the expression of TRPA1 and Nav1.8 in human dental pulp using
fluorescent and electron microscopic immunohistochemistry.
From the *Department of Oral Anatomy and Neurobiology,
School of Dentistry, Kyungpook National University, Daegu,
Korea; and †Mir Dental Hospital, Daegu, Korea.
Supported by Basic Science Research Program through the Materials and Methods
National Research Foundation of Korea funded by the Ministry Tissue Preparation
of Education, Science and Technology (2011-0028240). This study was approved by the Research and Ethics Committee of Kyungpook
Address requests for reprints to Dr Yong Chul Bae, Depart-
ment of Oral Anatomy and Neurobiology, School of Dentistry,
National University, Daegu, Korea, and informed consent was obtained from all human
Kyungpook National University, 188-1, 2-Ga, Samdeok-Dong, subjects who participated in the experimental investigation after the nature of the proce-
Jung-Gu, Daegu 700-412, South Korea. E-mail address: dure had been fully explained. Twelve pulps from healthy premolars extracted during
ycbae@knu.ac.kr the course of orthodontic treatment (males, age range: 17–21 years old) and 3 pulps
0099-2399/$ - see front matter from a painful premolar and molar diagnosed with irreversible pulpitis (males, age
Copyright ª 2012 American Association of Endodontists.
doi:10.1016/j.joen.2012.04.024 range: 18–23 years old) at Kyungpook National University Hospital and Mir Dental
Hospital, Daegu, Korea, were used for this study. Four normal pulps were used for
immunofluorescent staining for TRPA1 and/or Nav1.8, 5 normal pulps and 3 painful

JOE — Volume 38, Number 8, August 2012 Expression of TRPA1 in Human Dental Pulp 1087
Clinical Research
pulps for immunofluorescent staining for TRPA1 and/or neurofilament Data of the normal pulp and the painful pulp with irreversible pulpitis
200 (NF200), and 3 normal pulps for electron microscopic (EM) were compared using an unpaired Student’s t test. Quantitative assess-
immunohistochemistry for TRPA1. Immediately after extraction, teeth ment of colocalization of TRPA1 with Nav1.8 was performed on 3
were stored in 0.1 mol/L phosphate buffer (PB) (pH = 7.4) at 4 C images from each of the 16 sections of 4 healthy coronal pulps. The
for 1 to 4 hours. Then, the teeth were cut longitudinally with a water- omission of the primary or secondary antibodies or preadsorption of
cooled high-speed diamond bur, and the pulps were fixed for 2 hours the TRPA1 antibody with a blocking peptide at a final concentration
in 4% (w/v) paraformaldehyde (PFA) in 0.1 mol/L PB (pH = 7.4) for of 100 mg/mL completely abolished the specific immunostaining
immunofluorescence or in a mixture of 4% PFA and 0.05% glutaralde- (Fig. 1).
hyde for EM immunohistochemistry. For immunofluorescence, the
pulps were cryoprotected in 30% sucrose in PB overnight at 4 C and
EM Immunohistochemistry
cut on a freezing microtome at 40 mm. For EM immunohistochemistry,
the pulps were cut on a vibratome at 60 mm and also cryoprotected in The procedure for pre-embedding immunohistochemistry was
30% sucrose in PB overnight at 4 C. performed as described elsewhere (7, 16). Briefly, cryoprotected
sections were frozen on dry ice for 20 minutes, thawed in PBS, and
treated with 1% sodium borohydride in PBS for 30 minutes. To
suppress endogenous peroxidase, sections were incubated with 3%
Immunofluorescence and Analysis
H2O2 for 10 minutes and blocked with 10% normal donkey serum
for Immunopositive Axons for 30 minutes. After overnight incubation with rabbit anti-TRPA1 anti-
For immunofluorescence, sections were permeabilized with 50% serum (1:1,000, Neuromics), sections were incubated in biotinylated
ethanol for 30 minutes to enhance penetration, blocked with 10% donkey antirabbit antiserum (1:200, Jackson ImmunoResearch) for
normal donkey serum (Jackson ImmunoResearch, West Grove, PA) 2 hours, rinsed, incubated with ExtrAvidin peroxidase (1:5,000,
for 30 minutes to mask secondary antibody binding sites, and incubated Sigma-Aldrich) for 1 hour, and visualized with nickel-intensified diami-
overnight in a mixture of rabbit anti-TRPA1 antiserum (1:1,000, nobenzidine. Sections were then osmicated (1% osmium tetroxide in
RA14135; Neuromics, Edina, MN) and mouse anti-Nav1.8 antiserum PB), dehydrated in graded alcohols, and embedded in Durcupan
(1:100, 75-166; NeuroMab, Davis, CA) or in a mixture of rabbit ACM (Fluka, Buchs, Switzerland). Thin sections were cut, mounted
anti-TRPA1 antiserum and mouse anti-NF200 antiserum (1:200,000, on formvar-coated single-slot nickel grids, counterstained with Sato’s
N0142; Sigma-Aldrich, St Louis, MO) in phosphate-buffered saline lead, and examined on a Hitachi H 7500 electron microscope (Hitachi,
(PBS, 0.01 mol/L, pH = 7.4). After washing with PBS, the sections Tokyo, Japan).
were incubated with appropriate secondary antibodies (fluorescence
isothiocyanate or Cy3-conjugated antibodies raised in donkey, 1:200
in PBS, Jackson ImmunoResearch) for 2 hours. Results
The sections were mounted on slides and examined under Strong immunofluorescent staining for TRPA1 was observed in
a fluorescent microscope (Zeiss Axioplan 2; Carl Zeiss Inc, Jena, axons and axon collaterals in human dental pulp (Fig. 2). A small
Germany) or a laser confocal microscope (LSM 510 META, Carl Zeiss number of axons within nerve bundles wrapped with connective tissue
Inc). The quantitative analysis of TRPA1 expression within were TRPA1 positive (+) in the radicular pulp and in the core of the
NF200-immunopositive axons was performed on 3 to 4 sections from coronal pulp (Fig. 2A and B). In contrast, in the peripheral pulp,
each of the 5 healthy pulps and from each of the 3 painful pulps with TRPA1+ axons branched extensively and formed a network in the
irreversible pulpitis. Three to 4 images from each section were captured vicinity of the cell-rich zone corresponding to the plexus of Raschow
from similar regions in the central and peripheral portions of the in the occlusal and the cervical regions (Fig. 2C and D). TRPA1+ axons
coronal pulp with an Exi digital camera (Q-Imaging Inc, Surrey, CA) ascending toward the dentin between odontoblasts were rarely
using 20 objectives (1,360  1,036 pixels) and analyzed using observed. The proportion of NF200+ axons expressing TRPA1 signifi-
NIH ImageJ software (available at http://rsb.info.nih.gov/ij/). The cantly increased in the painful pulp compared with the normal pulp (P
threshold level for classifying pixels as immunopositive for TRPA1 < .05). Immunoreactivity for TRPA1 was more intense in the painful
and NF200 was set between 100 and 120 gray levels from images pulp than in the normal pulp (Figs. 3 and 4). Immunoreactivity for
with 256 gray levels. The area fraction (%) of immunopositive axons TRPA1 was also observed in the odontoblasts and their processes
(TRPA1/NF200) was presented as the mean  the standard error. (Fig. 5A–D). A large number of TRPA1+ axons were immunostained

Figure 1. Immunofluorescent staining for (A) TRPA1 in normal human dental pulp is completely abolished by (B) preadsorption with a control peptide, proving
the specificity of the TRPA1 antiserum (200, scale bars = 50 mm).

1088 Kim et al. JOE — Volume 38, Number 8, August 2012


Clinical Research

Figure 2. Immunofluorescent staining for TRPA1 in (A) the radicular portion, (B) the core of the coronal portion, and (C and D) the peripheral portion (C,
occlusal region; D, cervical region) of normal human dental pulp. (A and B) Relatively sparse TRPA1+ axons in the nerve bundle issue few axon collaterals in the
radicular pulp and the core of the coronal pulp. (C and D) TRPA1+ axons branch extensively and form a network in the (C) occlusal and (D) cervical regions of the
peripheral pulp (200, scale bars = 50 mm).

for Nav1.8. Of the 450 TRPA1+ axons examined in the coronal pulp in observed near the axolemma of unmyelinated axons in the peripheral
16 sections from 4 pulps, 177 (39.3%) were Nav1.8+ (Fig. 6A–C). pulp (Fig. 7).
At the electron microscopic level, TRPA1 immunoreactivity was
observed in the form of an electron-dense amorphous product in the Discussion
axoplasm of both myelinated and unmyelinated axons in the radicular The main findings of this study are that TRPA1 is densely expressed
pulp and the central portion of the coronal pulp. However, it was usually in the axons and odontoblasts in the peripheral portion of human dental
pulp and the expression of TRPA1 is increased in the pulpal axons after
pulpal inflammation. In addition, approximately 40% of TRPA1+ axons
coexpress the sodium channel Nav1.8.
TRPA1 is selectively expressed by small- and medium-sized, noci-
ceptive neurons in the DRG and TG and has been implicated in the medi-
ation of noxious cold (1, 7, 17) and the cold and mechanical
hypersensitivity after inflammation (18). Recently, Park et al (5)
showed that rat TG neurons innervating the dental pulp express
messenger RNA for TRPA1 and that the application of icilin or noxious
cold to these neurons increases their intracellular calcium and evoked
cationic currents, suggesting that TRPA1 may contribute to dental pain
evoked by noxious cold. The fact that a large number of pulpal axons in
the present study expressed TRPA1 is consistent with a TRPA1-mediated
cold nociception within human dental pulp.
TRPA1 was densely expressed in a large number of axons that
branched extensively outside the connective tissue sheath in the periph-
eral pulp and in a few axons within nerve bundles in the core of the
coronal pulp and in the radicular pulp. Electron microscopy revealed
that TRPA1 is expressed mostly in unmyelinated peripheral axons, in
which it was typically localized at the axonal membrane where it could
act as a functional receptor. In contrast, in myelinated axons in the core
of the coronal pulp and in the radicular pulp, TRPA1 was localized in the
Figure 3. The area fraction (%) of TRPA1+ axons in NF200+ nerve fibers in axoplasm away from the membrane, possibly representing a pool of
the normal and painful (irreversible pulpitis) pulp samples. *A significant receptors that is being transported to the peripheral pulp. These find-
difference between normal and painful pulp samples (P < .05). ings suggest that the TRPA1-mediated sensitivity to noxious cold is

JOE — Volume 38, Number 8, August 2012 Expression of TRPA1 in Human Dental Pulp 1089
Clinical Research

Figure 4. Double immunofluorescent staining for (A and D) TRPA1 and (B and E) NF200 in the peripheral region of (A–C) normal human pulps and (D–F) painful
pulps. Numbers of axons showing TRPA1 expression in NF200+ axons increase in the painful pulp compared with the normal pulp (200, scale bars = 50 mm).

Figure 5. Immunofluorescent staining for TRPA1 in the (A–C) occlusal and (D) cervical regions of normal human dental pulp. Odontoblasts show intense immu-
noreactivity for TRPA1 in their cell bodies and in their processes (arrowheads). An arrow in C points to a TRPA1+ axon beneath the odontoblast layer (400,
scale bars = 50 mm).

1090 Kim et al. JOE — Volume 38, Number 8, August 2012


Clinical Research

Figure 6. Double immunofluorescent staining for (A) TRPA1 and (B) Nav1.8 in normal human dental pulp. (C) Many TRPA1+ axons are Nav1.8-positive
(arrowheads) (200, scale bar = 20 mm).

mediated primarily by axons in the peripheral pulp rather than in the transducing noxious cold information. TRPA1 immunoreactivity was
core of the coronal pulp or in the radicular pulp. In addition, very strong in the processes and in the cell bodies of the odontoblasts.
few axons passing between odontoblasts and entering dentinal tubules Because, as described previously, TRPA1+ axons were rarely seen
were TRPA1 positive, suggesting that axons in the dentinal tubules either ascending toward the dentinal tubule between odontoblasts, we specu-
express another TRP receptor or are insensitive to cold. In the late that noxious cold may be sensed primarily by odontoblast processes
present study, the proportion of axons expressing TRPA1 within rather than by axons in the dentinal tubule.
NF200+ axons significantly increased, suggesting that TRPA1 is involved About 40% of TRPA1+ axons coexpressed Nav1.8 in this study,
in cold and mechanical allodynia and/or hyperalgesia after dental pulp corroborating previous reports that TG neurons that innervate the
inflammation. dental pulp express messenger RNA for both TRPA1 and Nav1.8
Several previous studies have proposed that odontoblasts may act (5, 6) and that DRG neurons of Nav1.8-null mice have significantly
as sensory cells because they express mechanosensitive K+ channels attenuated the expression of TRPA1 (15). Considering that among
(19, 20), L-type Ca2+ channels (21), and thermosensitive TRP channels the 9 subtypes of Nav channels in humans, Nav1.8 can generate
including TRPA1 and the transient receptor potential vanilloid 1 an electrical impulse and is involved in the transmission of nocicep-
(22, 23). They also express voltage-gated sodium channels (24, 25) tive information under cold conditions and plays a crucial role in
and may thus be electrically excitable and able to generate action pain perception at a low temperature (14), our finding supports
potentials (26). TRPA1 is functionally expressed in other non- the notion that Nav1.8 is involved in the conduction of TRPA1-
neuronal cells, including keratinocytes (27), fibroblasts (28), and syn- mediated cold nociception in human dental pulp. The TRPA1+
oviocytes (29), which may act in concert with sensory afferents to axons in the dental pulp that do not express Nav1.8 may express
monitor the thermal environment. other Navs, possibly Nav1.7 or Nav1.9, both of which have been
In agreement with 1 recent report (8), in this study we showed that shown to be overexpressed after pulpal inflammation and have
TRPA1 is expressed by odontoblasts, thus providing morphologic been implicated in the perception of pain after inflammation and
evidence that human odontoblasts may be involved in sensing and nerve injury (30–32).

Figure 7. Electron micrographs showing immunoperoxidase staining for TRPA1 in normal human dental pulp. (A) As shown for an example, the TRPA1 immu-
noreactivity, indicated by patches of an electron-dense reaction product (arrow), is localized in the central regions of the axoplasm of myelinated axons in the
radicular pulp and (B) adjacent to the axonal membrane of unmyelinated axons in the peripheral pulp. Scale bars = 500 nm.

JOE — Volume 38, Number 8, August 2012 Expression of TRPA1 in Human Dental Pulp 1091
Clinical Research
Acknowledgments 15. Abrahamsen B, Zhao J, Asante CO, et al. The cell and molecular basis of mechanical,
cold, and inflammatory pain. Science 2008;321:702–5.
The authors thank Dr Juli Valtschanoff for helpful discussion 16. Kim YS, Kim YJ, Paik SK, et al. Expression of metabotropic glutamate receptor
and careful reading of the manuscript. mGluR5 in human dental pulp. J Endod 2009;35:690–4.
The authors deny any conflicts of interest related to this study. 17. Sawada Y, Hosokawa H, Hori A, et al. Cold sensitivity of recombinant TRPA1 chan-
nels. Brain Res 2007;1160:39–46.
18. Stucky CL, Dubin AE, Jeske NA, et al. Roles of transient receptor potential channels
in pain. Brain Res Rev 2009;60:2–23.
19. Allard B, Couble ML, Magloire H, Bleicher F. Characterization and gene expression
References of high conductance calcium-activated potassium channels displaying mechanosen-
1. Story GM, Peier AM, Reeve AJ, et al. ANKTM1, a TRP-like channel expressed in noci- sitivity in human odontoblasts. J Biol Chem 2000;275:25556–61.
ceptive neurons, is activated by cold temperatures. Cell 2003;112:819–29. 20. Magloire H, Lesage F, Couble ML, et al. Expression and localization of TREK-1 K+
2. Ji G, Zhou S, Carlton SM. Intact Adelta-fibers up-regulate transient receptor potential channels in human odontoblasts. J Dent Res 2003;82:542–5.
A1 and contribute to cold hypersensitivity in neuropathic rats. Neuroscience 2008; 21. Westenbroek RE, Anderson NL, Byers MR. Altered localization of Cav1.2 (L-type)
154:1054–66. calcium channels in nerve fibers, Schwann cells, odontoblasts, and fibroblasts of
3. Karashima Y, Talavera K, Everaerts W, et al. TRPA1 acts as a cold sensor in vitro and tooth pulp after tooth injury. J Neurosci Res 2004;75:371–83.
in vivo. Proc Natl Acad Sci U S A 2009;106:1273–8. 22. Okumura R, Shima K, Muramatsu T, et al. The odontoblast as a sensory receptor
4. Del Camino D, Murphy S, Heiry M, et al. TRPA1 contributes to cold hypersensitivity. cell? The expression of TRPV1 (VR-1) channels. Arch Histol Cytol 2005;68:
J Neurosci 2010;30:15165–74. 251–7.
5. Park CK, Kim MS, Fang Z, et al. Functional expression of thermo-transient receptor 23. Son AR, Yang YM, Hong JH, et al. Odontoblast TRP channels and thermo/mechanical
potential channels in dental primary afferent neurons: implication for tooth pain. transmission. J Dent Res 2009;88:1014–9.
J Biol Chem 2006;281:17304–11. 24. Byers MR, Westenbroek RE. Odontoblasts in developing, mature and ageing rat teeth
6. Kim HY, Chung G, Jo HJ, et al. Characterization of dental nociceptive neurons. J Dent have multiple phenotypes that variably express all nine voltage-gated sodium chan-
Res 2011;90:771–6. nels. Arch Oral Biol 2011;56:1199–220.
7. Kim YS, Son JY, Kim TH, Paik SK, et al. Expression of transient receptor potential 25. Davidson RM. Neural form of voltage-dependent sodium current in human cultured
ankyrin 1 (TRPA1) in the rat trigeminal sensory afferents and spinal dorsal horn. dental pulp cells. Arch Oral Biol 1994;39:613–20.
J Comp Neurol 2010;518:687–98. 26. Allard B, Magloire H, Couble ML, et al. Voltage-gated sodium channels confer excit-
8. El Karim IA, Linden GJ, Curtis TM, et al. Human odontoblasts express functional thermo- ability to human odontoblasts: possible role in tooth pain transmission. J Biol Chem
sensitive TRP channels: implications for dentin sensitivity. Pain 2011;152:2211–23. 2006;281:29002–10.
9. Amaya F, Decosterd I, Samad TA, et al. Diversity of expression of the sensory 27. Atoyan R, Shander D, Botchkareva NV. Non-neuronal expression of transient
neuron-specific TTX-resistant voltage-gated sodium ion channels SNS and SNS2. receptor potential type A1 (TRPA1) in human skin. J Invest Dermatol 2009;129:
Mol Cell Neurosci 2000;15:331–42. 2312–5.
10. Sleeper AA, Cummins TR, Dib-Hajj SD, et al. Changes in expression of two 28. El Karim IA, Linden GJ, Curtis TM, et al. Human dental pulp fibroblasts express the
tetrodotoxin-resistant sodium channels and their currents in dorsal root ganglion ‘‘cold-sensing’’ transient receptor potential channels TRPA1 and TRPM8. J Endod
neurons after sciatic nerve injury but not rhizotomy. J Neurosci 2000;20:7279–89. 2011;37:473–8.
11. Bongenhielm U, Nosrat CA, Nosrat I, et al. Expression of sodium channel SNS/PN3 29. Kochukov MY, McNearney TA, Fu Y, Westlund KN. Thermosensitive TRP ion chan-
and ankyrin(G) mRNAs in the trigeminal ganglion after inferior alveolar nerve injury nels mediate cytosolic calcium response in human synoviocytes. Am J Physiol Cell
in the rat. Exp Neurol 2000;164:384–95. Physiol 2006;291:C424–32.
12. Eriksson J, Jablonski A, Persson AK, et al. Behavioral changes and trigeminal 30. Beneng K, Renton T, Yilmaz Z, et al. Sodium channel Nav1.7 immunoreactivity in
ganglion sodium channel regulation in an orofacial neuropathic pain model. Pain painful human dental pulp and burning mouth syndrome. BMC Neurosci 2010;
2005;119:82–94. 11:71.
13. Akopian AN, Souslova V, England S, et al. The tetrodotoxin-resistant sodium channel 31. Luo S, Perry GM, Levinson SR, Henry MA. Nav1.7 expression is increased in painful
SNS has a specialized function in pain pathways. Nat Neurosci 1999;2:541–8. human dental pulp. Mol Pain 2008;4:16.
14. Zimmermann K, Leffler A, Babes A, et al. Sensory neuron sodium channel Nav1.8 is 32. Wells JE, Bingham V, Rowland KC, Hatton J. Expression of Nav1.9 channels in human
essential for pain at low temperatures. Nature 2007;447:855–8. dental pulp and trigeminal ganglion. J Endod 2007;33:1172–6.

1092 Kim et al. JOE — Volume 38, Number 8, August 2012

You might also like