Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

Acta Physiol 2012, 204, 158–168

REVIEW
Taste receptor signalling – from tongues to lungs

S. C. Kinnamon
Department of Otolaryngology and Rocky Mountain Taste and Smell Center, School of Medicine, University of Colorado Denver, Aurora,
CO, USA

Received 25 February 2011, Abstract


revision requested 18 March 2011, Taste buds are the transducing endorgans of gustation. Each taste bud
revision received 28 March 2011,
comprises 50–100 elongated cells, which extend from the basal lamina to the
accepted 6 April 2011
Correspondence: S. C. Kinnamon, surface of the tongue, where their apical microvilli encounter taste stimuli in
Department of Otolaryngology, the oral cavity. Salts and acids utilize apically located ion channels for
School of Medicine, University of transduction, while bitter, sweet and umami (glutamate) stimuli utilize
Colorado Denver, 12,700 East G-protein-coupled receptors (GPCRs) and second-messenger signalling
19th Avenue, Mailstop 8606,
mechanisms. This review will focus on GPCR signalling mechanisms. Two
Aurora, CO 80534, USA.
E-mail:
classes of taste GPCRs have been identified, the T1Rs for sweet and umami
sue.kinnamon@ucdenver.edu (glutamate) stimuli and the T2Rs for bitter stimuli. These low affinity GPCRs
all couple to the same downstream signalling effectors that include Gbc
activation of phospholipase Cb2, 1,4,5-inositol trisphosphate mediated
release of Ca2+ from intracellular stores and Ca2+-dependent activation of the
monovalent selective cation channel, TrpM5. These events lead to membrane
depolarization, action potentials and release of ATP as a transmitter to
activate gustatory afferents. The Ga subunit, a-gustducin, activates a phos-
phodiesterase to decrease intracellular cAMP levels, although the precise
targets of cAMP have not been identified. With the molecular identification
of the taste GPCRs, it has become clear that taste signalling is not limited to
taste buds, but occurs in many cell types of the airways. These include sol-
itary chemosensory cells, ciliated epithelial cells and smooth muscle cells.
Bitter receptors are most abundantly expressed in the airways, where they
respond to irritating chemicals and promote protective airway reflexes, uti-
lizing the same downstream signalling effectors as taste cells.
Keywords G-protein-coupled receptor signalling, gustducin, purinergic
signalling, solitary chemosensory cell, taste cell, TrpM5.

Taste buds are the transducing elements of gustatory Na+; and umami, for detection of l-amino acids – each
sensation. Housed in connective papillae on the tongue required by the body for energy balance, ionic homeo-
and scattered throughout the epithelium of the soft stasis or building proteins. The aversive qualities are
palate and larynx, these onion-shaped endorgans detect sour, which detects acids in unripe fruit and spoiled
nutrients in foods and guard against ingestion of toxic foods, and bitter, which detects a variety of plant
substances and spoiled foods. Unlike the olfactory alkaloids, many of which are toxic. Thus, detection of
system, which can discriminate thousands of individual aversive tastes guards the entrance of the alimentary
chemicals, the gustatory system can discriminate only canal against the ingestion of potential toxins.
five basic taste qualities – sweet, sour, salty, bitter and Because taste stimuli represent both ionic and com-
umami (the taste of glutamate and other l-amino acids). plex compounds, different mechanisms have evolved for
The appetitive taste qualities are sweet, for detection of their detection. Salts and acids are detected primarily by
sugars and sweeteners; salty, primarily for detection of apically located ion channels, while chemicals that elicit

 2011 The Author


158 Acta Physiologica  2011 Scandinavian Physiological Society, doi: 10.1111/j.1748-1716.2011.02308.x
17481716, 2012, 2, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/j.1748-1716.2011.02308.x by U. Miguel Hernandez De Elche, Wiley Online Library on [06/11/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Acta Physiol 2012, 204, 158–168 S C Kinnamon Æ Taste GPCR signalling
bitter, sweet and umami tastes are detected by (a)
G-protein-coupled receptors (GPCRs) and second-
messenger signalling pathways. In keeping with the
theme of the symposium, this review will focus on
GPCR signalling mechanisms. With the molecular
identification of the taste receptors and their signalling
effectors, it is now clear that taste receptor signalling
is not limited to taste buds but occurs in a variety
of tissues, including chemosensory cells of the alimen-
tary tract (see Iwatsuki et al., this volume), pancreas
(Nakagawa et al. 2009), brain (Ren et al. 2009, Singh
et al. 2011) and several cell types of the airway epithelium
(Finger et al. 2003, Shah et al. 2009, Deshpande et al.
2010, Tizzano et al. 2010). In all cases, these receptors
detect similar compounds using similar signalling effec-
tors, but elicit very different effects in the different
tissues. This review will focus on taste receptor signalling (b)
in taste buds and airway epithelia, where these processes
have been most extensively studied.

Taste buds, cell types and innervation


Taste buds are onion-shaped aggregates of approxi-
mately 50–100 elongate cells that extend from the basal
lamina to the surface of the tongue, where their apical
microvilli extend through an opening in the epithelium
to contact sapid chemicals in the oral cavity. Each taste
bud contains three types of elongate taste cells and a
renewing population of basal cells (Fig. 1). Type II or
‘receptor’ cells are the focus of this review, as they
contain the GPCRs and downstream signalling effectors
for bitter, sweet and umami taste stimuli (Clapp et al. Figure 1 (a) Diagrammatic illustration of a taste bud, showing
2004, DeFazio et al. 2006). Interestingly, Type II cells the three types of taste cells and a renewing population of basal
lack voltage-gated Ca2+ channels and other presynaptic (B) cells. Type II cells contain the GPCRs signaling effectors for
specializations, although they associate closely with bitter, sweet and umami stimuli, and are the focus of this
afferent nerve processes (Clapp et al. 2006). Recent review. Type I cells are generally considered to have a support
data have shown that these cells release ATP as a function, while Type III cells respond to sour stimuli and form
transmitter, which activates P2X receptors on afferent prominent synapses with afferent nerve fibres. Type II cells also
associate closely with afferent nerve fibres, but do not form
nerve fibres (Finger et al. 2005) and P2Y receptors on
conventional synapses. (b) Image of two taste buds, showing
adjacent taste cells (Hayato et al. 2007, Huang et al.
Type II cells stained with an antibody against the Type III IP3
2009). Type I or ‘glial-like’ cells are believed to play receptor and nerve fibres stained with an antibody against the
primarily a support function in the taste bud, as their purinergic receptor P2X2. Apical staining of P2X2 may rep-
membranes wrap around other cells in a glial-like resent non-specific binding. (Image courtesy of A. Montoya
fashion and they express enzymes for the degradation of and J. Kinnamon, University of Denver).
the ATP released from the Type II cells (Bartel et al.
2006). Type I cells also express the epithelial Na+ release of ATP from Type II cells (Huang et al. 2009),
channel (Vandenbeuch et al. 2008), so they may also but the role of the other transmitters is not yet clear. In
play a role in the transduction of salt taste. Type III addition to modulating adjacent taste cells, Type III
or ‘presynaptic’ cells transduce sour (acidic) stimuli cells are the only cells in the taste bud to make
(Huang et al. 2006, 2008b) and also respond to ATP conventional synapses with sensory afferents (Murray
released from receptor cells (Huang et al. 2009). These & Murray 1971, Royer & Kinnamon 1991). However,
cells contain and release several transmitters, including the role of these transmitters in activating nerve fibres is
serotonin (Huang et al. 2005), noradrenaline (Huang unclear, since the double knockout of P2X2 and P2X3
et al. 2008a) and possibly GABA (DeFazio et al. 2006, eliminates all taste-evoked neural activity (Finger et al.
Starostik et al. 2010). Serotonin appears to modulate 2005).

 2011 The Author


Acta Physiologica  2011 Scandinavian Physiological Society, doi: 10.1111/j.1748-1716.2011.02308.x 159
17481716, 2012, 2, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/j.1748-1716.2011.02308.x by U. Miguel Hernandez De Elche, Wiley Online Library on [06/11/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Taste GPCR signalling Æ S C Kinnamon Acta Physiol 2012, 204, 158–168

Taste buds on the anterior two-thirds of the tongue inosine-5-monophosphate (IMP) and guanosine-
are housed in fungiform papillae, which in rodents 5-monophosphate (GMP), which bind in an allosteric
contain one to two taste buds each and are innervated fashion to the T1R1 venus fly trap domain and stabilize
by the chorda tympani branch of the facial nerve. Taste the closed (active) state of the receptor (Zhang et al.
buds on the posterior tongue are housed in foliate and 2008). The sweet receptor T1R2 + T1R3 binds sugars,
circumvallate papillae, each of which contains hundreds d-amino acids, synthetic sweeteners and some sweet
of taste buds that are innervated primarily by the proteins – basically all compounds that are recognized
glossopharyngeal nerve. Taste buds on the soft palate as sweet (Nelson et al. 2001). Although most ligand
are innervated by another branch of the lingual nerve, binding for small sweeteners occurs in the N-terminal
the greater superficial petrosal nerve, while taste buds domain of T1R2, some binding, particularly for large
on the epiglottis and larynx are innervated by superior sweet proteins, also occurs in regions of T1R3 (Jiang
laryngeal nerve, a branch of the vagus nerve. Despite et al. 2004, 2005, Nie et al. 2005). None of the T1R
differences in their location and innervation, the struc- binding domains has been successfully crystallized, so
ture of taste buds in different regions of the oral cavity is structural details of ligand binding remain to be
remarkably conserved. In addition, despite what is often elucidated. T1R3 null mice have been generated in
published in textbooks, there is no distinct map of two different labs, with differing results. In the Zuker
chemosensitivity on the tongue, although variations in lab, genetic elimination of T1R3 abolished responses to
thresholds exist in the different taste fields. virtually all sweet and umami compounds, suggesting
that T1R1 + T1R3 and T1R2 + T1R3 are the only
receptors to mediate these qualities (Zhao et al. 2003).
G-protein-coupled taste receptors
In contrast, the Margolskee lab found that while T1R3
Two classes of taste GPCRs have been identified knockout mice fail to respond to most sweet stimuli,
molecularly, T1Rs and T2Rs. T1Rs mediate sweet and considerable responses remain to umami compounds,
umami (glutamate) taste, while T2Rs mediate bitter suggesting the existence of multiple receptors for umami
taste. In general, taste receptors are low affinity recep- stimuli (Damak et al. 2003, Delay et al. 2006, Maruy-
tors compared with other GPCRs, with binding affin- ama et al. 2006, Yoshida et al. 2009b). Candidate
ities in the high lm–mm range. These concentrations, receptors are taste-specific isoforms of two metabotropic
however, are similar to the concentration of most glutamate receptors, mGluR1 (San Gabriel et al. 2009a)
nutrients in foods. Because of their low affinity, the taste and mGlurR4 (Chaudhari et al. 1996). However,
GPCRs were all cloned by mapping the human and knockouts will be required to verify a role in taste
mouse genomes, using information obtained from the transduction.
linkage analysis of taste polymorphisms. A detailed
discussion of the genetic origins of these taste receptors
T2Rs
is reviewed in Bachmanov & Beauchamp (2007).
Bitter taste is mediated by the T2R GPCRs, products of
the Tas2R gene family. T2Rs are classical ‘A’ type
T1Rs
receptors that are similar in structure to the opsins and
T1Rs are classical ‘C’ type receptors, with large the olfactory receptors (Adler et al. 2000, Chandrashe-
N-terminal ligand binding domains that exhibit a venus kar et al. 2000). They have short N-terminal domains,
fly trap ligand binding module similar to the metabo- with ligand binding in the extracellular loops and
tropic glutamate receptors, the GABAB receptor and the transmembrane domains. Recent data suggest that
calcium sensing receptor. Three different T1Rs have carboxy terminal regions are particularly important
been identified, which are products of the Tas1R genes: for agonist selectivity (Brockhoff et al. 2010). The
T1R1, T1R2 and T1R3 (Max et al. 2001, Montmayeur family consists of about 30 members in mammals, each
et al. 2001, Nelson et al. 2001, 2002, Sainz et al. of which binds structurally similar bitter compounds.
2001). These receptors are functional only as heterodi- While some receptors are rather narrowly tuned, others
mers, with T1R3 serving as an obligate partner for both are broadly tuned and respond to many bitter com-
the umami receptor (T1R1 + T1R3) and the sweet pounds (Meyerhof et al. 2010). These receptors have
receptor (T1R2 + T1R3). In heterologous systems, the been considered to function as monomers, but recent
umami receptor responds broadly to all l-amino acids data suggest that they can also form functional oligo-
in rodents (Nelson et al. 2002), but only to l-glutamate mers (Kuhn et al. 2010). Although the role of oligo-
in humans (Li et al. 2002). Ligand binding occurs in the merization in T2R function has not been clearly
N-terminal domain of the T1R1 monomer. Impor- elucidated, literally thousands of structurally diverse
tantly, the umami receptor in both rodents and humans molecules taste bitter and oligomers would greatly
is strongly potentiated by 5¢-ribonucleotides such as increase the repertoire of stimuli that can activate a

 2011 The Author


160 Acta Physiologica  2011 Scandinavian Physiological Society, doi: 10.1111/j.1748-1716.2011.02308.x
17481716, 2012, 2, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/j.1748-1716.2011.02308.x by U. Miguel Hernandez De Elche, Wiley Online Library on [06/11/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Acta Physiol 2012, 204, 158–168 S C Kinnamon Æ Taste GPCR signalling
given receptor. Molecular studies have shown that of sweet, salty and umami tastes (Ohsu et al. 2010).
bitter responsive taste cells express most or all of the Thus, CaSR and the fatty acid GPCRs appear to
receptors, which is not surprising since bitter taste modulate tastes rather than elicit a specific taste quality,
evolved to avoid ingestion of toxic substances, and and both appear to selectively enhance the appetitive
bitter compounds are not readily distinguished psycho- taste qualities.
physically in humans (Adler et al. 2000). However,
physiological (Caicedo & Roper 2001) and molecular
Downstream signalling effectors
(Matsunami et al. 2000, Behrens et al. 2007) studies of
bitter responsive taste cells have shown that not all taste
Gbc-mediated signalling
cells express all bitter receptors, suggesting that some
bitter compounds could be discriminated. In general, T2Rs and T1Rs, as well as T1R1 and T1R2
are expressed in largely non-overlapping subsets of
Type II taste cells, suggesting that these qualities can all
Other taste GPCRs
be distinguished. However, T1Rs and T2Rs generally
Although T1Rs and T2Rs are generally considered to be activate the same downstream signalling effectors in
the primary taste receptors, other GPCRs have been Type II taste cells (Fig. 2) (Zhang et al. 2003). Taste
identified in taste buds and likely contribute to detection receptor binding leads to activation of a heterotrimeric
of nutrients, even though they do not appear to mediate G protein, which consists in most cells of Ga-gustducin
a distinct taste quality. These include GPR40 and (a-gust) (McLaughlin et al. 1992) and its bc partners,
GPR120, both of which are expressed in subsets of b3c13 (Huang et al. 1999). The dominant leg of the
taste cells and detect medium and long chain fatty acids pathway is mediated by the bc partners, which activate
(Cartoni et al. 2010). Another GPCR that is expressed phospholipase Cb2 (PLCb2) (Rossler et al. 1998) to
abundantly in taste buds is the calcium sensing receptor, convert the membrane lipid PIP2 into the second
CaSR (San Gabriel et al. 2009b, Bystrova et al. 2010). messengers 1,4,5-inositol trisphosphate (IP3) and diacyl-
Recent studies in humans have shown that agonists of glycerol (DAG). While the function of DAG remains
the CaSR, including calcium and glutathione, elicit the unclear, IP3 binds to the Type III IP3 receptor (IP3R3)
so-called ‘kokumi’ taste, which results in a potentiation (Clapp et al. 2001, Miyoshi et al. 2001), causing release

(a)

(b)

Figure 2 Diagrammatic representation of taste GPCRs (a) and downstream signaling effectors (b). Receptor binding leads to Gbc
activation of PLCb2, production of IP3, release of Ca2+ from intracellular stores, Ca2+-dependent activation of TrpM5, depolar-
ization, activation of voltage-gated Na+ channels (VGNC) and release of ATP through pannexin-1 hemichannels. The released ATP
activates purinergic receptors on afferent nerve fibres. Alpha gustducin tonically regulates cAMP levels via activation of a phos-
phodiesterase (PDE), which subsequently prevents phosphorylation and desensitization of Ca2+ signalling effectors.

 2011 The Author


Acta Physiologica  2011 Scandinavian Physiological Society, doi: 10.1111/j.1748-1716.2011.02308.x 161
17481716, 2012, 2, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/j.1748-1716.2011.02308.x by U. Miguel Hernandez De Elche, Wiley Online Library on [06/11/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Taste GPCR signalling Æ S C Kinnamon Acta Physiol 2012, 204, 158–168

of Ca2+ from intracellular stores and subsequent Ca2+- a-transducin are expected to activate a phosphodiester-
dependent activation of a monovalent selective cation ase (PDE) and decrease intracellular cAMP levels.
channel, transient receptor potential channel M5 Biochemical studies have shown that bitter stimuli do
(TrpM5) (Perez et al. 2002, Zhang et al. 2007). This decrease intracellular cAMP levels, and the decrease is
leads to membrane depolarization, action potential inhibited by antibodies to a-gust (Yan et al. 2001).
generation (Vandenbeuch & Kinnamon 2009, Yoshida Cyclic AMP is also decreased in taste tissue in response
et al. 2009a) and release of ATP through gap junction to umami stimuli (Abaffy et al. 2003). However, many
hemichannels, likely composed of pannexin-1 (Huang studies have shown that sugars increase cAMP levels in
et al. 2007, Romanov et al. 2007, Dando & Roper taste tissue (Bernhardt et al. 1996) and the increase is
2009, Murata et al. 2010). Recent evidence suggests not simply a secondary consequence of Gbc-mediated
that Type II taste cells also express the vesicular ATP release of Ca2+ from intracellular stores (Trubey et al.
transporter, VNUT. This leaves open the possibility that 2006). Gustducin knockout mice are significantly com-
ATP may also be released in a vesicular manner promised to bitter, sweet and umami stimuli, but the
(Iwatsuki et al. 2009). Evidence for this signalling effect for sweet is much less than for bitter and umami
pathway comes from immunocytochemical and molec- (Wong et al. 1996, Ruiz et al. 2003, Glendinning et al.
ular studies showing that the component signalling 2005). Thus, the role of a-gust in sweet taste is much
effectors are co-expressed in both bitter and sweet/ less clear than for bitter and umami taste. Part of the
umami responsive Type II taste cells (Clapp et al. 2001, lack of effect of the gustducin knockout on sweet taste
2004, DeFazio et al. 2006). Further, stimulation of is that the sweet receptor T1R3 is not generally
isolated Type II taste cells with bitter, sweet or umami co-expressed with a-gust in posterior tongue. Instead,
taste stimuli elicits increases in intracellular Ca2+ that T1R3 is usually co-expressed with a Gq family protein,
do not require extracellular Ca2+, are blocked by the Ga14 in circumvallate and foliate taste buds (Shindo
PLC inhibitor U73122, and are sensitive to thapsigar- et al. 2008, Tizzano et al. 2008). Whether Ga14 medi-
gin, which inhibits the Ca2+ ATPase that refills intra- ates sweet transduction in these taste fields awaits
cellular Ca2+ stores (Ogura et al. 2002, Hacker et al. studies in Ga14 knockout mice.
2008). Finally, knockout of PLCb2, TrpM5 or IP3R3 What is the role of the decreased cAMP in taste
strongly reduces or eliminates afferent nerve responses signalling? Although molecular evidence has indicated
to most bitter, sweet and umami taste stimuli (Zhang expression of a cyclic nucleotide-gated channel in taste
et al. 2003, Damak et al. 2006, Hisatsune et al. 2007). buds (Misaka et al. 1997), there is no physiological
The apparent role of TrpM5 in this process is to evidence for cyclic nucleotide-gated currents in taste
translate the Ca2+ that is released from the intracellular cells. To determine other possible functions of the
stores into a membrane depolarization that is sufficient gustducin-mediated decrease in cAMP, biochemical
to activate voltage-gated Na+ channels and elicit action assays were performed on isolated circumvallate taste
potentials, which may be required to open the pannexin buds of gustducin knockout mice. Interestingly, the
hemichannels. In support of this hypothesis, knockout knockout mice were found to have highly elevated levels
of TrpM5 or inhibition of voltage-gated Na+ channels of cAMP relative to wild-type mice (Clapp et al. 2008).
with tetrodotoxin inhibits ATP release (Huang & Roper These levels were elevated in the absence of any taste
2010, Murata et al. 2010). Further, loose patch record- stimuli, suggesting that the taste receptors and/or G
ings from identified Type II taste cells during taste protein have tonic activity in the absence of taste
stimulation showed that ATP release was proportional ligands. The elevated cAMP likely activates protein
to the frequency of action potentials elicited by the taste kinase A to phosphorylate and inhibit PLC signalling
stimulus (Murata et al. 2010). effectors, since H-89, a specific protein kinase A
inhibitor, rescued responses to bitter stimuli in the taste
cells of gustducin knockout mice (Clapp et al. 2008).
Ga-mediated signalling
These data suggest that gustducin tonically regulates
All T2R receptors, and the T1R receptors in the anterior cAMP levels in taste cells to keep phosphorylation levels
tongue and palate, are co-expressed with the Ga low and prevent chronic adaptation to bitter taste
subunit, gustducin (Adler et al. 2000, Kim et al. 2003, stimuli. Whether a-gust plays a similar role in the
Stone et al. 2007). Ga-gustducin was the first protein to transduction of umami and sweet taste has not been
be molecularly identified in taste cells (McLaughlin determined.
et al. 1992), but its role in taste signal transduction is
still not completely understood. Gustducin has consid-
Taste receptor signalling in the airways
erable sequence homology to transducin, which is also
expressed in taste buds (McLaughlin et al. 1994). The cloning of taste signalling effectors and the
By analogy to the visual system, both a-gust and production of several lines of transgenic mice expressing

 2011 The Author


162 Acta Physiologica  2011 Scandinavian Physiological Society, doi: 10.1111/j.1748-1716.2011.02308.x
17481716, 2012, 2, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/j.1748-1716.2011.02308.x by U. Miguel Hernandez De Elche, Wiley Online Library on [06/11/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Acta Physiol 2012, 204, 158–168 S C Kinnamon Æ Taste GPCR signalling
green fluorescent protein (GFP) from their promoters, In rodents, SCCs are found in the upper airways,
particularly a-gust, TrpM5 and T1R3 has revealed an scattered in the nasal respiratory epithelium (Finger
extensive expression in the airways, from the upper et al. 2003, Tizzano et al. 2010), at the entrance of the
airways to the lungs. T2R signalling has been most vomeronasal duct (Ogura et al. 2010) and on the larynx
extensively studied, but T1Rs are also expressed in a (Tizzano et al. 2011). In the lower airways, they are
number of regions of the airways. The cell types found in the trachea, the bronchi and larger bronchi-
expressing these signalling effectors include solitary oles, but not in the smaller bronchioles or the alveoli
chemosensory (also called chemoreceptor) cells (SCCs) (Tizzano et al. 2010). In the respiratory epithelium and
(Finger et al. 2003, Sbarbati et al. 2004, Kaske et al. the larynx, the SCCs are heavily innervated by pepti-
2007, Lin et al. 2008, Ogura et al. 2010, Tizzano et al. dergic fibres of the trigeminal and vagus nerves respec-
2010, 2011), ciliated epithelial cells (Shah et al. 2009), tively. Unlike Type II taste receptor cells that lack
and most recently, smooth muscle cells lining the conventional synapses with sensory afferents (Clapp
airways (Deshpande et al. 2010). et al. 2004, 2006), the SCCs innervated by peptidergic
fibres appear to have classical synapses with pre- and
postsynaptic specializations (Finger et al. 2003). A
Solitary chemosensory cells
recent study suggests that a subset of SCCs expresses
Solitary chemosensory cells were first described in cholinergic markers and contacts nerve fibres expressing
aquatic vertebrates (Kotrschal & Whitear 1988, White- nicotinic acetylcholine receptors (Krasteva et al. 2010),
ar 1992), where they are extensively expressed on the suggesting acetylcholine may be the transmitter. In
gill arches and skin. Each SCC has an apical process contrast to the upper airways, SCCs in the trachea and
with microvilli that reaches the surface of the epithelium lower airways are much more sparsely innervated,
and a basolateral membrane that is densely supplied suggesting these SCCs may mediate different functions
with nerve endings, suggesting a sensory function. The from the SCCs in the upper airway epithelia.
morphology of SCCs resembles that of taste receptor The SCCs of the nasal respiratory epithelium have
cells, but unlike taste cells, which are aggregated in taste been studied most extensively. These cells express both
buds, SCCs are scattered singly in the epithelium T2Rs and T1Rs, but T1Rs are much less abundantly
(Fig. 3). In air breathing vertebrates, SCCs are found expressed than T2Rs. Unlike taste cells, where T1Rs
scattered in the airway epithelium, suggesting a respi- and T2Rs are expressed in non-overlapping subsets of
ratory function. Although SCCs have not been exten- cells, SCCs in the airways often express both, suggesting
sively studied in non-mammalian terrestrial vertebrates, different classes of compounds (i.e. sweet and bitter)
SCCs have been described in the airways of alligators, may elicit similar effects in the airways (Ohmoto et al.
where they are scattered among the olfactory receptor 2008, Tizzano et al. 2011). The downstream signalling
cells (Hansen 2007). SCCs appear to be restricted to the effectors are identical to taste receptor cells, with Gbc
vertebrate lineage, as none have been identified in stimulation of PLCb2, production of IP3, release of Ca2+
invertebrate species (Finger 2006). from intracellular stores, activation of TrpM5, depo-
larization and release of transmitter. The only difference
is that SCCs appear to use conventional vesicular
synaptic transmission to activate the afferent nerves,
while taste cells release ATP via non-vesicular mecha-
nisms. Solitary chemosensory cells have been isolated
from a-gust-GFP mice and TrpM5-GFP mice and
studied with Ca2+ imaging. GFP-labelled SCCs respond
rather selectively to the bitter stimulus denatonium,
suggesting these cells are more narrowly tuned than
bitter responsive taste cells, which respond to a variety
of bitter compounds. As in taste cells, the response is
inhibited by the PLC inhibitor U73122 (Gulbransen
et al. 2008). In addition to denatonium, SCCs also
respond to homoserine lactones (HSLs) that are pro-
duced by Gram-negative pathogenic bacteria as quorum
Figure 3 Laser scanning confocal image of two TrpM5-GFP-
labelled solitary chemosensory cells of the mouse nasal epi- signalling molecules. When the concentration of these
thelium. Nerve endings are stained with an antibody against molecules reaches a critical level, the bacteria become
substance P, a transmitter expressed in peptidergic trigeminal pathogenic and form a biofilm, attacking the epithe-
nerve fibres. (Image courtesy of M. Tizzano, University of lium. Solitary chemosensory cells detect the HSLs at the
Colorado Denver). appropriate concentration and activate the trigeminal

 2011 The Author


Acta Physiologica  2011 Scandinavian Physiological Society, doi: 10.1111/j.1748-1716.2011.02308.x 163
17481716, 2012, 2, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/j.1748-1716.2011.02308.x by U. Miguel Hernandez De Elche, Wiley Online Library on [06/11/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Taste GPCR signalling Æ S C Kinnamon Acta Physiol 2012, 204, 158–168

nerve to produce a pronounced apnea that protects the represents a species difference, or possibly an effect of
airway from further inhalation. These responses are cell culture conditions. It will be important to document
absent in both TrpM5 and a-gust knockout mice, the presence of these receptors on freshly isolated
suggesting that activation of the trigeminal nerve by human airway epithelia.
HSLs requires the integrity of the SCCs (Tizzano et al.
2010). Important questions that remain are the nature
Airway smooth muscle cells
of the stimuli that activate the T1Rs and the physio-
logical role of the SCCs of the lower airways. As most of T2R signalling has recently been reported on cultured
the lower airway SCCs are not innervated, activation of human smooth muscle cells that line the airway. Several
these cells likely results in secretory functions or T2Rs were expressed in the muscle cells, along with
mucociliary clearance mechanisms, rather than protec- a-gust (Deshpande et al. 2010). Stimulation with a
tive nerve reflexes. variety of bitter compounds caused an increase in
intracellular Ca2+, which was blocked or reduced by
pharmacological inhibitors of Gbc, PLCb2 and IP3
Ciliated epithelial cells
Interestingly, stimulation with bitter compounds caused
Cultured human airway epithelial cells have been a potent relaxation of the smooth muscle. This was
shown to express T2Rs and some of their downstream unexpected, since acetylcholine, which causes an
signalling effectors (Shah et al. 2009). In this case, the increase in intracellular Ca2+ via activation of Gaq,
receptors are found on the ciliated cells rather than on evokes muscle contraction. The authors found that the
the SCCs described above. Several T2Rs and a-gust are bitter taste-mediated increase in intracellular Ca2+ was
expressed on the motile cilia, with PLCb2 expressed accompanied by a Ca2+-dependent increase in K+
beneath the tight junctions. The cultured airway cells conductance mediated by the BK channel, which
responded to several bitter compounds with increases in resulted in membrane hyperpolarization and relaxation.
intracellular Ca2+ and a concomitant increase in the This suggests that the T2R signalling cascade is in a
frequency of ciliary beating. Unlike the SCCs, TrpM5 membrane compartment that is distinct from the
does not appear to be expressed, which is not surprising acetylcholine receptor signalling cascade. The authors
since epithelial cells are not generally electrically excit- suggest that inhaled bitter compounds could be used in
able. The authors suggest that the ciliated cells detect a therapeutic manner to treat airway diseases, such as
noxious inhaled substances or products of bacterial asthma and chronic pulmonary obstructive disease. One
infection and increase ciliary beating to remove the concern, however, is how these compounds pass from
harmful substances from the airways. It is of interest the airway epithelium to the smooth muscle to mediate
that subsequent studies using immunocytochemical the effects. Again, as was the case with ciliated epithelial
probes and transgene expression have failed to find cells discussed above, recent studies have failed to
evidence of T2R signalling on ciliated airway cells of document expression of T2R downstream signalling
rodents (Tizzano et al. 2011). It is possible that this effectors in smooth muscle of the rodent airway

Figure 4 Diagrammatic illustration of differences in signalling effectors in taste cells, SCCs of the airway, ciliated epithelial
cells and smooth muscle cells lining the airways. In all cases taste GPCRs activate the downstream PLC signalling effectors, but the
effects of increased Ca2+ differ among the different cell types.

 2011 The Author


164 Acta Physiologica  2011 Scandinavian Physiological Society, doi: 10.1111/j.1748-1716.2011.02308.x
17481716, 2012, 2, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/j.1748-1716.2011.02308.x by U. Miguel Hernandez De Elche, Wiley Online Library on [06/11/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Acta Physiol 2012, 204, 158–168 S C Kinnamon Æ Taste GPCR signalling
(Tizzano et al. 2011). It will be important to document The author thanks Dr John Kinnamon for the unpublished
that these signalling effectors are present in vivo. image in Figure 1 and Dr Marco Tizzano for the unpublished
image in Figure 3. Drs Aurelie Vandenbeuch and Thomas
Finger provided helpful comments on the manuscript.
Summary and future directions
This work was supported in part by NIH grants DC00766,
Taste receptor signalling is remarkably conserved in DC006021, DC007495, DC009820 and P30DC004657.
tissues ranging from taste receptor cells to several cell
types in the airways and several cell types in the
References
gastrointestinal epithelium (see review by Iwatsuki
et al. this volume). In all cases, the stimuli bind to Abaffy, T., Trubey, K.R. & Chaudhari, N. 2003. Adenylyl
low affinity receptors that activate Gbc, causing stim- cyclase expression and modulation of cAMP in rat taste cells.
ulation of PLCb2, production of IP3 and release of Ca2+ Am J Physiol Cell Physiol 284, C1420–C1428.
from intracellular stores. Following the increase in Adler, E., Hoon, M.A., Mueller, K.L., Chandrashekar, J.,
intracellular Ca2+, the downstream events vary depend- Ryba, N.J. & Zuker, C.S. 2000. A novel family of mam-
malian taste receptors. Cell 100, 693–702.
ing on the cell type (Fig. 4). In taste cells and solitary
Bachmanov, A.A. & Beauchamp, G.K. 2007. Taste receptor
chemoreceptor cells, the Ca2+ activates TrpM5 to
genes. Annu Rev Nutr 27, 389–414.
depolarize the cell and evoke release of transmitter, Bartel, D.L., Sullivan, S.L., Lavoie, E.G., Sevigny, J. & Finger,
while in smooth muscle cells the Ca2+ activates the T.E. 2006. Nucleoside triphosphate diphosphohydrolase-2 is
Ca2+-dependent K+ channel to hyperpolarize the cell the ecto-ATPase of type I cells in taste buds. J Comp Neurol
and cause muscle relaxation. In ciliated epithelial cells 497, 1–12.
that are not electrically excitable or dependent on Behrens, M., Foerster, S., Staehler, F., Raguse, J.D. & Mey-
membrane potential, the increase in Ca2+ simply causes erhof, W. 2007. Gustatory expression pattern of the human
an increase in ciliary beat frequency, a Ca2+-dependent TAS2R bitter receptor gene family reveals a heterogenous
process. Thus, similar upstream effectors activate dif- population of bitter responsive taste receptor cells. J Neu-
ferent physiological processes in different systems to rosci 27, 12630–12640.
Bernhardt, S.J., Naim, M., Zehavi, U. & Lindemann, B. 1996.
evoke different effects. Interestingly, the ‘bitter’ com-
Changes in IP3 and cytosolic Ca2+ in response to sugars and
pounds all appear to evoke processes that favour their
non-sugar sweeteners in transduction of sweet taste in the
expulsion from the body – in taste cells they evoke an rat. J Physiol 490(Pt 2), 325–336.
unpleasant taste, in SCCs they evoke protective airway Brockhoff, A., Behrens, M., Niv, M.Y. & Meyerhof, W. 2010.
reflexes such as sneezing or coughing and lower in the Structural requirements of bitter taste receptor activation.
airway they enhance ciliary beating and relaxation of Proc Natl Acad Sci USA 107, 11110–11115.
the airway, both of which would promote their expul- Bystrova, M.F., Romanov, R.A., Rogachevskaja, O.A.,
sion. Churbanov, G.D. & Kolesnikov, S.S. 2010. Functional
Several unanswered questions remain about signal- expression of the extracellular-Ca2+-sensing receptor in
ling in taste cells as well as airway epithelium. First, mouse taste cells. J Cell Sci 123, 972–982.
what is the role of gustducin in sweet taste? Gustducin Caicedo, A. & Roper, S.D. 2001. Taste receptor cells that
discriminate between bitter stimuli. Science 291, 1557–1560.
should decrease cAMP levels, but previous biochemical
Cartoni, C., Yasumatsu, K., Ohkuri, T., Shigemura, N.,
studies have shown that most sugars increase cAMP in
Yoshida, R., Godinot, N., le Coutre, J., Ninomiya, Y. &
taste tissue. Studies examining cAMP signalling in Damak, S. 2010. Taste preference for fatty acids is mediated
individual taste cells will be required to resolve the role by GPR40 and GPR120. J Neurosci 30, 8376–8382.
of cAMP in sweet taste. Further, what is the role of Chandrashekar, J., Mueller, K.L., Hoon, M.A., Adler, E., Feng,
T1Rs in the airway epithelium? What compounds L., Guo, W., Zuker, C.S. & Ryba, N.J. 2000. T2Rs function
activate them and what is the effect? Do other natural as bitter taste receptors. Cell 100, 703–711.
compounds, including bacterial or viral gene products Chaudhari, N., Yang, H., Lamp, C., Delay, E., Cartford, C.,
activate T2Rs in vivo? Finally, what is the role of taste Than, T. & Roper, S. 1996. The taste of monosodium glu-
signalling in human airways in vivo? The two human tamate: membrane receptors in taste buds. J Neurosci 16,
studies described above, both used cultured airway cells 3817–3826.
Clapp, T.R., Stone, L.M., Margolskee, R.F. & Kinnamon, S.C.
and culture conditions, can affect gene expression. Fresh
2001. Immunocytochemical evidence for co-expression of
biopsies of human tissue will be required to address this
Type III IP3 receptor with signaling components of bitter
question, as well as whether human airway epithelium taste transduction. BMC Neurosci 2, 6.
has trigeminally innervated SCCs similar to rodents. Clapp, T.R., Yang, R., Stoick, C.L., Kinnamon, S.C. & Kin-
namon, J.C. 2004. Morphologic characterization of rat taste
receptor cells that express components of the phospholipase
Conflict of interest
C signaling pathway. J Comp Neurol 468, 311–321.
None.

 2011 The Author


Acta Physiologica  2011 Scandinavian Physiological Society, doi: 10.1111/j.1748-1716.2011.02308.x 165
17481716, 2012, 2, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/j.1748-1716.2011.02308.x by U. Miguel Hernandez De Elche, Wiley Online Library on [06/11/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Taste GPCR signalling Æ S C Kinnamon Acta Physiol 2012, 204, 158–168

Clapp, T.R., Medler, K.F., Damak, S., Margolskee, R.F. & Hayato, R., Ohtubo, Y. & Yoshii, K. 2007. Functional
Kinnamon, S.C. 2006. Mouse taste cells with G protein- expression of ionotropic purinergic receptors on mouse taste
coupled taste receptors lack voltage-gated calcium channels bud cells. J Physiol 584, 473–488.
and SNAP-25. BMC Biol 4, 7. Hisatsune, C., Yasumatsu, K., Takahashi-Iwanaga, H., Oga-
Clapp, T.R., Trubey, K.R., Vandenbeuch, A., Stone, L.M., wa, N., Kuroda, Y., Yoshida, R., Ninomiya, Y. & Mikos-
Margolskee, R.F., Chaudhari, N. & Kinnamon, S.C. 2008. hiba, K. 2007. Abnormal taste perception in mice lacking the
Tonic activity of Galpha-gustducin regulates taste cell type 3 inositol 1,4,5-trisphosphate receptor. J Biol Chem
responsivity. FEBS Lett 582, 3783–3787. 282, 37225–37231.
Damak, S., Rong, M., Yasumatsu, K., Kokrashvili, Z., Huang, Y.A. & Roper, S.D. 2010. Intracellular Ca(2+) and
Varadarajan, V., Zou, S., Jiang, P., Ninomiya, Y. & TRPM5-mediated membrane depolarization produce ATP
Margolskee, R.F. 2003. Detection of sweet and umami taste secretion from taste receptor cells. J Physiol 588, 2343–
in the absence of taste receptor T1r3. Science 301, 850–853. 2350.
Damak, S., Rong, M., Yasumatsu, K., Kokrashvili, Z., Perez, Huang, L., Shanker, Y.G., Dubauskaite, J., Zheng, J.Z., Yan,
C.A., Shigemura, N., Yoshida, R., Mosinger, B. Jr, Glen- W., Rosenzweig, S., Spielman, A.I., Max, M. & Margolskee,
dinning, J.I., Ninomiya, Y. & Margolskee, R.F. 2006. R.F. 1999. Ggamma13 colocalizes with gustducin in taste
Trpm5 null mice respond to bitter, sweet, and umami com- receptor cells and mediates IP3 responses to bitter denato-
pounds. Chem Senses 31, 253–264. nium. Nat Neurosci 2, 1055–1062.
Dando, R. & Roper, S.D. 2009. Cell-to-cell communication in Huang, Y.J., Maruyama, Y., Lu, K.S., Pereira, E., Plonsky, I.,
intact taste buds through ATP signalling from pannexin 1 Baur, J.E., Wu, D. & Roper, S.D. 2005. Mouse taste buds
gap junction hemichannels. J Physiol 587, 5899–5906. use serotonin as a neurotransmitter. J Neurosci 25, 843–847.
DeFazio, R.A., Dvoryanchikov, G., Maruyama, Y., Kim, J.W., Huang, A.L., Chen, X., Hoon, M.A., Chandrashekar, J., Guo,
Pereira, E., Roper, S.D. & Chaudhari, N. 2006. Separate W., Trankner, D., Ryba, N.J. & Zuker, C.S. 2006. The cells
populations of receptor cells and presynaptic cells in mouse and logic for mammalian sour taste detection. Nature 442,
taste buds. J Neurosci 26, 3971–3980. 934–938.
Delay, E.R., Hernandez, N.P., Bromley, K. & Margolskee, Huang, Y.J., Maruyama, Y., Dvoryanchikov, G., Pereira, E.,
R.F. 2006. Sucrose and monosodium glutamate taste Chaudhari, N. & Roper, S.D. 2007. The role of pannexin 1
thresholds and discrimination ability of T1R3 knockout hemichannels in ATP release and cell-cell communication in
mice. Chem Senses 31, 351–357. mouse taste buds. Proc Natl Acad Sci USA 104, 6436–6441.
Deshpande, D.A., Wang, W.C., McIlmoyle, E.L., Robinett, Huang, Y.A., Maruyama, Y. & Roper, S.D. 2008a. Norepi-
K.S., Schillinger, R.M., An, S.S., Sham, J.S. & Liggett, S.B. nephrine is coreleased with serotonin in mouse taste buds.
2010. Bitter taste receptors on airway smooth muscle J Neurosci 28, 13088–13093.
bronchodilate by localized calcium signaling and reverse Huang, Y.A., Maruyama, Y., Stimac, R. & Roper, S.D. 2008b.
obstruction. Nat Med 16, 1299–1304. Presynaptic (Type III) cells in mouse taste buds sense sour
Finger, T.E. 2006. Evolution of taste. In: J. Kaas (ed.) Evolu- (acid) taste. J Physiol 586, 2903–2912.
tion of the Nervous System, pp. 423–441. Elsevier Press, Huang, Y.A., Dando, R. & Roper, S.D. 2009. Autocrine and
Oxford, UK. paracrine roles for ATP and serotonin in mouse taste buds.
Finger, T.E., Bottger, B., Hansen, A., Anderson, K.T., Alimo- J Neurosci 29, 13909–13918.
hammadi, H. & Silver, W.L. 2003. Solitary chemoreceptor Iwatsuki, K., Ichikawa, R., Hiasa, M., Moriyama, Y., Torii, K.
cells in the nasal cavity serve as sentinels of respiration. Proc & Uneyama, H. 2009. Identification of the vesicular nucle-
Natl Acad Sci USA 100, 8981–8986. otide transporter (VNUT) in taste cells. Biochem Biophys
Finger, T.E., Danilova, V., Barrows, J., Bartel, D.L., Vigers, Res Commun 388, 1–5.
A.J., Stone, L., Hellekant, G. & Kinnamon, S.C. 2005. ATP Jiang, P., Ji, Q., Liu, Z., Snyder, L.A., Benard, L.M., Mar-
signaling is crucial for communication from taste buds to golskee, R.F. & Max, M. 2004. The cysteine-rich region of
gustatory nerves. Science 310, 1495–1499. T1R3 determines responses to intensely sweet proteins.
Glendinning, J.I., Bloom, L.D., Onishi, M., Zheng, K.H., J Biol Chem 279, 45068–45075.
Damak, S., Margolskee, R.F. & Spector, A.C. 2005. Jiang, P., Cui, M., Zhao, B., Snyder, L.A., Benard, L.M.,
Contribution of alpha-gustducin to taste-guided licking Osman, R., Max, M. & Margolskee, R.F. 2005. Identifica-
responses of mice. Chem Senses 30, 299–316. tion of the cyclamate interaction site within the transmem-
Gulbransen, B.D., Clapp, T.R., Finger, T.E. & Kinnamon, S.C. brane domain of the human sweet taste receptor subunit
2008. Nasal solitary chemoreceptor cell responses to bitter T1R3. J Biol Chem 280, 34296–34305.
and trigeminal stimulants in vitro. J Neurophysiol 99, 2929– Kaske, S., Krasteva, G., Konig, P., Kummer, W., Hofmann, T.,
2937. Gudermann, T. & Chubanov, V. 2007. TRPM5, a taste-
Hacker, K., Laskowski, A., Feng, L., Restrepo, D. & Medler, signaling transient receptor potential ion-channel, is a
K. 2008. Evidence for two populations of bitter responsive ubiquitous signaling component in chemosensory cells. BMC
taste cells in mice. J Neurophysiol 99, 1503–1514. Neurosci 8, 49.
Hansen, A. 2007. Olfactory and solitary chemosensory cells: Kim, M.R., Kusakabe, Y., Miura, H., Shindo, Y., Ninomiya,
two different chemosensory systems in the nasal cavity of the Y. & Hino, A. 2003. Regional expression patterns of taste
American alligator, Alligator mississippiensis. BMC Neuro- receptors and gustducin in the mouse tongue. Biochem
sci 8, 64. Biophys Res Commun 312, 500–506.

 2011 The Author


166 Acta Physiologica  2011 Scandinavian Physiological Society, doi: 10.1111/j.1748-1716.2011.02308.x
17481716, 2012, 2, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/j.1748-1716.2011.02308.x by U. Miguel Hernandez De Elche, Wiley Online Library on [06/11/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Acta Physiol 2012, 204, 158–168 S C Kinnamon Æ Taste GPCR signalling
Kotrschal, K. & Whitear, M. 1988. Chemosensory anterior expressed in pancreatic beta-cells activates the calcium and
dorsal fin in rocklings (Gaidropsarus and Ciliata, Teleostei, cyclic AMP signaling systems and stimulates insulin secre-
Gadidae): somatotopic representation of the ramus recurrens tion. PLoS ONE 4, e5106.
facialis as revealed by transganglionic transport of HRP. Nelson, G., Hoon, M.A., Chandrashekar, J., Zhang, Y., Ryba,
J Comp Neurol 268, 109–120. N.J. & Zuker, C.S. 2001. Mammalian sweet taste receptors.
Krasteva, G., Canning, B.J., Veres, T., Papadakis, T., Hart- Cell 106, 381–390.
mann, P., Muchlfield, C., Schiecker, K., Hans, K., Tallini, Nelson, G., Chandrashekar, J., Hoon, M.A., Feng, L., Zhao,
Y.N. & Braun, A.e.a. 2010. Tracheal bursh cells are neuro- G., Ryba, N.J. & Zuker, C.S. 2002. An amino-acid taste
nally connected cholinergic sensory cells (abstract). Soc receptor. Nature 416, 199–202.
Neurosci 773.7. Nie, Y., Vigues, S., Hobbs, J.R., Conn, G.L. & Munger, S.D.
Kuhn, C., Bufe, B., Batram, C. & Meyerhof, W. 2010. Olig- 2005. Distinct contributions of T1R2 and T1R3 taste
omerization of TAS2R bitter taste receptors. Chem Senses receptor subunits to the detection of sweet stimuli. Curr Biol
35, 395–406. 15, 1948–1952.
Li, X., Staszewski, L., Xu, H., Durick, K., Zoller, M. & Adler, Ogura, T., Margolskee, R.F. & Kinnamon, S.C. 2002. Taste
E. 2002. Human receptors for sweet and umami taste. Proc receptor cell responses to the bitter stimulus denatonium
Natl Acad Sci USA 99, 4692–4696. involve Ca2+ influx via store-operated channels. J Neuro-
Lin, W., Ogura, T., Margolskee, R.F., Finger, T.E. & Restrepo, physiol 87, 3152–3155.
D. 2008. TRPM5-expressing solitary chemosensory cells Ogura, T., Krosnowski, K., Zhang, L., Bekkerman, M. & Lin,
respond to odorous irritants. J Neurophysiol 99, 1451–1460. W. 2010. Chemoreception regulates chemical access to
Maruyama, Y., Pereira, E., Margolskee, R.F., Chaudhari, N. & mouse vomeronasal organ: role of solitary chemosensory
Roper, S.D. 2006. Umami responses in mouse taste cells cells. PLoS ONE 5, e11924.
indicate more than one receptor. J Neurosci 26, 2227–2234. Ohmoto, M., Matsumoto, I., Yasuoka, A., Yoshihara, Y. &
Matsunami, H., Montmayeur, J.P. & Buck, L.B. 2000. Abe, K. 2008. Genetic tracing of the gustatory and trigemi-
A family of candidate taste receptors in human and mouse. nal neural pathways originating from T1R3-expressing taste
Nature 404, 601–604. receptor cells and solitary chemoreceptor cells. Mol Cell
Max, M., Shanker, Y.G., Huang, L., Rong, M., Liu, Z., Neurosci 38, 505–517.
Campagne, F., Weinstein, H., Damak, S. & Margolskee, Ohsu, T., Amino, Y., Nagasaki, H., Yamanaka, T., Takeshita, S.,
R.F. 2001. Tas1r3, encoding a new candidate taste receptor, Hatanaka, T., Maruyama, Y., Miyamura, N. & Eto, Y.
is allelic to the sweet responsiveness locus Sac. Nat Genet 28, 2010. Involvement of the calcium-sensing receptor in human
58–63. taste perception. J Biol Chem 285, 1016–1022.
McLaughlin, S.K., McKinnon, P.J. & Margolskee, R.F. 1992. Perez, C.A., Huang, L., Rong, M., Kozak, J.A., Preuss, A.K.,
Gustducin is a taste-cell-specific G protein closely related to Zhang, H., Max, M. & Margolskee, R.F. 2002. A transient
the transducins. Nature 357, 563–569. receptor potential channel expressed in taste receptor cells.
McLaughlin, S.K., McKinnon, P.J., Spickofsky, N., Danho, W. Nat Neurosci 5, 1169–1176.
& Margolskee, R.F. 1994. Molecular cloning of G proteins Ren, X., Zhou, L., Terwilliger, R., Newton, S.S. & de Araujo,
and phosphodiesterases from rat taste cells. Physiol Behav I.E. 2009. Sweet taste signaling functions as a hypothalamic
56, 1157–1164. glucose sensor. Front Integr Neurosci 3, 12.
Meyerhof, W., Batram, C., Kuhn, C., Brockhoff, A., Chudoba, Romanov, R.A., Rogachevskaja, O.A., Bystrova, M.F., Jiang, P.,
E., Bufe, B., Appendino, G. & Behrens, M. 2010. The Margolskee, R.F. & Kolesnikov, S.S. 2007. Afferent neuro-
molecular receptive ranges of human TAS2R bitter taste transmission mediated by hemichannels in mammalian taste
receptors. Chem Senses 35, 157–170. cells. EMBO J 26, 657–667.
Misaka, T., Kusakabe, Y., Emori, Y., Gonoi, T., Arai, S. & Rossler, P., Kroner, C., Freitag, J., Noe, J. & Breer, H. 1998.
Abe, K. 1997. Taste buds have a cyclic nucleotide-activated Identification of a phospholipase C beta subtype in rat taste
channel, CNGgust. J Biol Chem 272, 22623–22629. cells. Eur J Cell Biol 77, 253–261.
Miyoshi, M.A., Abe, K. & Emori, Y. 2001. IP(3) receptor type Royer, S.M. & Kinnamon, J.C. 1991. HVEM serial-section
3 and PLCbeta2 are co-expressed with taste receptors T1R analysis of rabbit foliate taste buds: I. Type III cells and their
and T2R in rat taste bud cells. Chem Senses 26, 259–265. synapses. J Comp Neurol 306, 49–72.
Montmayeur, J.P., Liberles, S.D., Matsunami, H. & Buck, L.B. Ruiz, C.J., Wray, K., Delay, E., Margolskee, R.F. & Kinna-
2001. A candidate taste receptor gene near a sweet taste mon, S.C. 2003. Behavioral evidence for a role of alpha-
locus. Nat Neurosci 4, 492–498. gustducin in glutamate taste. Chem Senses 28, 573–579.
Murata, Y., Yasuo, T., Yoshida, R., Obata, K., Yanagawa, Y., Sainz, E., Korley, J.N., Battey, J.F. & Sullivan, S.L. 2001.
Margolskee, R.F. & Ninomiya, Y. 2010. Action potential- Identification of a novel member of the T1R family of
enhanced ATP release from taste cells through hemichannels. putative taste receptors. J Neurochem 77, 896–903.
J Neurophysiol 104, 896–901. San Gabriel, A., Maekawa, T., Uneyama, H. & Torii, K.
Murray, R.G. & Murray, A. 1971. Relations and possible 2009a. Metabotropic glutamate receptor type 1 in taste tis-
significance of taste bud cells. Contrib Sens Physiol 5, 47–95. sue. Am J Clin Nutr 90, 743S–746S.
Nakagawa, Y., Nagasawa, M., Yamada, S., Hara, A., San Gabriel, A., Uneyama, H., Maekawa, T. & Torii, K.
Mogami, H., Nikolaev, V.O., Lohse, M.J., Shigemura, N., 2009b. The calcium-sensing receptor in taste tissue. Biochem
Ninomiya, Y. & Kojima, I. 2009. Sweet taste receptor Biophys Res Commun 378, 414–418.

 2011 The Author


Acta Physiologica  2011 Scandinavian Physiological Society, doi: 10.1111/j.1748-1716.2011.02308.x 167
17481716, 2012, 2, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/j.1748-1716.2011.02308.x by U. Miguel Hernandez De Elche, Wiley Online Library on [06/11/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Taste GPCR signalling Æ S C Kinnamon Acta Physiol 2012, 204, 158–168

Sbarbati, A., Merigo, F., Benati, D., Tizzano, M., Bernardi, P., Vandenbeuch, A., Clapp, T.R. & Kinnamon, S.C. 2008.
Crescimanno, C. & Osculati, F. 2004. Identification and Amiloride-sensitive channels in type I fungiform taste cells in
characterization of a specific sensory epithelium in the rat mouse. BMC Neurosci 9, 1.
larynx. J Comp Neurol 475, 188–201. Whitear, M. 1992. Solitary chemoreceptor cells. In: T.J. HARA
Shah, A.S., Ben-Shahar, Y., Moninger, T.O., Kline, J.N. & (ed.) Chemoreception in Fishes, 2nd edn, pp. 103–125.
Welsh, M.J. 2009. Motile cilia of human airway epithelia are Elsevier Press, London.
chemosensory. Science 325, 1131–1134. Wong, G.T., Gannon, K.S. & Margolskee, R.F. 1996. Trans-
Shindo, Y., Miura, H., Carninci, P., Kawai, J., Hayashizaki, Y., duction of bitter and sweet taste by gustducin. Nature 381,
Ninomiya, Y., Hino, A., Kanda, T. & Kusakabe, Y. 2008. G 796–800.
alpha14 is a candidate mediator of sweet/umami signal Yan, W., Sunavala, G., Rosenzweig, S., Dasso, M., Brand, J.G.
transduction in the posterior region of the mouse tongue. & Spielman, A.I. 2001. Bitter taste transduced by PLC-
Biochem Biophys Res Commun 376, 504–508. beta(2)-dependent rise in IP(3) and alpha-gustducin-depen-
Singh, N., Vrontakis, M., Parkinson, F. & Chelikani, P. 2011. dent fall in cyclic nucleotides. Am J Physiol Cell Physiol 280,
Functional bitter taste receptors are expressed in brain cells. C742–C751.
Biochem Biophys Res Commun 406, 146–151. Yoshida, R., Miyauchi, A., Yasuo, T., Jyotaki, M., Murata, Y.,
Starostik, M.R., Rebello, M.R., Cotter, K.A., Kulik, A. & Yasumatsu, K., Shigemura, N., Yanagawa, Y., Obata, K.,
Medler, K.F. 2010. Expression of GABAergic receptors in Ueno, H., Margolskee, R.F. & Ninomiya, Y. 2009a. Dis-
mouse taste receptor cells. PLoS ONE 5, e13639. crimination of taste qualities among mouse fungiform taste
Stone, L.M., Barrows, J., Finger, T.E. & Kinnamon, S.C. 2007. bud cells. J Physiol 587, 4425–4439.
Expression of T1Rs and gustducin in palatal taste buds of Yoshida, R., Yasumatsu, K., Shirosaki, S., Jyotaki, M., Horio,
mice. Chem Senses 32, 255–262. N., Murata, Y., Shigemura, N., Nakashima, K. & Nin-
Tizzano, M., Dvoryanchikov, G., Barrows, J.K., Kim, S., omiya, Y. 2009b. Multiple receptor systems for umami taste
Chaudhari, N. & Finger, T.E. 2008. Expression of Galpha14 in mice. Ann N Y Acad Sci 1170, 51–54.
in sweet-transducing taste cells of the posterior tongue. BMC Zhang, Y., Hoon, M.A., Chandrashekar, J., Mueller, K.L.,
Neurosci 9, 110. Cook, B., Wu, D., Zuker, C.S. & Ryba, N.J. 2003.
Tizzano, M., Gulbransen, B.D., Vandenbeuch, A., Clapp, T.R., Coding of sweet, bitter, and umami tastes: different
Herman, J.P., Sibhatu, H.M., Churchill, M.E., Silver, W.L., receptor cells sharing similar signaling pathways. Cell 112,
Kinnamon, S.C. & Finger, T.E. 2010. Nasal chemosensory 293–301.
cells use bitter taste signaling to detect irritants and bacterial Zhang, Z., Zhao, Z., Margolskee, R.F. & Liman, E.R. 2007.
signals. Proc Natl Acad Sci USA 107, 3210–3215. The transduction channel TRPM5 is gated by intracellular
Tizzano, M., Cristofoletti, M., Sbarbati, A. & Finger, T.E. calcium in taste cells. J Neurosci 27, 5777–5786.
2011. Expression of taste receptors in solitary chemosensory Zhang, F., Klebansky, B., Fine, R.M., Xu, H., Pronin, A., Liu,
cells of rodent airways. BMC Pulm Med 11, 3. H., Tachdjian, C. & Li, X. 2008. Molecular mechanism for
Trubey, K.R., Culpepper, S., Maruyama, Y., Kinnamon, S.C. the umami taste synergism. Proc Natl Acad Sci USA 105,
& Chaudhari, N. 2006. Tastants evoke cAMP signal in taste 20930–20934.
buds that is independent of calcium signaling. Am J Physiol Zhao, G.Q., Zhang, Y., Hoon, M.A., Chandrashekar, J.,
Cell Physiol 291, C237–C244. Erlenbach, I., Ryba, N.J. & Zuker, C.S. 2003. The recep-
Vandenbeuch, A. & Kinnamon, S.C. 2009. Why do taste cells tors for mammalian sweet and umami taste. Cell 115, 255–
generate action potentials? J Biol 8, 42. 266.

 2011 The Author


168 Acta Physiologica  2011 Scandinavian Physiological Society, doi: 10.1111/j.1748-1716.2011.02308.x

You might also like