Comprehensive Physiology Wiley Online Library

Physiology of Taste Processing in the Tongue, Gut, and Brain

Full Article on Wiley Online Library



Abstract

The gustatory system detects and informs us about the nature of various chemicals we put in our mouth. Some of these have nutritive value (sugars, amino acids, salts, and fats) and are appetitive and avidly ingested, whereas others (atropine, quinine, nicotine) are aversive and rapidly rejected. However, the gustatory system is mainly responsible for evoking the perception of a limited number of qualities that humans taste as sweet, umami, bitter, sour, salty, and perhaps fat [free fatty acids (FFA)] and starch (malto‐oligosaccharides). The complex flavors and mouthfeel that we experience while eating food result from the integration of taste, odor, texture, pungency, and temperature. The latter three arise primarily from the somatosensory (trigeminal) system. The sensory organs used for detecting and transducing many chemicals are found in taste buds (TBs) located throughout the tongue, soft palate esophagus, and epiglottis. In parallel with the taste system, the trigeminal nerve innervates the peri‐gemmal epithelium to transmit temperature, mechanical stimuli, and painful or cooling sensations such as those produced by changes in temperature as well as from chemicals like capsaicin and menthol, respectively. This article gives an overview of the current knowledge about these TB cells' anatomy and physiology and their trigeminal induced sensations. We then discuss how taste is represented across gustatory cortices using an intermingled and spatially distributed population code. Finally, we review postingestion processing (interoception) and central integration of the tongue‐gut‐brain interaction, ultimately determining our sensations as well as preferences toward the wholesomeness of nutritious foods. © 2021 American Physiological Society. Compr Physiol 11:2489‐2523, 2021.

Figure 1. Figure 1. Anatomy of the tongue and taste buds. (A) The drawing of a human tongue 275. Slight elevations in the dorsal surface demarcate fungiform papillae. Circumvallate papillae (big arrows), with the exception of the central one, form an inverted V‐shape along the anterior of the sulcus terminalis. (B‐D) Scanning electron micrographs of the mucosal surface of a canine lingual epithelium. (B) A section of the anterior epithelium with three round fungiform papillae (Fu) and numerous pointed filiform papillae (Fi). * Indicates a magnification area shown in (C). (D) A magnified view of a taste pore (TP) opening into the fungiform papillae. Note that the cornified layers are shed in sheets. (E) A longitudinal section of a taste bud from a rabbit foliate papilla. Several nuclei of types I and II cells are shown. A single type III cell appears at the left edge of the TB. Arrows mark afferent nerve fibers. The TB extends from the basal lamina (BL) to the epithelium (E) surface, where the taste pore (TP) is in contact with the external oral environment. CTi, connective tissue. (A) Modified, with permission, from Witt M, 2019 285. (B‐D) Modified, with permission, from Holland VF, et al., 1989 109. (E) Modified, with permission, from Royer SM and Kinnamon JC, 1994 221.
Figure 2. Figure 2. Dorsal surface of a human tongue with taste papillae and taste bud cells. A drawing of the human tongue showing three types of taste papillae containing TBs as well as their associated afferent fibers. In fungiform papillae, TBs are located mainly around the tip of the tongue, while in circumvallate and foliate papillae, they are in trenches on the posterior and lateral areas of the tongue, respectively (middle panel). Right panel shows a schematic representation of a TB, the peripheral end organ responsible for initiating and transmitting signals that arise in taste perception. TBs are composed of four different types, I, II, III, and IV, surrounded by stratified, squamous epithelial cells. The apical and basolateral sides of taste receptor cells (TRCs) are demarked by tight junctions (dashed black lines). The TB shows a small pit with a “crater‐like” shape, called the taste pore (TP), where tastants enter to activate TRCs. Afferent nerve fibers arising from gustatory ganglion neurons are shown in black. Some fibers selectively contact one type of TRC, whereas others can target more than one type (see text and Ref. 114). Other gustatory fibers also contact and terminate in the perigemmal epithelium (black). The chorda tympani (CT) is a branch of the facial nerve (CN VII), and GPN refers to the glossopharyngeal nerve (CN IX). Free ending fibers (red) from the lingual branch (LN) of the trigeminal nerve (CN V) are embedded in the keratinocytes surrounding the TB of fungiform papillae [see red V(LN)]. Not shown is the vagus nerve (CN X) that innervates the root of the tongue, epiglottis, and larynx. Modified, with permission, from Witt M, 2019 285.
Figure 3. Figure 3. 3‐D reconstruction of type I taste cells shows their two apical morphologies and how they embrace type II and III cells. (A) Short type I cells (light green) terminate in an apical microvillus that does not reach the taste pore, whereas Tall type I cells (dark green) have a longer branched apical process penetrating the taste pore. (B) A drawing of a TB depicting how type I cells (blue) wrap‐around type II (green) and III (orange) cells. (C) An electron microscopic section of a type I cell enwrapping a type II cell with a thin lamellae sheet (light green). The type II cell is contacting a nerve fiber (nf, yellow) and contains an atypical mitochondrion (atM) that may serve as a source of ATP. (A,C) Modified, with permission, from Yang R, et al., 2019 289. (B) Modified, with permission, from Witt M, 2019 285.
Figure 4. Figure 4. Reconstruction of a type III taste cell and two associated nerve fibers (nF). (A) Accumulations of synaptic vesicles (Ves) near the point of cell‐nerve contact indicate synapses (see arrows and circles in C). At one of these contacts is an atypical mitochondrion (atM‐red) with irregular cristae close to the cell membrane. (B) Type III cell nuclei are irregular and elongated with deep invaginations (arrows). Also visible are two adjacent type I cell nuclei (nu) with small invaginations. Classical chemical synapses (C) enlarged at the lower left with synaptic vesicles (arrowheads) appear at some contact points between the type III cell and nerve fibers (nf, yellow). C: Enlargements from panel (B) showing electron‐dense synaptic vesicles (arrowheads) at points of contact with the nerve fibers. Modified, with permission, from Yang R, et al., 2019 289.
Figure 5. Figure 5. Type IV basal precursor cells. (A) Type IV (basal) cells have nuclei (magenta) situated in the lower quarter of the TB. In contrast, the nuclei of typical type II (light blue) and type III (red) cells lie higher in the taste bud. (B) Type IV cells have a basal process extending to the basal lamina (Figure 1E; see BL) at the base of the TB. This process frequently contacts the basal plexus of nerve fibers penetrating the base of the bud. Some basal cells also have an apically directed process that may contact type II or type III cells. Modified, with permission, from Yang R, et al., 2019 289.
Figure 6. Figure 6. Thermal transient receptor potential (TRPs) channels. A selected group of thermal TRPs (and their temperature sensitivities) that are activated either indirectly by tastants (TRPM5; sweet, bitter, and umami—see text) or directly by various spices (TRPA1, TRPV1) or other compounds (TRPM8, TRPV2). A ligand for TRPV4 is bisandrographolide A, extracted from the King of Bitter. (inset) Modified, with permission, from Roper SD, 2014 216; Modified, with permission, from Simon SA and Gutierrez R, 2017 240.
Figure 7. Figure 7. Schematic diagram of gustatory and trigeminal afferent nerve fibers. (A) Chorda Tympani (CT) afferent fibers from geniculate ganglion (GG) neurons can innervate adjacent and distal TBs. Some fibers only contact type II cells (blue), whereas others only type III cells (gray). In addition, some fibers can contact both type II and III cells (purple) (see text). The cell bodies of the CT and the greater superficial petrosal (GSP) nerves reside in the GG and project to rNTS (see pictures in C). Some CT fibers do not target fungiform taste bud cells but instead terminate in lingual epithelial tissue (black fibers) (see Figure 8C, cluster M5). The “somatosensory” GG cells innervating the outer ear are not shown. Along with gustatory fibers from the CT run trigeminal innervation. Included in that group are neurons that contain transient receptor potential (TRP) cation channel subfamily M member 8 (TRPM8) neurons that respond to menthol and from the transient receptor potential cation channel subfamily V member 1 (TRPV1), which is a receptor for capsaicin the principal ingredient in chili peppers responsible for its burning sensation (Figure 6). Trigeminal nerve fibers project to the spinal trigeminal nucleus (SpVc) and the rNTS 302. (B) A cresyl‐violet‐stained of a rat GG soma also shown both the CT and GSP fibers (see arrows). (C) Upper picture depicts histology of CT fibers innervating the rNTS, and the lower panel shows fluorescent glossopharyngeal nerve (GPN) in red and GSP fibers in green. 4 denotes the 4th ventricle. (A) Modified, with permission, from Zaidi FN, et al., 2008 302. (C) Modified, with permission, from Martin LJ, et al., 2018 164.
Figure 8. Figure 8. Single‐cell genetic profiling of mouse geniculate ganglion (GG) neurons found two major cell types: those expressing the transcriptional factor Phox2b+ that conveys gustatory information and those expressing Phox2b fibers that are somatosensory neurons that innervate the outer ear. (A) Labeling the GG. The immunostaining of purinergic receptor P2X3 (a marker for afferent nerve fibers is in red) and purple represents the GG neurons expressing Phox2b+ and P2X3+ are labeled in purple (merge). The nonnuclear “green” signal is an unspecific binding of the mouse antibody. (B) A meta‐analysis of three experiments showing the genetic profiling of individual GG neurons 4,64,304. In the middle panel, a dot represents a single GG neuron classified into seven distinct clusters (M0‐M6) arranged according to its five main genes (see correlation matrix in C). (C) Clusters from M1‐M6 corresponds to gustatory GG neurons (expressing Phox2b+). The somatosensory GG neurons belong to cluster M0 (Phox2b). The cluster M2 is composed of GG neurons expressing the Spon1 gene, and they are responsive to sweet tastants. The cluster M6 is involved in acid taste transduction and is characterized by the expression of the Penk gene. M5 is an ensemble of GG neurons that may be involved in mechano‐transduction information from the tongue. These cells expressed the gene for Piezo2. Modified, with permission, from Anderson CB and Larson ED, 2020 4.
Figure 9. Figure 9. Primary canonical signaling pathways are shared by sweet and bitter tastants and lamino acids. After chemicals are bound to their associated GPCRs, G‐protein βγ subunits then → PLC‐β2 → PIP2 → IP3 and activation of endoplasmic reticulum IP3R3 receptors → increasing intracellular [Ca2+]i → cation influx through TRPM4 and TRPM5 channels → membrane depolarization → opening the CALHM1/3 channels → secretion of ATP that activates P2X2/P2X3 receptors on primary afferent neurons. In parallel, activation of voltage‐gated Na+ channels (Nav) amplifies the membrane depolarization and increases ATP secretion (see text). Some type II cells also express a sodium‐glucose‐linked transporter 1 (SGLT1) that can distinguish glucose from noncaloric artificial sweeteners that can also activate T1R2/T1R3 receptors. Type II cells can also detect and transduce long‐chain fatty acids (FAs). FAs bind to the translocator CD36 to activate specific G protein‐coupled receptors (like GPR120 receptors) to initiate a transduction cascade that in turn produces a second messenger leading to release intracellular [Ca2+]i (see the text on fat for details ). It remains unknown as to whether CD36 serves as a chaperon for GPR120 to transduce FFAs or both works independently. The basal part of the type II cells can also respond in an endocrine, autocrine, or paracrine manner to hormones such as leptin, GLP‐1, and ghrelin (see text for additional details). The TRC in the middle depicts a type II cell that expresses the l‐amino acid receptor, T1R1/T1R3. Shown at the right is a type II cell with three T2R receptors activated by bitter tastants. It remains to determine whether type II bitter cells also express receptors for peptides. Modified, with permission, from Gao N, et al., 2009 84; Liu P, et al., 2011 153.
Figure 10. Figure 10. Morphology of a type II cell and a schematic of the structure of the human “sweet taste” receptor hT1R2/3 with its many binding sites. Reconstruction (3‐D) of a type II cell from a mouse's circumvallate papillae. Red spots indicate the position of atypical mitochondria (atM), which may act as a source of ATP to be released to activate receptors on primary afferent fibers. Also shown is a microvillus that extends through the taste pore (up—not shown) into the oral cavity. Inset. The heterodimer T1R2/T1R3 is composed of an extracellular amino terminal domain (ATD), containing a venus flytrap domain (VFD) and a cysteine‐rich domain (CRD). The VFD has two lobes that change their conformation to “open” or “closed” states, like that seen in the carnivorous plant with the same name. The VFD is connected via the CRD to the seven helical transmembrane domains (TMD). Importantly, sweet‐tasting molecules bind to different receptor sites (arrows), inducing conformational changes eliciting a canonical signaling cascade, shown in Figures 9 and 11. (Left) Modified, with permission, from Yang R, et al., 2019 289. (Right) Modified, with permission, from Laffitte A, et al., 2014 136.
Figure 11. Figure 11. Sweet, umami, and bitter taste responses require the co‐activation of both a narrowly tuned type II cell (left) and a broadly tuned type III cell (right). The integration of information between two types of taste cells could occur in the taste bud (TB) or by those afferent fibers that contact both type II and type III cells. At right is shown a recently identified broadly responsive type III cell (BRtypeIII cell) that responds to acid, sweet, umami, and bitter tastants via not yet identified receptors that activate PLC‐β3 → IP3R1 that then produces an increase of intracellular Ca2+ that probably activate TRPM4 channels (see ?) present in those cells. This leads to the activation of voltage‐gated calcium channels (VGCCs) that cause the release of synaptic vesicles whose transmitters activate primary neurons 10. It is still unknown what transmitter uses the BRTypeIII cell to communicate with afferent nerves (see small ? next to vesicles). Wherever the logic AND circuit occurs (see large ? marks), it should happen between the TB and GG before taste information is sent to the rNTS (light blue somas). From the rNTS, taste information is partitioned into oromotor reticular formation (RF; red neurons), where the central pattern generator for rhythmic licking is located 266 or to a classical taste/visceral pathway in the parabrachial nucleus (PBN; dark blue neurons). Modified, with permission, from Travers JB, et al., 1997 266; Banik DD, et al., 2020 10.
Figure 12. Figure 12. The signaling cascade pathways for acid taste transduction. Type III cells express the proton permeable ion channel, otopetrin‐1 (OTOP1) 265,304. Influx of H+ through OTOP1 acidifies the cytoplasm, which could inhibit potassium Kir2.1 channels that induce a small depolarizing (inward) current sufficient to gate voltage‐gated Ca2+ channels and produce action potentials (Nav), leading to the subsequent release of synaptic vesicles (black circles) containing 5‐HT that may bind to 5‐HT3aR channels on afferent nerves. Alternatively, it perhaps could co‐release ATP that activates the purinergic receptors P2X2/3 (not shown). A parallel pathway for weak acids has been proposed that involves a passive diffusion for weak acids (HA) into the cell and the subsequent dissociation of H+ and cytoplasmic acidification inactivates the Kir2.1 channel. Also depicted is the possibility that the associated anion (A) binding to OTOP1 may produce an allosteric effect that could account for the weak/strong acid paradox (see text). Type III cells and acid‐activated trigeminal nerves (drawing at right) are both necessary to trigger aversive responses induced by acid stimuli. Modified, with permission, from Zhang J, et al., 2019 304; Teng B, et al., 2019 265.
Figure 13. Figure 13. Cell types involved in low NaCl attraction and high NaCl rejection. Amiloride‐sensitive cells: Left cell displays, the low concentration of salty taste is transduced by an all‐electrical pathway bypassing intracellular calcium signaling to secrete ATP via the CALHM1/3 channel. From left to right, the second cell depicts a benzamil‐inhibitable type I GAD65+ expressing cell whose optogenetic activation (with ChR2) can drive low sodium appetite during salt deprivation. These cells signaling pathways are still unknown but likely involve the β‐ENaC subunit 156. Amiloride‐insensitive cells: High Salt. The third cell on the right is involved in detecting the Cl anion under high NaCl concentrations. The receptor and signaling pathways that transduce Cl are currently unknown 211. Another subpopulation of type II cells expressing the bitter receptor T2R32+ is also recruited after applying high sodium concentrations 193. The signaling pathway for this TRC has not been described, although it is expected to be the same canonical pathway used by bitter‐sensitive type II cells. The type III cell expressing Pkdl1+ has also been shown to be involved in high sodium detection 193. Trigeminal nerve: Hypertonic (>1M), salt concentrations diffuse into the perigemmal epithelium and activate trigeminal nerve endings. Modified, with permission, from Lossow K, et al., 2020 156; Oka Y, et al., 2013 193; Roebber JK, et al., 2019 211.
Figure 14. Figure 14. Responses from specialist and generalist taste cells are found throughout the entire rodent gustatory pathway. (A) Results of calcium imaging TRCs in anesthetized mice. Seventy percent of TRCs exhibited narrowly tuned responses to only one taste quality: sweet, bitter, umami, NaCl, and acid. For the remainder, 30% of the cells responded to more than one tastant [upper cylindrical responses 102]. The colors in the TRCs depict the best‐stimulus of narrowly tuned cells. (B) Electrophysiological recordings of tastant‐evoked action potentials from a specialist TRC that is only responsive to HCl and the response to a generalist cell that is activated by MSG, QHCl, HCl, and NaCl. Bar indicates the duration of the stimulus. Note that these responses occurred even in isolated taste buds from Ref. 298. (C) Afferent fibers of the CT have cell bodies in the geniculate ganglion (GG), and segregated GG populations are more sensitive to one taste quality (Penk for acid; Cdh13 for bitter; spon1for sweet; Cdh4 umami; and Egr2 for NaCl 304. The inset: GG neurons selective to one taste quality are spatially intermingled throughout the GG (see Ref. 11). In the rNTS, Pdyn (prodynorphin) expressing neurons convey acid information 304, whereas Sst (somatostatin) neurons respond to bitter, and Calb2 (Calbindin 2) to sweet tastants. All these rNTS neurons are also spatially intermingled (not shown). In contrast, Satb2 expressing neurons in the PBN are broadly tuned and respond to all taste qualities, but their activation enhances sucrose palatability and makes bitter tastants more aversive. CGRP (calcitonin gene‐related peptide) neurons are not involved in taste processing per se. However, they sense aversive and visceral malaise stimuli in general 119. (D) In the Insular cortex (IC), taste responses are spatially distributed and intermixed, with no apparent taste “map” organization (note the color‐coded for tastants is different from panels A and B, S, sucrose; N, NaCl; CA, citric acid; Q, quinine; and W, water). From the gustatory cortex (IC), taste information goes to reaches the orbitofrontal cortex (OFC) that is commonly referred to as the secondary taste cortex. rNTS, rostral nucleus tractus solitarius; PBN, parabrachial nucleus; VPMpc, ventral posterior medial thalamus (parvocellular region); anterior insular cortex (aIC), and posterior (pIC); MCA, medial cerebral artery. (A) Modified, with permission, from Han J and Choi M, 2018 102. (B) Modified, with permission, from Yoshida R, et al., 2009 298. (C) Modified, with permission, from Jarvie BC, et al., 2021 119; Zhang J, et al., 2019 304. (D) Modified, with permission, from Chen K, et al., 2020 40.
Figure 15. Figure 15. Gut nutrient‐sensing can induce food preferences and promote food‐seeking behavior. Across days, trained rodents develop a preference toward instrumental actions such as lever pressing resulting in an intragastric infusion of sucrose over sucralose (Splenda®), an artificial nonnutritive sweet tastant. Modified, with permission, from Fernandes AB, et al., 2020 72.
Figure 16. Figure 16. Schematic representation of the triad tongue, gut, brain interaction. Shown is the dorsal tongue whose taste responsive neurons first project to the rostral rNTS and vagal neurons innervating the gut, which project to ventro‐caudal vcNTS. These two pathways will result in dopamine (DA) release in the dorsal (DS) or ventral striatum (VS) by activating dopaminergic neurons in the substantial nigra par compacta (SNpc) or the ventral tegmental area (VTA), respectively. The ARC nucleus in the hypothalamus contains AgRP neurons that reflect the animals' internal “hunger” state and can be modulated by hormones like CCK, ghrelin, PYY, leptin, and GLP‐1 that signal an internal state of hunger or satiety. The lower right shows EEC cells that are sensitive to free fatty acids (FFA's—left) and l‐amino acids (right). It also shows a neuropod cell the latter forms synapses with vagal nerves activated by glucose via SGLT1 and uses glutamate as a transmitter to rapidly send information to the brain. For noncaloric sweeteners, these neuropod cells release ATP as a transmitter. The tongue, gut, and brain form a closed‐loop feedback system essential to determine the taste, internal state, and nutritive value of food. Multiple populations of neurons and brain circuits integrate the exteroceptive (tongue‐related) and interoceptive (gut‐related) events triggered by food intake to interpret and generate a conscious taste percept and an unconscious preference for food (see text for details). (inset) Modified, with permission, from Kaelberer MM, et al., 2020 127.


Figure 1. Anatomy of the tongue and taste buds. (A) The drawing of a human tongue 275. Slight elevations in the dorsal surface demarcate fungiform papillae. Circumvallate papillae (big arrows), with the exception of the central one, form an inverted V‐shape along the anterior of the sulcus terminalis. (B‐D) Scanning electron micrographs of the mucosal surface of a canine lingual epithelium. (B) A section of the anterior epithelium with three round fungiform papillae (Fu) and numerous pointed filiform papillae (Fi). * Indicates a magnification area shown in (C). (D) A magnified view of a taste pore (TP) opening into the fungiform papillae. Note that the cornified layers are shed in sheets. (E) A longitudinal section of a taste bud from a rabbit foliate papilla. Several nuclei of types I and II cells are shown. A single type III cell appears at the left edge of the TB. Arrows mark afferent nerve fibers. The TB extends from the basal lamina (BL) to the epithelium (E) surface, where the taste pore (TP) is in contact with the external oral environment. CTi, connective tissue. (A) Modified, with permission, from Witt M, 2019 285. (B‐D) Modified, with permission, from Holland VF, et al., 1989 109. (E) Modified, with permission, from Royer SM and Kinnamon JC, 1994 221.


Figure 2. Dorsal surface of a human tongue with taste papillae and taste bud cells. A drawing of the human tongue showing three types of taste papillae containing TBs as well as their associated afferent fibers. In fungiform papillae, TBs are located mainly around the tip of the tongue, while in circumvallate and foliate papillae, they are in trenches on the posterior and lateral areas of the tongue, respectively (middle panel). Right panel shows a schematic representation of a TB, the peripheral end organ responsible for initiating and transmitting signals that arise in taste perception. TBs are composed of four different types, I, II, III, and IV, surrounded by stratified, squamous epithelial cells. The apical and basolateral sides of taste receptor cells (TRCs) are demarked by tight junctions (dashed black lines). The TB shows a small pit with a “crater‐like” shape, called the taste pore (TP), where tastants enter to activate TRCs. Afferent nerve fibers arising from gustatory ganglion neurons are shown in black. Some fibers selectively contact one type of TRC, whereas others can target more than one type (see text and Ref. 114). Other gustatory fibers also contact and terminate in the perigemmal epithelium (black). The chorda tympani (CT) is a branch of the facial nerve (CN VII), and GPN refers to the glossopharyngeal nerve (CN IX). Free ending fibers (red) from the lingual branch (LN) of the trigeminal nerve (CN V) are embedded in the keratinocytes surrounding the TB of fungiform papillae [see red V(LN)]. Not shown is the vagus nerve (CN X) that innervates the root of the tongue, epiglottis, and larynx. Modified, with permission, from Witt M, 2019 285.


Figure 3. 3‐D reconstruction of type I taste cells shows their two apical morphologies and how they embrace type II and III cells. (A) Short type I cells (light green) terminate in an apical microvillus that does not reach the taste pore, whereas Tall type I cells (dark green) have a longer branched apical process penetrating the taste pore. (B) A drawing of a TB depicting how type I cells (blue) wrap‐around type II (green) and III (orange) cells. (C) An electron microscopic section of a type I cell enwrapping a type II cell with a thin lamellae sheet (light green). The type II cell is contacting a nerve fiber (nf, yellow) and contains an atypical mitochondrion (atM) that may serve as a source of ATP. (A,C) Modified, with permission, from Yang R, et al., 2019 289. (B) Modified, with permission, from Witt M, 2019 285.


Figure 4. Reconstruction of a type III taste cell and two associated nerve fibers (nF). (A) Accumulations of synaptic vesicles (Ves) near the point of cell‐nerve contact indicate synapses (see arrows and circles in C). At one of these contacts is an atypical mitochondrion (atM‐red) with irregular cristae close to the cell membrane. (B) Type III cell nuclei are irregular and elongated with deep invaginations (arrows). Also visible are two adjacent type I cell nuclei (nu) with small invaginations. Classical chemical synapses (C) enlarged at the lower left with synaptic vesicles (arrowheads) appear at some contact points between the type III cell and nerve fibers (nf, yellow). C: Enlargements from panel (B) showing electron‐dense synaptic vesicles (arrowheads) at points of contact with the nerve fibers. Modified, with permission, from Yang R, et al., 2019 289.


Figure 5. Type IV basal precursor cells. (A) Type IV (basal) cells have nuclei (magenta) situated in the lower quarter of the TB. In contrast, the nuclei of typical type II (light blue) and type III (red) cells lie higher in the taste bud. (B) Type IV cells have a basal process extending to the basal lamina (Figure 1E; see BL) at the base of the TB. This process frequently contacts the basal plexus of nerve fibers penetrating the base of the bud. Some basal cells also have an apically directed process that may contact type II or type III cells. Modified, with permission, from Yang R, et al., 2019 289.


Figure 6. Thermal transient receptor potential (TRPs) channels. A selected group of thermal TRPs (and their temperature sensitivities) that are activated either indirectly by tastants (TRPM5; sweet, bitter, and umami—see text) or directly by various spices (TRPA1, TRPV1) or other compounds (TRPM8, TRPV2). A ligand for TRPV4 is bisandrographolide A, extracted from the King of Bitter. (inset) Modified, with permission, from Roper SD, 2014 216; Modified, with permission, from Simon SA and Gutierrez R, 2017 240.


Figure 7. Schematic diagram of gustatory and trigeminal afferent nerve fibers. (A) Chorda Tympani (CT) afferent fibers from geniculate ganglion (GG) neurons can innervate adjacent and distal TBs. Some fibers only contact type II cells (blue), whereas others only type III cells (gray). In addition, some fibers can contact both type II and III cells (purple) (see text). The cell bodies of the CT and the greater superficial petrosal (GSP) nerves reside in the GG and project to rNTS (see pictures in C). Some CT fibers do not target fungiform taste bud cells but instead terminate in lingual epithelial tissue (black fibers) (see Figure 8C, cluster M5). The “somatosensory” GG cells innervating the outer ear are not shown. Along with gustatory fibers from the CT run trigeminal innervation. Included in that group are neurons that contain transient receptor potential (TRP) cation channel subfamily M member 8 (TRPM8) neurons that respond to menthol and from the transient receptor potential cation channel subfamily V member 1 (TRPV1), which is a receptor for capsaicin the principal ingredient in chili peppers responsible for its burning sensation (Figure 6). Trigeminal nerve fibers project to the spinal trigeminal nucleus (SpVc) and the rNTS 302. (B) A cresyl‐violet‐stained of a rat GG soma also shown both the CT and GSP fibers (see arrows). (C) Upper picture depicts histology of CT fibers innervating the rNTS, and the lower panel shows fluorescent glossopharyngeal nerve (GPN) in red and GSP fibers in green. 4 denotes the 4th ventricle. (A) Modified, with permission, from Zaidi FN, et al., 2008 302. (C) Modified, with permission, from Martin LJ, et al., 2018 164.


Figure 8. Single‐cell genetic profiling of mouse geniculate ganglion (GG) neurons found two major cell types: those expressing the transcriptional factor Phox2b+ that conveys gustatory information and those expressing Phox2b fibers that are somatosensory neurons that innervate the outer ear. (A) Labeling the GG. The immunostaining of purinergic receptor P2X3 (a marker for afferent nerve fibers is in red) and purple represents the GG neurons expressing Phox2b+ and P2X3+ are labeled in purple (merge). The nonnuclear “green” signal is an unspecific binding of the mouse antibody. (B) A meta‐analysis of three experiments showing the genetic profiling of individual GG neurons 4,64,304. In the middle panel, a dot represents a single GG neuron classified into seven distinct clusters (M0‐M6) arranged according to its five main genes (see correlation matrix in C). (C) Clusters from M1‐M6 corresponds to gustatory GG neurons (expressing Phox2b+). The somatosensory GG neurons belong to cluster M0 (Phox2b). The cluster M2 is composed of GG neurons expressing the Spon1 gene, and they are responsive to sweet tastants. The cluster M6 is involved in acid taste transduction and is characterized by the expression of the Penk gene. M5 is an ensemble of GG neurons that may be involved in mechano‐transduction information from the tongue. These cells expressed the gene for Piezo2. Modified, with permission, from Anderson CB and Larson ED, 2020 4.


Figure 9. Primary canonical signaling pathways are shared by sweet and bitter tastants and lamino acids. After chemicals are bound to their associated GPCRs, G‐protein βγ subunits then → PLC‐β2 → PIP2 → IP3 and activation of endoplasmic reticulum IP3R3 receptors → increasing intracellular [Ca2+]i → cation influx through TRPM4 and TRPM5 channels → membrane depolarization → opening the CALHM1/3 channels → secretion of ATP that activates P2X2/P2X3 receptors on primary afferent neurons. In parallel, activation of voltage‐gated Na+ channels (Nav) amplifies the membrane depolarization and increases ATP secretion (see text). Some type II cells also express a sodium‐glucose‐linked transporter 1 (SGLT1) that can distinguish glucose from noncaloric artificial sweeteners that can also activate T1R2/T1R3 receptors. Type II cells can also detect and transduce long‐chain fatty acids (FAs). FAs bind to the translocator CD36 to activate specific G protein‐coupled receptors (like GPR120 receptors) to initiate a transduction cascade that in turn produces a second messenger leading to release intracellular [Ca2+]i (see the text on fat for details ). It remains unknown as to whether CD36 serves as a chaperon for GPR120 to transduce FFAs or both works independently. The basal part of the type II cells can also respond in an endocrine, autocrine, or paracrine manner to hormones such as leptin, GLP‐1, and ghrelin (see text for additional details). The TRC in the middle depicts a type II cell that expresses the l‐amino acid receptor, T1R1/T1R3. Shown at the right is a type II cell with three T2R receptors activated by bitter tastants. It remains to determine whether type II bitter cells also express receptors for peptides. Modified, with permission, from Gao N, et al., 2009 84; Liu P, et al., 2011 153.


Figure 10. Morphology of a type II cell and a schematic of the structure of the human “sweet taste” receptor hT1R2/3 with its many binding sites. Reconstruction (3‐D) of a type II cell from a mouse's circumvallate papillae. Red spots indicate the position of atypical mitochondria (atM), which may act as a source of ATP to be released to activate receptors on primary afferent fibers. Also shown is a microvillus that extends through the taste pore (up—not shown) into the oral cavity. Inset. The heterodimer T1R2/T1R3 is composed of an extracellular amino terminal domain (ATD), containing a venus flytrap domain (VFD) and a cysteine‐rich domain (CRD). The VFD has two lobes that change their conformation to “open” or “closed” states, like that seen in the carnivorous plant with the same name. The VFD is connected via the CRD to the seven helical transmembrane domains (TMD). Importantly, sweet‐tasting molecules bind to different receptor sites (arrows), inducing conformational changes eliciting a canonical signaling cascade, shown in Figures 9 and 11. (Left) Modified, with permission, from Yang R, et al., 2019 289. (Right) Modified, with permission, from Laffitte A, et al., 2014 136.


Figure 11. Sweet, umami, and bitter taste responses require the co‐activation of both a narrowly tuned type II cell (left) and a broadly tuned type III cell (right). The integration of information between two types of taste cells could occur in the taste bud (TB) or by those afferent fibers that contact both type II and type III cells. At right is shown a recently identified broadly responsive type III cell (BRtypeIII cell) that responds to acid, sweet, umami, and bitter tastants via not yet identified receptors that activate PLC‐β3 → IP3R1 that then produces an increase of intracellular Ca2+ that probably activate TRPM4 channels (see ?) present in those cells. This leads to the activation of voltage‐gated calcium channels (VGCCs) that cause the release of synaptic vesicles whose transmitters activate primary neurons 10. It is still unknown what transmitter uses the BRTypeIII cell to communicate with afferent nerves (see small ? next to vesicles). Wherever the logic AND circuit occurs (see large ? marks), it should happen between the TB and GG before taste information is sent to the rNTS (light blue somas). From the rNTS, taste information is partitioned into oromotor reticular formation (RF; red neurons), where the central pattern generator for rhythmic licking is located 266 or to a classical taste/visceral pathway in the parabrachial nucleus (PBN; dark blue neurons). Modified, with permission, from Travers JB, et al., 1997 266; Banik DD, et al., 2020 10.


Figure 12. The signaling cascade pathways for acid taste transduction. Type III cells express the proton permeable ion channel, otopetrin‐1 (OTOP1) 265,304. Influx of H+ through OTOP1 acidifies the cytoplasm, which could inhibit potassium Kir2.1 channels that induce a small depolarizing (inward) current sufficient to gate voltage‐gated Ca2+ channels and produce action potentials (Nav), leading to the subsequent release of synaptic vesicles (black circles) containing 5‐HT that may bind to 5‐HT3aR channels on afferent nerves. Alternatively, it perhaps could co‐release ATP that activates the purinergic receptors P2X2/3 (not shown). A parallel pathway for weak acids has been proposed that involves a passive diffusion for weak acids (HA) into the cell and the subsequent dissociation of H+ and cytoplasmic acidification inactivates the Kir2.1 channel. Also depicted is the possibility that the associated anion (A) binding to OTOP1 may produce an allosteric effect that could account for the weak/strong acid paradox (see text). Type III cells and acid‐activated trigeminal nerves (drawing at right) are both necessary to trigger aversive responses induced by acid stimuli. Modified, with permission, from Zhang J, et al., 2019 304; Teng B, et al., 2019 265.


Figure 13. Cell types involved in low NaCl attraction and high NaCl rejection. Amiloride‐sensitive cells: Left cell displays, the low concentration of salty taste is transduced by an all‐electrical pathway bypassing intracellular calcium signaling to secrete ATP via the CALHM1/3 channel. From left to right, the second cell depicts a benzamil‐inhibitable type I GAD65+ expressing cell whose optogenetic activation (with ChR2) can drive low sodium appetite during salt deprivation. These cells signaling pathways are still unknown but likely involve the β‐ENaC subunit 156. Amiloride‐insensitive cells: High Salt. The third cell on the right is involved in detecting the Cl anion under high NaCl concentrations. The receptor and signaling pathways that transduce Cl are currently unknown 211. Another subpopulation of type II cells expressing the bitter receptor T2R32+ is also recruited after applying high sodium concentrations 193. The signaling pathway for this TRC has not been described, although it is expected to be the same canonical pathway used by bitter‐sensitive type II cells. The type III cell expressing Pkdl1+ has also been shown to be involved in high sodium detection 193. Trigeminal nerve: Hypertonic (>1M), salt concentrations diffuse into the perigemmal epithelium and activate trigeminal nerve endings. Modified, with permission, from Lossow K, et al., 2020 156; Oka Y, et al., 2013 193; Roebber JK, et al., 2019 211.


Figure 14. Responses from specialist and generalist taste cells are found throughout the entire rodent gustatory pathway. (A) Results of calcium imaging TRCs in anesthetized mice. Seventy percent of TRCs exhibited narrowly tuned responses to only one taste quality: sweet, bitter, umami, NaCl, and acid. For the remainder, 30% of the cells responded to more than one tastant [upper cylindrical responses 102]. The colors in the TRCs depict the best‐stimulus of narrowly tuned cells. (B) Electrophysiological recordings of tastant‐evoked action potentials from a specialist TRC that is only responsive to HCl and the response to a generalist cell that is activated by MSG, QHCl, HCl, and NaCl. Bar indicates the duration of the stimulus. Note that these responses occurred even in isolated taste buds from Ref. 298. (C) Afferent fibers of the CT have cell bodies in the geniculate ganglion (GG), and segregated GG populations are more sensitive to one taste quality (Penk for acid; Cdh13 for bitter; spon1for sweet; Cdh4 umami; and Egr2 for NaCl 304. The inset: GG neurons selective to one taste quality are spatially intermingled throughout the GG (see Ref. 11). In the rNTS, Pdyn (prodynorphin) expressing neurons convey acid information 304, whereas Sst (somatostatin) neurons respond to bitter, and Calb2 (Calbindin 2) to sweet tastants. All these rNTS neurons are also spatially intermingled (not shown). In contrast, Satb2 expressing neurons in the PBN are broadly tuned and respond to all taste qualities, but their activation enhances sucrose palatability and makes bitter tastants more aversive. CGRP (calcitonin gene‐related peptide) neurons are not involved in taste processing per se. However, they sense aversive and visceral malaise stimuli in general 119. (D) In the Insular cortex (IC), taste responses are spatially distributed and intermixed, with no apparent taste “map” organization (note the color‐coded for tastants is different from panels A and B, S, sucrose; N, NaCl; CA, citric acid; Q, quinine; and W, water). From the gustatory cortex (IC), taste information goes to reaches the orbitofrontal cortex (OFC) that is commonly referred to as the secondary taste cortex. rNTS, rostral nucleus tractus solitarius; PBN, parabrachial nucleus; VPMpc, ventral posterior medial thalamus (parvocellular region); anterior insular cortex (aIC), and posterior (pIC); MCA, medial cerebral artery. (A) Modified, with permission, from Han J and Choi M, 2018 102. (B) Modified, with permission, from Yoshida R, et al., 2009 298. (C) Modified, with permission, from Jarvie BC, et al., 2021 119; Zhang J, et al., 2019 304. (D) Modified, with permission, from Chen K, et al., 2020 40.


Figure 15. Gut nutrient‐sensing can induce food preferences and promote food‐seeking behavior. Across days, trained rodents develop a preference toward instrumental actions such as lever pressing resulting in an intragastric infusion of sucrose over sucralose (Splenda®), an artificial nonnutritive sweet tastant. Modified, with permission, from Fernandes AB, et al., 2020 72.


Figure 16. Schematic representation of the triad tongue, gut, brain interaction. Shown is the dorsal tongue whose taste responsive neurons first project to the rostral rNTS and vagal neurons innervating the gut, which project to ventro‐caudal vcNTS. These two pathways will result in dopamine (DA) release in the dorsal (DS) or ventral striatum (VS) by activating dopaminergic neurons in the substantial nigra par compacta (SNpc) or the ventral tegmental area (VTA), respectively. The ARC nucleus in the hypothalamus contains AgRP neurons that reflect the animals' internal “hunger” state and can be modulated by hormones like CCK, ghrelin, PYY, leptin, and GLP‐1 that signal an internal state of hunger or satiety. The lower right shows EEC cells that are sensitive to free fatty acids (FFA's—left) and l‐amino acids (right). It also shows a neuropod cell the latter forms synapses with vagal nerves activated by glucose via SGLT1 and uses glutamate as a transmitter to rapidly send information to the brain. For noncaloric sweeteners, these neuropod cells release ATP as a transmitter. The tongue, gut, and brain form a closed‐loop feedback system essential to determine the taste, internal state, and nutritive value of food. Multiple populations of neurons and brain circuits integrate the exteroceptive (tongue‐related) and interoceptive (gut‐related) events triggered by food intake to interpret and generate a conscious taste percept and an unconscious preference for food (see text for details). (inset) Modified, with permission, from Kaelberer MM, et al., 2020 127.
References
 1.Ackroff K, Yiin Y‐M, Sclafani A. Post‐oral infusion sites that support glucose‐conditioned flavor preferences in rats. Physiol Behav 99: 402‐411, 2010. DOI: 10.1016/j.physbeh.2009.12.012.
 2.Adler E, Hoon MA, Mueller KL, Chandrashekar J, Ryba NJ, Zuker CS. A novel family of mammalian taste receptors. Cell 100: 693‐702, 2000. DOI: 10.1016/s0092‐8674(00)80705‐9.
 3.Andersen CA, Nielsen L, Møller S, Kidmose P. Cortical response to fat taste. Chem Senses 45: 283‐291, 2020. DOI: 10.1093/chemse/bjaa019.
 4.Anderson CB, Larson ED. Single cell transcriptional profiling of Phox2b‐expressing geniculate ganglion neurons. bioRxiv 812578, 2020. DOI: 10.1101/812578.
 5.Aponte Y, Atasoy D, Sternson SM. AGRP neurons are sufficient to orchestrate feeding behavior rapidly and without training. Nat Neurosci 14: 351‐355, 2011. DOI: 10.1038/nn.2739.
 6.Avery JA. Against gustotopic representation in the human brain: There is no Cartesian Restaurant. Curr Opin Physiol., 2021. DOI: 10.1016/j.cophys.2021.01.005.
 7.Baez‐Santiago MA, Reid EE, Moran A, Maier JX, Marrero‐Garcia Y, Katz DB. Dynamic taste responses of parabrachial pontine neurons in awake rats. J Neurophysiol 115: 1314‐1323, 2016. DOI: 10.1152/jn.00311.2015.
 8.Bai L, Mesgarzadeh S, Ramesh KS, Huey EL, Liu Y, Gray LA, Aitken TJ, Chen Y, Beutler LR, Ahn JS, Madisen L, Zeng H, Krasnow MA, Knight ZA. Genetic identification of vagal sensory neurons that control feeding. Cell 179: 1129‐1143.e23, 2019. DOI: 10.1016/j.cell.2019.10.031.
 9.Baldwin MW, Toda Y, Nakagita T, O'Connell MJ, Klasing KC, Misaka T, Edwards SV, Liberles SD. Evolution of sweet taste perception in hummingbirds by transformation of the ancestral umami receptor. Science 345: 929‐933, 2014. DOI: 10.1126/science.1255097.
 10.Banik DD, Benfey ED, Martin LE, Kay KE, Loney GC, Nelson AR, Ahart ZC, Kemp BT, Kemp BR, Torregrossa A‐M, Medler KF. A subset of broadly responsive Type III taste cells contribute to the detection of bitter, sweet and umami stimuli. PLOS Genet 16: e1008925, 2020. DOI: 10.1371/journal.pgen.1008925.
 11.Barretto RPJ, Gillis‐Smith S, Chandrashekar J, Yarmolinsky DA, Schnitzer MJ, Ryba NJP, Zuker CS. The neural representation of taste quality at the periphery. Nature 517: 373‐376, 2015. DOI: 10.1038/nature13873.
 12.Bartel DL, Sullivan SL, Lavoie EG, Sévigny J, Finger TE. Nucleoside triphosphate diphosphohydrolase‐2 is the ecto‐ATPase of type I cells in taste buds. J Comp Neurol 497: 1‐12, 2006. DOI: 10.1002/cne.20954.
 13.Baumer‐Harrison C, Raymond MA, Myers TA, Sussman KM, Rynberg ST, Ugartechea AP, Lauterbach D, Mast TG, Breza JM. Optogenetic stimulation of Type I GAD65+ cells in taste buds activates gustatory neurons and drives appetitive licking behavior in sodium‐depleted mice. J Neurosci, 2020. DOI: 10.1523/JNEUROSCI.0597‐20.2020.
 14.Behrens M, Meyerhof W. Bitter taste receptor research comes of age: From characterization to modulation of TAS2Rs. Semin Cell Dev Biol 24: 215‐221, 2013. DOI: 10.1016/j.semcdb.2012.08.006.
 15. Behrens M, Meyerhof W. Gustatory and extragustatory functions of mammalian taste receptors. Physiol Behav 105(1): 4‐13, 2011. DOI: 10.1016/j.physbeh.2011.02.010.
 16.Beidler LM. Properties of chemoreceptors of tongue of rat. J Neurophysiol 16: 595‐607, 1953. DOI: 10.1152/jn.1953.16.6.595.
 17.Beidler LM. A theory of taste stimulation. J Gen Physiol 38: 133‐139, 1954.
 18.Beidler LM, Smallman RL. Renewal of cells within taste buds. J Cell Biol 27: 263‐272, 1965. DOI: 10.1083/jcb.27.2.263.
 19.Ben Abu N, Mason PE, Klein H, Dubovski N, Ben Shoshan‐Galeczki Y, Malach E, Pražienková V, Maletínská L, Tempra C, Chamorro VC, Cvačka J, Behrens M, Niv MY, Jungwirth P. Sweet taste of heavy water. Commun Biol 4: 1‐10, 2021. DOI: 10.1038/s42003‐021‐01964‐y.
 20.Betley JN, Xu S, Cao ZFH, Gong R, Magnus CJ, Yu Y, Sternson SM. Neurons for hunger and thirst transmit a negative‐valence teaching signal. Nature 521: 180‐185, 2015. DOI: 10.1038/nature14416.
 21.Beutler LR, Chen Y, Ahn JS, Lin Y‐C, Essner RA, Knight ZA. Dynamics of gut‐brain communication underlying hunger. Neuron 96: 461‐475.e5, 2017. DOI: 10.1016/j.neuron.2017.09.043.
 22.Beutler LR, Corpuz TV, Ahn JS, Kosar S, Song W, Chen Y, Knight ZA. Obesity causes selective and long‐lasting desensitization of AgRP neurons to dietary fat. eLife 9: e55909, 2020. DOI: 10.7554/eLife.55909.
 23.Bigiani A. Mouse taste cells with glialike membrane properties. J Neurophysiol 85: 1552‐1560, 2001. DOI: 10.1152/jn.2001.85.4.1552.
 24.Bigiani A. The origin of saltiness: Oral detection of NaCl. Curr Opin Physiol 19: 156‐161, 2021. DOI: 10.1016/j.cophys.2020.11.006.
 25.Boudreau JC, Oravec J, White TD, Madigan C, Chu S‐P. Geniculate neuralgia and facial nerve sensory systems. Arch Otolaryngol 103: 473‐481, 1977. DOI: 10.1001/archotol.1977.00780250067007.
 26.Breslin PAS. An evolutionary perspective on food and human taste. Curr Biol 23: R409‐R418, 2013. DOI: 10.1016/j.cub.2013.04.010.
 27.Breza JM, Contreras RJ. Anion size modulates salt taste in rats. J Neurophysiol 107: 1632‐1648, 2012. DOI: 10.1152/jn.00621.2011.
 28.Breza JM, Nikonov AA, Contreras RJ. Response latency to lingual taste stimulation distinguishes neuron types within the geniculate ganglion. J Neurophysiol 103: 1771‐1784, 2010. DOI: 10.1152/jn.00785.2009.
 29.Buchanan KL, Rupprecht LE, Sahasrabudhe A, Kaelberer MM, Klein M, Villalobos J, Liu WW, Yang A, Gelman J, Park S, Anikeeva P, Bohórquez DV. A gut sensor for sugar preference. BioRxiv, 2020. DOI: 10.1101/2020.03.06.981365.
 30.Byrnes N, Nestrud MA, Hayes JE. Perceptual mapping of chemesthetic stimuli in naïve assessors. Chemosens Percept 8: 19‐32, 2015. DOI: 10.1007/s12078‐015‐9178‐7.
 31.Cai H, Maudsley S, Martin B. What is the role of metabolic hormones in taste buds of the tongue. Gut Brain Control Metab 42: 134‐146, 2014. DOI: 10.1159/000358322.
 32.Caicedo A, Roper SD. Taste receptor cells that discriminate between bitter stimuli. Science 291: 1557‐1560, 2001. DOI: 10.1126/science.1056670.
 33.Cartoni C, Yasumatsu K, Ohkuri T, Shigemura N, Yoshida R, Godinot N, le Coutre J, Ninomiya Y, Damak S. Taste preference for fatty acids is mediated by GPR40 and GPR120. J Neurosci Off J Soc Neurosci 30: 8376‐8382, 2010. DOI: 10.1523/JNEUROSCI.0496‐10.2010.
 34.Castillo‐Azofeifa D, Losacco JT, Salcedo E, Golden EJ, Finger TE, Barlow LA. Sonic hedgehog from both nerves and epithelium is a key trophic factor for taste bud maintenance. Development 144: 3054‐3065, 2017. DOI: 10.1242/dev.150342.
 35.Caterina MJ, Schumacher MA, Tominaga M, Rosen TA, Levine JD, Julius D. The capsaicin receptor: A heat‐activated ion channel in the pain pathway. Nature 389: 816‐824, 1997. DOI: 10.1038/39807.
 36.Chandrashekar J, Kuhn C, Oka Y, Yarmolinsky DA, Hummler E, Ryba NJP, Zuker CS. The cells and peripheral representation of sodium taste in mice. Nature 464: 297‐301, 2010. DOI: 10.1038/nature08783.
 37.Chandrashekar J, Yarmolinsky D, von Buchholtz L, Oka Y, Sly W, Ryba NJP, Zuker CS. The taste of carbonation. Science 326: 443‐445, 2009. DOI: 10.1126/science.1174601.
 38.Chang RB, Waters H, Liman ER. A proton current drives action potentials in genetically identified sour taste cells. Proc Natl Acad Sci 107: 22320‐22325, 2010. DOI: 10.1073/pnas.1013664107.
 39.Chaudhari N, Landin AM, Roper SD. A metabotropic glutamate receptor variant functions as a taste receptor. Nat Neurosci 3: 113‐119, 2000. DOI: 10.1038/72053.
 40.Chen K, Kogan JF, Fontanini A. Spatially distributed representation of taste quality in the gustatory insular cortex of behaving mice. Curr Biol, 2020. DOI: 10.1016/j.cub.2020.10.014.
 41.Chen K, Kogan JF, Fontanini A. Spatially distributed representation of taste quality in the gustatory insular cortex of awake behaving mice. bioRxiv. 2020.07.01.183095, 2020. DOI: 10.1101/2020.07.01.183095.
 42.Chen X, Gabitto M, Peng Y, Ryba NJP, Zuker CS. A gustotopic map of taste qualities in the mammalian brain. Science 333: 1262‐1266, 2011. DOI: 10.1126/science.1204076.
 43.Chen Y, Lin Y‐C, Zimmerman CA, Essner RA, Knight ZA. Hunger neurons drive feeding through a sustained, positive reinforcement signal. eLife 5: e18640, 2016. DOI: 10.7554/eLife.18640.
 44.Chevrot M, Passilly‐Degrace P, Ancel D, Bernard A, Enderli G, Gomes M, Robin I, Issanchou S, Vergès B, Nicklaus S, Besnard P. Obesity interferes with the orosensory detection of long‐chain fatty acids in humans. Am J Clin Nutr 99: 975‐983, 2014. DOI: 10.3945/ajcn.113.077198.
 45.Cioni P, Strambini GB. Effect of heavy water on protein flexibility. Biophys J 82: 3246‐3253, 2002.
 46.Cui M, Jiang P, Maillet E, Max M, Margolskee RF, Osman R. The heterodimeric sweet taste receptor has multiple potential ligand binding sites. Curr Pharm Des 12: 4591‐4600, 2006. DOI: 10.2174/138161206779010350.
 47.D'Autréaux F, Coppola E, Hirsch M‐R, Birchmeier C, Brunet J‐F. Homeoprotein Phox2b commands a somatic‐to‐visceral switch in cranial sensory pathways. Proc Natl Acad Sci 108: 20018‐20023, 2011. DOI: 10.1073/pnas.1110416108.
 48.Dagan‐Wiener A, Di Pizio A, Nissim I, Bahia MS, Dubovski N, Margulis E, Niv MY. BitterDB: Taste ligands and receptors database in 2019. Nucleic Acids Res 47: D1179‐D1185, 2019. DOI: 10.1093/nar/gky974.
 49.Daly K, Al‐Rammahi M, Moran A, Marcello M, Ninomiya Y, Shirazi‐Beechey SP. Sensing of amino acids by the gut‐expressed taste receptor T1R1‐T1R3 stimulates CCK secretion. Am J Physiol Gastrointest Liver Physiol 304: G271‐G282, 2013. DOI: 10.1152/ajpgi.00074.2012.
 50.Damak S, Rong M, Yasumatsu K, Kokrashvili Z, Varadarajan V, Zou S, Jiang P, Ninomiya Y, Margolskee RF. Detection of sweet and umami taste in the absence of taste receptor T1r3. Science 301: 850‐853, 2003. DOI: 10.1126/science.1087155.
 51.Dando R, Dvoryanchikov G, Pereira E, Chaudhari N, Roper SD. Adenosine enhances sweet taste through A2B receptors in the taste bud. J Neurosci 32: 322‐330, 2012. DOI: 10.1523/JNEUROSCI.4070‐11.2012.
 52.Danilova V, Danilov Y, Roberts T, Tinti J‐M, Nofre C, Hellekant G. Sense of taste in a new world monkey, the common marmoset: Recordings from the chorda tympani and glossopharyngeal nerves. J Neurophysiol 88: 579‐594, 2002. DOI: 10.1152/jn.2002.88.2.579.
 53.Danilova V, Hellekant G, Tinti JM, Nofre C. Gustatory responses of the hamster Mesocricetus auratus to various compounds considered sweet by humans. J Neurophysiol 80: 2102‐2112, 1998. DOI: 10.1152/jn.1998.80.4.2102.
 54.David Dickman J, Smith DV. Response properties of fibers in the hamster superior laryngeal nerve. Brain Res 450: 25‐38, 1988. DOI: 10.1016/0006‐8993(88)91541‐7.
 55.Davidenko O, Darcel N, Fromentin G, Tomé D. Control of protein and energy intake ‐ brain mechanisms. Eur J Clin Nutr 67: 455‐461, 2013. DOI: 10.1038/ejcn.2013.73.
 56.de Araujo IE, Oliveira‐Maia AJ, Sotnikova TD, Gainetdinov RR, Caron MG, Nicolelis MAL, Simon SA. Food reward in the absence of taste receptor signaling. Neuron 57: 930‐941, 2008. DOI: 10.1016/j.neuron.2008.01.032.
 57.de Araujo IE, Schatzker M, Small DM. Rethinking food reward. Annu Rev Psychol 71: 139‐164, 2020. DOI: 10.1146/annurev‐psych‐122216‐011643.
 58.Di Lorenzo PM. Taste in the brain is encoded by sensorimotor state changes. Curr Opin Physiol 20: 39‐45, 2021. DOI: 10.1016/j.cophys.2020.12.003.
 59.Dolensek N, Gehrlach DA, Klein AS, Gogolla N. Facial expressions of emotion states and their neuronal correlates in mice. Science 368: 89‐94, 2020. DOI: 10.1126/science.aaz9468.
 60.Domingos AI, Vaynshteyn J, Voss HU, Ren X, Gradinaru V, Zang F, Deisseroth K, de Araujo IE, Friedman J. Leptin regulates the reward value of nutrient. Nat Neurosci 14: 1562‐1568, 2011. DOI: 10.1038/nn.2977.
 61.Doolin RE, Gilbertson TA. Distribution and characterization of functional amiloride‐sensitive sodium channels in rat tongue. J Gen Physiol 107: 545‐554, 1996. DOI: 10.1085/jgp.107.4.545.
 62.DuBois GE. Molecular mechanism of sweetness sensation. Physiol Behav 164: 453‐463, 2016. DOI: 10.1016/j.physbeh.2016.03.015.
 63.Dutta Banik D, Martin LE, Freichel M, Torregrossa A‐M, Medler KF. TRPM4 and TRPM5 are both required for normal signaling in taste receptor cells. Proc Natl Acad Sci U S A 115: E772‐E781, 2018. DOI: 10.1073/pnas.1718802115.
 64.Dvoryanchikov G, Hernandez D, Roebber JK, Hill DL, Roper SD, Chaudhari N. Transcriptomes and neurotransmitter profiles of classes of gustatory and somatosensory neurons in the geniculate ganglion. Nat Commun 8: 760, 2017. DOI: 10.1038/s41467‐017‐01095‐1.
 65.Dvoryanchikov G, Sinclair MS, Perea‐Martinez I, Wang T, Chaudhari N. Inward rectifier channel, ROMK, is localized to the apical tips of glial‐like cells in mouse taste buds. J Comp Neurol 517: spc1, 2009. DOI: 10.1002/cne.22202.
 66.Elliott EJ, Simon SA. The anion in salt taste: A possible role for paracellular pathways. Brain Res 535: 9‐17, 1990. DOI: 10.1016/0006‐8993(90)91817‐Z.
 67.Erickson RP. The evolution and implications of population and modular neural coding ideas. Progress in Brain Research 130: 9‐29, 2001. DOI: 10.1016/s0079-6123(01)30003-1. PMID: 11480291.
 68.Erickson RP. A study of the science of taste: On the origins and influence of the core ideas. Behav Brain Sci 31: 59‐75, 2008. DOI: 10.1017/S0140525X08003348.
 69.Farbman AI. Fine structure of the taste bud. J Ultrastruct Res 12: 328‐350, 1965. DOI: 10.1016/S0022‐5320(65)80103‐4.
 70.Farbman AI. Renewal of taste bud cells in rat circumvallate papillae. Cell Tissue Kinet 13: 349‐357, 1980. DOI: 10.1111/j.1365‐2184.1980.tb00474.x.
 71.Feng P, Zhao H. Complex evolutionary history of the vertebrate sweet/umami taste receptor genes. Chin Sci Bull 58: 2198‐2204, 2013. DOI: 10.1007/s11434‐013‐5811‐5.
 72.Fernandes AB, Alves da Silva J, Almeida J, Cui G, Gerfen CR, Costa RM, Oliveira‐Maia AJ. Postingestive modulation of food seeking depends on vagus‐mediated dopamine neuron activity. Neuron 106: 778‐788.e6, 2020. DOI: 10.1016/j.neuron.2020.03.009.
 73.Finger TE, Danilova V, Barrows J, Bartel DL, Vigers AJ, Stone L, Hellekant G, Kinnamon SC. ATP signaling is crucial for communication from taste buds to gustatory nerves. Science 310: 1495‐1499, 2005. DOI: 10.1126/science.1118435.
 74.Fletcher ML, Ogg MC, Lu L, Ogg RJ, Boughter JD. Overlapping representation of primary tastes in a defined region of the gustatory cortex. J Neurosci Off J Soc Neurosci 37: 7595‐7605, 2017. DOI: 10.1523/JNEUROSCI.0649‐17.2017.
 75.Fonseca E, de Lafuente V, Simon SA, Gutierrez R. Sucrose intensity coding and decision‐making in rat gustatory cortices. eLife 7, 2018. DOI: 10.7554/eLife.41152.
 76.Fonseca E, Sandoval‐Herrera V, Simon SA, Gutierrez R. Behavioral disassociation of perceived sweet taste intensity and hedonically positive palatability. eNeuro 7, 2020. DOI: 10.1523/ENEURO.0268‐20.2020.
 77.Friedman JM. Leptin and the endocrine control of energy balance. Nat Metab 1: 754‐764, 2019. DOI: 10.1038/s42255‐019‐0095‐y.
 78.Fu O, Iwai Y, Kondoh K, Misaka T, Minokoshi Y, Nakajima K. SatB2‐expressing neurons in the parabrachial nucleus encode sweet taste. Cell Rep 27: 1650‐1656.e4, 2019. DOI: 10.1016/j.celrep.2019.04.040.
 79.Fu O, Iwai Y, Narukawa M, Ishikawa AW, Ishii KK, Murata K, Yoshimura Y, Touhara K, Misaka T, Minokoshi Y, Nakajima K. Hypothalamic neuronal circuits regulating hunger‐induced taste modification. Nat Commun 10: 4560, 2019. DOI: 10.1038/s41467‐019‐12478‐x.
 80.Fukuwatari T, Kawada T, Tsuruta M, Hiraoka T, Iwanaga T, Sugimoto E, Fushiki T. Expression of the putative membrane fatty acid transporter (FAT) in taste buds of the circumvallate papillae in rats. FEBS Lett 414: 461‐464, 1997. DOI: 10.1016/s0014‐5793(97)01055‐7.
 81.Fushiki T, Kawai T. Chemical reception of fats in the oral cavity and the mechanism of addiction to dietary fat. Chem Senses 30 (Suppl 1): i184‐i185, 2005. DOI: 10.1093/chemse/bjh175.
 82.Gaillard D, Laugerette F, Darcel N, El‐Yassimi A, Passilly‐Degrace P, Hichami A, Khan NA, Montmayeur J‐P, Besnard P. The gustatory pathway is involved in CD36‐mediated orosensory perception of long‐chain fatty acids in the mouse. FASEB J Off Publ Fed Am Soc Exp Biol 22: 1458‐1468, 2008. DOI: 10.1096/fj.07‐8415com.
 83.Ganchrow JR, Steiner JE, Daher M. Neonatal facial expressions in response to different qualities and intensities of gustatory stimuli. Infant Behav Dev 6: 473‐484, 1983. DOI: 10.1016/S0163‐6383(83)90301‐6.
 84.Gao N, Lu M, Echeverri F, Laita B, Kalabat D, Williams ME, Hevezi P, Zlotnik A, Moyer BD. Voltage‐gated sodium channels in taste bud cells. BMC Neurosci 10: 20, 2009. DOI: 10.1186/1471‐2202‐10‐20.
 85.Garcia A, Coss A, Luis‐Islas J, Puron‐Sierra L, Luna M, Villavicencio M, Gutierrez R. Lateral hypothalamic GABAergic neurons encode and potentiate sucrose's palatability. Front Neurosci 14, 2021. DOI: 10.3389/fnins.2020.608047.
 86.Garcia J, Lasiter PS, Bermudez‐Rattoni F, Deems DA. A general theory of aversion learning. Ann N Y Acad Sci 443: 8‐21, 1985. DOI: 10.1111/j.1749‐6632.1985.tb27060.x.
 87.Geerling JC, Loewy AD. Central regulation of sodium appetite. Exp Physiol 93: 177‐209, 2008. DOI: 10.1113/expphysiol.2007.039891.
 88.Gilbertson TA, Liu L, Kim I, Burks CA, Hansen DR. Fatty acid responses in taste cells from obesity‐prone and ‐resistant rats. Physiol Behav 86: 681‐690, 2005. DOI: 10.1016/j.physbeh.2005.08.057.
 89.Gilbertson TA, Yu T, Shah BP. Gustatory Mechanisms for Fat Detection. In: Montmayeur JP, le Coutre J, editors. Fat Detection: Taste, Texture, and Post Ingestive Effects. CRC Press/Taylor & Francis, 2010. Chapter 3. Available from: https://www.ncbi.nlm.nih.gov/books/NBK53558/
 90.Goldstein N, McKnight AD, Carty JRE, Arnold M, Betley JN, Alhadeff AL. Hypothalamic detection of macronutrients via multiple gut‐brain pathways. Cell Metab, 2021. DOI: 10.1016/j.cmet.2020.12.018.
 91.Green BG. Chemesthesis: Pungency as a component of flavor. Trends Food Sci Technol 7: 415‐420, 1996. DOI: 10.1016/S0924‐2244(96)10043‐1.
 92.Gribble FM, Reimann F. Enteroendocrine cells: Chemosensors in the intestinal epithelium. Annu Rev Physiol 78: 277‐299, 2016. DOI: 10.1146/annurev‐physiol‐021115‐105439.
 93.Griffioen‐Roose S, Mars M, Siebelink E, Finlayson G, Tomé D, de Graaf C. Protein status elicits compensatory changes in food intake and food preferences. Am J Clin Nutr 95: 32‐38, 2012. DOI: 10.3945/ajcn.111.020503.
 94.Grill HJ, Berridge KC. Taste reactivity as a measure of the neural control of palatability. Prog Psychobiol Physiol Psychol 11: 1‐61, 1985.
 95.Grill HJ, Norgren R. The taste reactivity test. I. Mimetic responses to gustatory stimuli in neurologically normal rats. Brain Res 143: 263‐279, 1978. DOI: 10.1016/0006‐8993(78)90568‐1.
 96.Grill HJ, Norgren R. Chronically decerebrate rats demonstrate satiation but not bait shyness. Science 201: 267‐269, 1978. DOI: 10.1126/science.663655.
 97.Gutierrez R, Carmena JM, Nicolelis MAL, Simon SA. Orbitofrontal ensemble activity monitors licking and distinguishes among natural rewards. J Neurophysiol 95: 119‐133, 2006. DOI: 10.1152/jn.00467.2005.
 98.Gutierrez R, Fonseca E, Simon SA. The neuroscience of sugars in taste, gut‐reward, feeding circuits, and obesity. Cell Mol Life Sci 77: 3469‐3502, 2020. DOI: 10.1007/s00018‐020‐03458‐2.
 99.Gutierrez R, Simon SA, Nicolelis MAL. Licking‐induced synchrony in the taste–reward circuit improves cue discrimination during learning. J Neurosci 30: 287‐303, 2010. DOI: 10.1523/JNEUROSCI.0855‐09.2010.
 100.Hajnal A, Smith GP, Norgren R. Oral sucrose stimulation increases accumbens dopamine in the rat. Am J Physiol Regul Integr Comp Physiol 286: R31‐R37, 2004. DOI: 10.1152/ajpregu.00282.2003.
 101.Hamosh M, Scow RO. Lingual lipase and its role in the digestion of dietary lipid. J Clin Invest 52: 88‐95, 1973.
 102.Han J, Choi M. Comprehensive functional screening of taste sensation in vivo. bioRxiv 371682, 2018. DOI: 10.1101/371682.
 103.Han W, Tellez LA, Perkins MH, Perez IO, Qu T, Ferreira J, Ferreira TL, Quinn D, Liu Z‐W, Gao X‐B, Kaelberer MM, Bohórquez DV, Shammah‐Lagnado SJ, de Lartigue G, de Araujo IE. A neural circuit for gut‐induced reward. Cell 175: 665‐678.e23, 2018. DOI: 10.1016/j.cell.2018.08.049.
 104.Harper AE, Benevenga NJ, Wohlhueter RM. Effects of ingestion of disproportionate amounts of amino acids. Physiol Rev 50: 428‐558, 1970. DOI: 10.1152/physrev.1970.50.3.428.
 105.Heck GL, Mierson S, DeSimone JA. Salt taste transduction occurs through an amiloride‐sensitive sodium transport pathway. Science 223: 403‐405, 1984. DOI: 10.1126/science.6691151.
 106.Herness S, Zhao F‐L. The neuropeptides CCK and NPY and the changing view of cell‐to‐cell communication in the taste bud. Physiol Behav 97: 581‐591, 2009. DOI: 10.1016/j.physbeh.2009.02.043.
 107.Herness S, Zhao F‐L, Lu S, Kaya N, Shen T. Expression and physiological actions of cholecystokinin in rat taste receptor cells. J Neurosci Off J Soc Neurosci 22: 10018‐10029, 2002.
 108.Higgins SC, Gueorguiev M, Korbonits M. Ghrelin, the peripheral hunger hormone. Ann Med 39: 116‐136, 2007. DOI: 10.1080/07853890601149179.
 109.Holland VF, Zampighi GA, Simon SA. Morphology of fungiform papillae in canine lingual epithelium: Location of intercellular junctions in the epithelium. J Comp Neurol 279: 13‐27, 1989. DOI: 10.1002/cne.902790103.
 110.Holman GL. Intragastric reinforcement effect. J Comp Physiol Psychol 69: 432‐441, 1969. DOI: 10.1037/h0028233.
 111.Huang AL, Chen X, Hoon MA, Chandrashekar J, Guo W, Tränkner D, Ryba NJP, Zuker CS. The cells and logic for mammalian sour taste detection. Nature 442: 934‐938, 2006. DOI: 10.1038/nature05084.
 112.Huang AY, Wu SY. Substance P as a putative efferent transmitter mediates GABAergic inhibition in mouse taste buds. Br J Pharmacol 175: 1039‐1053, 2018. DOI: 10.1111/bph.14142.
 113.Huang L, Ledochowitsch P, Knoblich U, Lecoq J, Murphy GJ, Reid C, de Vries SEJ, Koch C, Zeng H, Buice MA, Waters J, Li L. Relationship between simultaneously recorded spiking activity and fluorescence signal in GCaMP6 transgenic mice. eLife 10, 2021. DOI: 10.7554/eLife.51675.
 114.Huang T, Ohman LC, Clements AV, Whiddon ZD, Krimm RF. Variable branching characteristics of peripheral taste neurons indicates differential convergence. bioRxiv 2020.08.20.260059, 2020. DOI: 10.1101/2020.08.20.260059.
 115.Huang YA, Maruyama Y, Stimac R, Roper SD. Presynaptic (Type III) cells in mouse taste buds sense sour (acid) taste. J Physiol 586: 2903‐2912, 2008. DOI: 10.1113/jphysiol.2008.151233.
 116.Huang YA, Roper SD. Intracellular Ca2+ and TRPM5‐mediated membrane depolarization produce ATP secretion from taste receptor cells. J Physiol 588: 2343‐2350, 2010. DOI: 10.1113/jphysiol.2010.191106.
 117.Ishimaru Y, Inada H, Kubota M, Zhuang H, Tominaga M, Matsunami H. Transient receptor potential family members PKD1L3 and PKD2L1 form a candidate sour taste receptor. Proc Natl Acad Sci U S A 103: 12569‐12574, 2006. DOI: 10.1073/pnas.0602702103.
 118.Jang H‐J, Kokrashvili Z, Theodorakis MJ, Carlson OD, Kim B‐J, Zhou J, Kim HH, Xu X, Chan SL, Juhaszova M, Bernier M, Mosinger B, Margolskee RF, Egan JM. Gut‐expressed gustducin and taste receptors regulate secretion of glucagon‐like peptide‐1. Proc Natl Acad Sci U S A 104: 15069‐15074, 2007. DOI: 10.1073/pnas.0706890104.
 119.Jarvie BC, Chen JY, King HO, Palmiter RD. Satb2 neurons in the parabrachial nucleus mediate taste perception. Nat Commun 12: 224, 2021. DOI: 10.1038/s41467‐020‐20100‐8.
 120.Jensterle M, DeVries JH, Battelino T, Battelino S, Yildiz B, Janez A. Glucagon‐like peptide‐1, a matter of taste? Rev Endocr Metab Disord, 2020. DOI: 10.1007/s11154‐020‐09609‐x.
 121.Jezzini A, Caruana F, Stoianov I, Gallese V, Rizzolatti G. Functional organization of the insula and inner perisylvian regions. Proc Natl Acad Sci 109: 10077‐10082, 2012. DOI: 10.1073/pnas.1200143109.
 122.Jezzini A, Mazzucato L, Camera GL, Fontanini A. Processing of hedonic and chemosensory features of taste in medial prefrontal and insular networks. J Neurosci 33: 18966‐18978, 2013. DOI: 10.1523/JNEUROSCI.2974‐13.2013.
 123.Jiang P, Josue J, Li X, Glaser D, Li W, Brand JG, Margolskee RF, Reed DR, Beauchamp GK. Major taste loss in carnivorous mammals. Proc Natl Acad Sci 109: 4956‐4961, 2012. DOI: 10.1073/pnas.1118360109.
 124.Jin H, Fishman ZH, Ye M, Wang L, Zuker CS. Top‐down control of sweet and bitter taste in the mammalian brain. Cell 184: 257‐271.e16, 2021. DOI: 10.1016/j.cell.2020.12.014.
 125.Julius D. TRP channels and pain. Annu Rev Cell Dev Biol 29: 355‐384, 2013. DOI: 10.1146/annurev‐cellbio‐101011‐155833.
 126.Kaelberer MM, Buchanan KL, Klein ME, Barth BB, Montoya MM, Shen X, Bohórquez DV. A gut‐brain neural circuit for nutrient sensory transduction. Science 361, 2018. DOI: 10.1126/science.aat5236.
 127.Kaelberer MM, Rupprecht LE, Liu WW, Weng P, Bohórquez DV. Neuropod cells: The emerging biology of gut‐brain sensory transduction. Annu Rev Neurosci 43: 337‐353, 2020. DOI: 10.1146/annurev‐neuro‐091619‐022657.
 128.Kataoka S, Yang R, Ishimaru Y, Matsunami H, Kinnamon JC, Finger TE. The candidate sour taste receptor, PKD2L1, is expressed by type III taste cells in the mouse. Chem Senses 33: 243‐254, 2008. DOI: 10.1093/chemse/bjm083.
 129.Katz DB, Simon SA, Nicolelis MAL. Taste‐specific neuronal ensembles in the gustatory cortex of awake rats. J Neurosci Off J Soc Neurosci 22: 1850‐1857, 2002.
 130.Kawai K, Sugimoto K, Nakashima K, Miura H, Ninomiya Y. Leptin as a modulator of sweet taste sensitivities in mice. Proc Natl Acad Sci 97: 11044‐11049, 2000. DOI: 10.1073/pnas.190066697.
 131.Keast RS, Costanzo A. Is fat the sixth taste primary? Evidence and implications. Flavour 4: 5, 2015. DOI: 10.1186/2044‐7248‐4‐5.
 132.Keller KL. Genetic influences on oral fat perception and preference. J Food Sci 77: S143‐S147, 2012. DOI: 10.1111/j.1750‐3841.2011.02585.x.
 133.Kim U‐K, Breslin PAS, Reed D, Drayna D. Genetics of human taste perception. J Dent Res 83: 448‐453, 2004. DOI: 10.1177/154405910408300603.
 134.Kulkarni BV, Mattes RD. Lingual lipase activity in the orosensory detection of fat by humans. Am J Physiol Regul Integr Comp Physiol 306: R879‐R885, 2014. DOI: 10.1152/ajpregu.00352.2013.
 135.Kusuhara Y, Yoshida R, Ohkuri T, Yasumatsu K, Voigt A, Hübner S, Maeda K, Boehm U, Meyerhof W, Ninomiya Y. Taste responses in mice lacking taste receptor subunit T1R1. J Physiol 591: 1967‐1985, 2013. DOI: 10.1113/jphysiol.2012.236604.
 136.Laffitte A, Neiers F, Briand L. Functional roles of the sweet taste receptor in oral and extraoral tissues. Curr Opin Clin Nutr Metab Care 17: 379‐385, 2014. DOI: 10.1097/MCO.0000000000000058.
 137.Lapis TJ, Penner MH, Lim J. Humans can taste glucose oligomers independent of the hT1R2/hT1R3 sweet taste receptor. Chem Senses 41: 755‐762, 2016. DOI: 10.1093/chemse/bjw088.
 138.Larson ED, Vandenbeuch A, Anderson CB, Kinnamon SC. Function, innervation, and neurotransmitter signaling in mice lacking Type‐II taste cells. eNeuro 7, 2020. DOI: 10.1523/ENEURO.0339‐19.2020.
 139.Laugerette F, Passilly‐Degrace P, Patris B, Niot I, Febbraio M, Montmayeur J‐P, Besnard P. CD36 involvement in orosensory detection of dietary lipids, spontaneous fat preference, and digestive secretions. J Clin Invest 115: 3177‐3184, 2005. DOI: 10.1172/JCI25299.
 140.Lawless HT, Skinner EZ. The duration and perceived intensity of sucrose taste. Percept Psychophys 25: 180‐184, 1979. DOI: 10.3758/BF03202983.
 141.Leijon SCM, Neves AF, Breza JM, Simon SA, Chaudhari N, Roper SD. Oral thermosensing by murine trigeminal neurons: Modulation by capsaicin, menthol and mustard oil. J Physiol 597: 2045‐2061, 2019. DOI: 10.1113/JP277385.
 142.Lemon CH, Margolskee RF. Contribution of the T1r3 taste receptor to the response properties of central gustatory neurons. J Neurophysiol 101: 2459‐2471, 2009. DOI: 10.1152/jn.90892.2008.
 143.Levitan D, Lin J‐Y, Wachutka J, Mukherjee N, Nelson SB, Katz DB. Single and population coding of taste in the gustatory cortex of awake mice. J Neurophysiol 122: 1342‐1356, 2019. DOI: 10.1152/jn.00357.2019.
 144.Li JX, Yoshida T, Monk KJ, Katz DB. Lateral hypothalamus contains two types of palatability‐related taste responses with distinct dynamics. J Neurosci 33: 9462‐9473, 2013. DOI: 10.1523/JNEUROSCI.3935‐12.2013.
 145.Li X, Li W, Wang H, Bayley DL, Cao J, Reed DR, Bachmanov AA, Huang L, Legrand‐Defretin V, Beauchamp GK, Brand JG. Cats lack a sweet taste receptor. J Nutr 136: 1932S‐1934S, 2006.
 146.Lim J, Pullicin AJ. Oral carbohydrate sensing: Beyond sweet taste. Physiol Behav 202: 14‐25, 2019. DOI: 10.1016/j.physbeh.2019.01.021.
 147.Liman ER, Kinnamon SC. Sour taste: Receptors, cells and circuits. Curr Opin Physiol 20: 8‐15, 2021. DOI: 10.1016/j.cophys.2020.12.006.
 148.Lin C, Colquitt L, Wise P, Breslin PAS, Rawson NE, Genovese F, Maina I, Joseph P, Fomuso L, Slade L, Brooks D, Miclo A, Hayes JE, Sullo A, Reed DR. Studies of human twins reveal genetic variation that affects dietary fat perception. Chem Senses 45: 467‐481, 2020. DOI: 10.1093/chemse/bjaa036.
 149.Liou AP, Chavez DI, Espero E, Hao S, Wank SA, Raybould HE. Protein hydrolysate‐induced cholecystokinin secretion from enteroendocrine cells is indirectly mediated by the intestinal oligopeptide transporter PepT1. Am J Physiol Gastrointest Liver Physiol 300: G895‐G902, 2011. DOI: 10.1152/ajpgi.00521.2010.
 150.Liou AP, Lu X, Sei Y, Zhao X, Pechhold S, Carrero RJ, Raybould HE, Wank S. The G‐protein‐coupled receptor GPR40 directly mediates long‐chain fatty acid‐induced secretion of cholecystokinin. Gastroenterology 140: 903‐912, 2011. DOI: 10.1053/j.gastro.2010.10.012.
 151.Liu Y, Xu H, Dahir N, Calder A, Lin F, Gilbertson TA (2021) GPR84 is Essential for the Taste of Medium Chain Saturated Fatty Acids. J Neurosci: JN-RM-2530-20.
 152.Liu B, Ha M, Meng X‐Y, Khaleduzzaman M, Zhang Z, Li X, Cui M. Functional characterization of the heterodimeric sweet taste receptor T1R2 and T1R3 from a New World monkey species (squirrel monkey) and its response to sweet‐tasting proteins. Biochem Biophys Res Commun 427: 431‐437, 2012. DOI: 10.1016/j.bbrc.2012.09.083.
 153.Liu H, Fontanini A. State dependency of chemosensory coding in the gustatory thalamus (VPMpc) of alert rats. J Neurosci 35: 15479‐15491, 2015. DOI: 10.1523/JNEUROSCI.0839‐15.2015.
 154.Liu P, Shah BP, Croasdell S, Gilbertson TA. Transient receptor potential channel Type M5 is essential for fat taste. J Neurosci 31: 8634‐8642, 2011. DOI: 10.1523/JNEUROSCI.6273‐10.2011.
 155.Livneh Y, Ramesh RN, Burgess CR, Levandowski KM, Madara JC, Fenselau H, Goldey GJ, Diaz VE, Jikomes N, Resch JM, Lowell BB, Andermann ML. Homeostatic circuits selectively gate food cue responses in insular cortex. Nature 546: 611‐616, 2017. DOI: 10.1038/nature22375.
 156.Loper HB, La Sala M, Dotson C, Steinle N. Taste perception, associated hormonal modulation, and nutrient intake. Nutr Rev 73: 83‐91, 2015. DOI: 10.1093/nutrit/nuu009.
 157.Lossow K, Hermans‐Borgmeyer I, Meyerhof W, Behrens M. Segregated expression of ENaC subunits in taste cells. Chem Senses 45: 235‐248, 2020. DOI: 10.1093/chemse/bjaa004.
 158.Lundy RF, Contreras RJ. Temperature and amiloride alter taste nerve responses to Na+, K+, and NH+4 salts in rats. Brain Res 744: 309‐317, 1997. DOI: 10.1016/S0006‐8993(96)01118‐3.
 159.Lundy RF, Contreras RJ. Gustatory neuron types in rat geniculate ganglion. J Neurophysiol 82: 2970‐2988, 1999. DOI: 10.1152/jn.1999.82.6.2970.
 160.Lyall V, Alam RI, Phan DQ, Ereso GL, Phan TH, Malik SA, Montrose MH, Chu S, Heck GL, Feldman GM, DeSimone JA. Decrease in rat taste receptor cell intracellular pH is the proximate stimulus in sour taste transduction. Am J Physiol Cell Physiol 281: C1005‐C1013, 2001. DOI: 10.1152/ajpcell.2001.281.3.C1005.
 161.Ma Z, Taruno A, Ohmoto M, Jyotaki M, Lim JC, Miyazaki H, Niisato N, Marunaka Y, Lee RJ, Hoff H, Payne R, Demuro A, Parker I, Mitchell CH, Henao‐Mejia J, Tanis JE, Matsumoto I, Tordoff MG, Foskett JK. CALHM3 is essential for rapid ion channel‐mediated purinergic neurotransmission of GPCR‐mediated tastes. Neuron 98: 547‐561.e10, 2018. DOI: 10.1016/j.neuron.2018.03.043.
 162.Martin B, Dotson CD, Shin Y‐K, Ji S, Drucker DJ, Maudsley S, Munger SD. Modulation of taste sensitivity by GLP‐1 signaling in taste buds. Ann N Y Acad Sci 1170: 98‐101, 2009. DOI: 10.1111/j.1749‐6632.2009.03920.x.
 163.Martin C, Passilly‐Degrace P, Chevrot M, Ancel D, Sparks SM, Drucker DJ, Besnard P. Lipid‐mediated release of GLP‐1 by mouse taste buds from circumvallate papillae: Putative involvement of GPR120 and impact on taste sensitivity. J Lipid Res 53: 2256‐2265, 2012. DOI: 10.1194/jlr.M025874.
 164.Martin C, Passilly‐Degrace P, Gaillard D, Merlin J‐F, Chevrot M, Besnard P. The lipid‐sensor candidates CD36 and GPR120 are differentially regulated by dietary lipids in mouse taste buds: Impact on spontaneous fat preference. PLoS One 6: e24014, 2011. DOI: 10.1371/journal.pone.0024014.
 165.Martin LJ, Lane AH, Samson KK, Sollars SI. Regenerative failure following rat neonatal chorda tympani transection is associated with geniculate ganglion cell loss and terminal field plasticity in the nucleus of the solitary tract. bioRxiv 503326, 2018. DOI: 10.1101/503326.
 166.Masuda T, Taguchi W, Sano A, Ohta K, Kitabatake N, Tani F. Five amino acid residues in cysteine‐rich domain of human T1R3 were involved in the response for sweet‐tasting protein, thaumatin. Biochimie 95: 1502‐1505, 2013. DOI: 10.1016/j.biochi.2013.01.010.
 167.Mattes RD. Oral detection of short‐, medium‐, and long‐chain free fatty acids in humans. Chem Senses 34: 145‐150, 2009. DOI: 10.1093/chemse/bjn072.
 168.Mattes RD. Taste, teleology and macronutrient intake. Curr Opin Physiol 19: 162‐167, 2021. DOI: 10.1016/j.cophys.2020.11.003.
 169.McCaughey SA. Taste‐evoked responses to sweeteners in the nucleus of the solitary tract differ between C57BL/6ByJ and 129P3/J mice. J Neurosci Off J Soc Neurosci 27: 35‐45, 2007. DOI: 10.1523/JNEUROSCI.3672‐06.2007.
 170.Merigo F, Benati D, Cristofoletti M, Osculati F, Sbarbati A. Glucose transporters are expressed in taste receptor cells. J Anat 219: 243‐252, 2011. DOI: 10.1111/j.1469‐7580.2011.01385.x.
 171.Miller IJ. Peripheral interactions among single papilla inputs to gustatory nerve fibers. J Gen Physiol 57: 1‐25, 1971.
 172.Miller IJ. Branched chorda tympani neurons and interactions among taste receptors. J Comp Neurol 158: 155‐166, 1974. DOI: 10.1002/cne.901580204.
 173.Moayedi Y, Duenas‐Bianchi LF, Lumpkin EA. Somatosensory innervation of the oral mucosa of adult and aging mice. Sci Rep 8, 2018. DOI: 10.1038/s41598‐018‐28195‐2.
 174.Montell C, Rubin GM. Molecular characterization of the Drosophila trp locus: A putative integral membrane protein required for phototransduction. Neuron 2: 1313‐1323, 1989. DOI: 10.1016/0896‐6273(89)90069‐x.
 175.Morais S. The physiology of taste in fish: Potential implications for feeding stimulation and gut chemical sensing. Rev Fish Sci Aquac 25: 133‐149, 2017. DOI: 10.1080/23308249.2016.1249279.
 176.Morales‐Lázaro SL, Simon SA, Rosenbaum T. The role of endogenous molecules in modulating pain through transient receptor potential vanilloid 1 (TRPV1). J Physiol 591: 3109‐3121, 2013. DOI: 10.1113/jphysiol.2013.251751.
 177.Morera E, De Petrocellis L, Morera L, Moriello AS, Nalli M, Di Marzo V, Ortar G. Synthesis and biological evaluation of [6]‐gingerol analogues as transient receptor potential channel TRPV1 and TRPA1 modulators. Bioorg Med Chem Lett 22: 1674‐1677, 2012. DOI: 10.1016/j.bmcl.2011.12.113.
 178.Murata Y, Yasuo T, Yoshida R, Obata K, Yanagawa Y, Margolskee RF, Ninomiya Y. Action potential–enhanced ATP release from taste cells through hemichannels. J Neurophysiol 104: 896‐901, 2010. DOI: 10.1152/jn.00414.2010.
 179.Murray RG, Murray A. The fine structure of the taste buds of rhesus and cynomalgus monkeys. Anat Rec 138: 211‐233, 1960. DOI: 10.1002/ar.1091380304.
 180.Nakamura Y, Sanematsu K, Ohta R, Shirosaki S, Koyano K, Nonaka K, Shigemura N, Ninomiya Y. Diurnal variation of human sweet taste recognition thresholds is correlated with plasma leptin levels. Diabetes 57: 2661‐2665, 2008. DOI: 10.2337/db07‐1103.
 181.Nelson G, Chandrashekar J, Hoon MA, Feng L, Zhao G, Ryba NJP, Zuker CS. An amino‐acid taste receptor. Nature 416: 199‐202, 2002. DOI: 10.1038/nature726.
 182.Nelson G, Hoon MA, Chandrashekar J, Zhang Y, Ryba NJ, Zuker CS. Mammalian sweet taste receptors. Cell 106: 381‐390, 2001. DOI: 10.1016/s0092‐8674(01)00451‐2.
 183.Nie Y, Vigues S, Hobbs JR, Conn GL, Munger SD. Distinct contributions of T1R2 and T1R3 taste receptor subunits to the detection of sweet stimuli. Curr Biol 15: 1948‐1952, 2005. DOI: 10.1016/j.cub.2005.09.037.
 184.Nilius B, Appendino G. Spices: The savory and beneficial science of pungency. Rev Physiol Biochem Pharmacol 164: 1‐76, 2013. DOI: 10.1007/112_2013_11.
 185.Ninomiya K. Natural occurrence. Food Rev Int 14: 177‐211, 1998. DOI: 10.1080/87559129809541157.
 186.Ninomiya Y, Imoto T. Gurmarin inhibition of sweet taste responses in mice. Am J Physiol‐Regul Integr Comp Physiol 268: R1019‐R1025, 1995. DOI: 10.1152/ajpregu.1995.268.4.R1019.
 187.Ninomiya Y, Sako N, Imai Y. Enhanced gustatory neural responses to sugars in the diabetic db/db mouse. Am J Physiol 269: R930‐R937, 1995. DOI: 10.1152/ajpregu.1995.269.4.R930.
 188.Nissenbaum JW, Sclafani A. Qualitative differences in polysaccharide and sugar tastes in the rat: A two‐carbohydrate taste model. Neurosci Biobehav Rev 11: 187‐196, 1987. DOI: 10.1016/S0149‐7634(87)80025‐8.
 189.Nomura K, Nakanishi M, Ishidate F, Iwata K, Taruno A. All‐electrical Ca2+‐independent signal transduction mediates attractive sodium taste in taste buds. Neuron 106: 816‐829.e6, 2020. DOI: 10.1016/j.neuron.2020.03.006.
 190.O'Connor EC, Kremer Y, Lefort S, Harada M, Pascoli V, Rohner C, Lüscher C. Accumbal D1R neurons projecting to lateral hypothalamus authorize feeding. Neuron 88: 553‐564, 2015. DOI: 10.1016/j.neuron.2015.09.038.
 191.Ogiso K, Shimizu Y, Watanabe K, Tonosaki K. Possible involvement of undissociated acid molecules in the acid response of the chorda tympani nerve of the rat. J Neurophysiol 83: 2776‐2779, 2000. DOI: 10.1152/jn.2000.83.5.2776.
 192.Ohla K, Yoshida R, Roper SD, Di Lorenzo PM, Victor JD, Boughter JD, Fletcher M, Katz DB, Chaudhari N. Recognizing taste: Coding patterns along the neural axis in mammals. Chem Senses 44: 237‐247, 2019. DOI: 10.1093/chemse/bjz013.
 193.Ohman‐Gault L, Huang T, Krimm R. The transcription factor Phox2b distinguishes between oral and non‐oral sensory neurons in the geniculate ganglion. J Comp Neurol 525: 3935‐3950, 2017. DOI: 10.1002/cne.24312.
 194.Oka Y, Butnaru M, von Buchholtz L, Ryba NJP, Zuker CS. High salt recruits aversive taste pathways. Nature 494: 472‐475, 2013. DOI: 10.1038/nature11905.
 195.Ootani S, Umezaki T, Shin T, Murata Y. Convergence of afferents from the SLN and GPN in cat medullary swallowing neurons. Brain Res Bull 37: 397‐404, 1995. DOI: 10.1016/0361‐9230(95)00018‐6.
 196.Passilly‐Degrace P, Chevrot M, Bernard A, Ancel D, Martin C, Besnard P. Is the taste of fat regulated? Biochimie 96: 3‐7, 2014. DOI: 10.1016/j.biochi.2013.07.029.
 197.Peng Y, Gillis‐Smith S, Jin H, Tränkner D, Ryba NJP, Zuker CS. Sweet and bitter taste in the brain of awake behaving animals. Nature 527: 512‐515, 2015. DOI: 10.1038/nature15763.
 198.Perea‐Martinez I, Nagai T, Chaudhari N. Functional cell types in taste buds have distinct longevities. PLoS One 8: e53399, 2013. DOI: 10.1371/journal.pone.0053399.
 199.Pérez CA, Huang L, Rong M, Kozak JA, Preuss AK, Zhang H, Max M, Margolskee RF. A transient receptor potential channel expressed in taste receptor cells. Nat Neurosci 5: 1169, 2002. DOI: 10.1038/nn952.
 200.Perez IO, Villavicencio M, Simon SA, Gutierrez R. Speed and accuracy of taste identification and palatability: Impact of learning, reward expectancy, and consummatory licking. Am J Physiol‐Regul Integr Comp Physiol 305: R252‐R270, 2013. DOI: 10.1152/ajpregu.00492.2012.
 201.Pfaffmann C. Gustatory afferent impulses. J Cell Comp Physiol 17: 243‐258, 1941. DOI: 10.1002/jcp.1030170209.
 202.Prado L, Luis‐Islas J, Sandoval OI, Puron L, Gil MM, Luna A, Arias‐García MA, Galarraga E, Simon SA, Gutierrez R. Activation of glutamatergic fibers in the anterior NAc shell modulates reward activity in the aNAcSh, the lateral hypothalamus, and medial prefrontal cortex and transiently stops feeding. J Neurosci 36: 12511‐12529, 2016. DOI: 10.1523/JNEUROSCI.1605‐16.2016.
 203.Prescott J, Stevenson RJ. Pungency in food perception and preference. Food Rev Int 11: 665‐698, 1995. DOI: 10.1080/87559129509541064.
 204.Pullicin AJ, Penner MH, Lim J. Human taste detection of glucose oligomers with low degree of polymerization. PLoS One 12: e0183008, 2017. DOI: 10.1371/journal.pone.0183008.
 205.Pumplin DW, Yu C, Smith DV. Light and dark cells of rat vallate taste buds are morphologically distinct cell types. J Comp Neurol 378: 389‐410, 1997. DOI: 10.1002/(SICI)1096‐9861(19970217)378:3<389::AID‐CNE7>3.0.CO;2‐#.
 206.Qu T, Han W, Niu J, Tong J, de Araujo IE. On the roles of the Duodenum and the Vagus nerve in learned nutrient preferences. Appetite 139: 145‐151, 2019. DOI: 10.1016/j.appet.2019.04.014.
 207.Ramsey IS, Delling M, Clapham DE. An introduction to TRP channels. Annu Rev Physiol 68: 619‐647, 2006. DOI: 10.1146/annurev.physiol.68.040204.100431.
 208.Ramsey IS, DeSimone JA. Otopetrin‐1: A sour‐tasting proton channel. J Gen Physiol 150: 379‐382, 2018. DOI: 10.1085/jgp.201812003.
 209.Rehnberg BG, MacKinnon BI, Hettinger TP, Frank ME. Anion modulation of taste responses in sodium‐sensitive neurons of the hamster chorda tympani nerve. J Gen Physiol 101: 453‐465, 1993. DOI: 10.1085/jgp.101.3.453.
 210.Reimann F, Habib AM, Tolhurst G, Parker HE, Rogers GJ, Gribble FM. Glucose sensing in L cells: A primary cell study. Cell Metab 8: 532‐539, 2008. DOI: 10.1016/j.cmet.2008.11.002.
 211.Ren X, Tellez LA, de Araujo IE. Enantiomer‐specific selection of amino acids. Amino Acids 45: 1353‐1364, 2013. DOI: 10.1007/s00726‐013‐1595‐9.
 212.Roebber JK, Roper SD, Chaudhari N. The role of the anion in salt (NaCl) detection by mouse taste buds. J Neurosci Off J Soc Neurosci 39: 6224‐6232, 2019. DOI: 10.1523/JNEUROSCI.2367‐18.2019.
 213.Roland B, Deneux T, Franks KM, Bathellier B, Fleischmann A. Odor identity coding by distributed ensembles of neurons in the mouse olfactory cortex. eLife 6: e26337, 2017. DOI: 10.7554/eLife.26337.
 214.Rolls ET. The texture and taste of food in the brain. J Texture Stud 51: 23‐44, 2020. DOI: 10.1111/jtxs.12488.
 215.Rolls ET, Critchley HD, Browning AS, Hernadi I, Lenard L. Responses to the sensory properties of fat of neurons in the primate orbitofrontal cortex. J Neurosci 19: 1532‐1540, 1999. DOI: 10.1523/JNEUROSCI.19‐04‐01532.1999.
 216.Roper SD. Signal transduction and information processing in mammalian taste buds. Pflugers Arch 454: 759‐776, 2007. DOI: 10.1007/s00424‐007‐0247‐x.
 217.Roper SD. TRPs in taste and chemesthesis. In: Nilius B, Flockerzi V, editors. Mammalian Transient Receptor Potential (TRP) Cation Channels. Cham: Springer International Publishing, p. 827‐871, 2014.
 218.Roper SD. 3.09 ‐ Microphysiology of taste buds. In: Fritzsch B, editor. The Senses: A Comprehensive Reference (2nd ed). Oxford: Elsevier, 2020, p. 187‐210.
 219.Roper SD. Chemical and electrical synaptic interactions among taste bud cells. Curr Opin Physiol 20: 118‐125, 2021. DOI: 10.1016/j.cophys.2020.12.004.
 220.Rosenblueth A, Wiener N, Bigelow J. Behavior, purpose and teleology. Philos Sci 10: 18‐24, 1943. DOI: 10.1086/286788.
 221.Roussin AT, D'Agostino AE, Fooden AM, Victor JD, Lorenzo PMD. Taste coding in the nucleus of the solitary tract of the awake, freely licking rat. J Neurosci 32: 10494‐10506, 2012. DOI: 10.1523/JNEUROSCI.1856‐12.2012.
 222.Royer SM, Kinnamon JC. Application of serial sectioning and three‐dimensional reconstruction to the study of taste bud ultrastructure and organization. Microsc Res Tech 29: 381‐407, 1994. DOI: 10.1002/jemt.1070290508.
 223.Russek M. Demonstration of the influence of an hepatic glucosensitive mechanism on food‐intake. Physiol Behav 5: 1207‐1209, 1970. DOI: 10.1016/0031‐9384(70)90218‐0.
 224.Sadacca BF, Rothwax JT, Katz DB. Sodium concentration coding gives way to evaluative coding in cortex and amygdala. J Neurosci 32: 9999‐10011, 2012. DOI: 10.1523/JNEUROSCI.6059‐11.2012.
 225.Salazar H, Llorente I, Jara‐Oseguera A, García‐Villegas R, Munari M, Gordon SE, Islas LD, Rosenbaum T. A single N‐terminal cysteine in TRPV1 determines activation by pungent compounds from onion and garlic. Nat Neurosci 11: 255‐261, 2008. DOI: 10.1038/nn2056.
 226.Sammons JD, Weiss MS, Victor JD, Di Lorenzo PM. Taste coding of complex naturalistic taste stimuli and traditional taste stimuli in the parabrachial pons of the awake, freely licking rat. J Neurophysiol 116: 171‐182, 2016. DOI: 10.1152/jn.01119.2015.
 227.Samuelsen CL, Gardner MPH, Fontanini A. Thalamic contribution to cortical processing of taste and expectation. J Neurosci Off J Soc Neurosci 33: 1815‐1827, 2013. DOI: 10.1523/JNEUROSCI.4026‐12.2013.
 228.San Gabriel A, Maekawa T, Uneyama H, Torii K. Metabotropic glutamate receptor type 1 in taste tissue. Am J Clin Nutr 90: 743S‐746S, 2009. DOI: 10.3945/ajcn.2009.27462I.
 229.Santos‐Hernández M, Miralles B, Amigo L, Recio I. Intestinal signaling of proteins and digestion‐derived products relevant to satiety. J Agric Food Chem 66: 10123‐10131, 2018. DOI: 10.1021/acs.jafc.8b02355.
 230.Sclafani A. The sixth taste? Appetite 43: 1‐3, 2004. DOI: 10.1016/j.appet.2004.03.007.
 231.Sclafani A, Ackroff K, Abumrad NA. CD36 gene deletion reduces fat preference and intake but not post‐oral fat conditioning in mice. Am J Physiol‐Regul Integr Comp Physiol 293: R1823‐R1832, 2007. DOI: 10.1152/ajpregu.00211.2007.
 232.Sclafani A, Ackroff K, Schwartz GJ. Selective effects of vagal deafferentation and celiac–superior mesenteric ganglionectomy on the reinforcing and satiating action of intestinal nutrients. Physiol Behav 78: 285‐294, 2003. DOI: 10.1016/S0031‐9384(02)00968‐X.
 233.Sclafani A, Koepsell H, Ackroff K. SGLT1 sugar transporter/sensor is required for post‐oral glucose appetition. Am J Physiol‐Regul Integr Comp Physiol 310: R631‐R639, 2016. DOI: 10.1152/ajpregu.00432.2015.
 234.Sclafani A, Marambaud P, Ackroff K. Sucrose‐conditioned flavor preferences in sweet ageusic T1r3 and Calhm1 knockout mice. Physiol Behav 126: 25‐29, 2014. DOI: 10.1016/j.physbeh.2013.12.003.
 235.Sclafani A, Zukerman S, Glendinning JI, Margolskee RF. Fat and carbohydrate preferences in mice. Am J Physiol Regul Integr Comp Physiol 293: R1504‐R1513, 2007. DOI: 10.1152/ajpregu.00364.2007.
 236.Sclafani A, Nissenbaum JW, Vigorito M. Starch preference in rats. Neurosci Biobehav Rev 11: 253‐262, 1987.
 237.Servant G, Tachdjian C, Li X, Karanewsky DS. The sweet taste of true synergy: Positive allosteric modulation of the human sweet taste receptor. Trends Pharmacol Sci 32: 631‐636, 2011. DOI: 10.1016/j.tips.2011.06.007.
 238.Shin Y‐K, Martin B, Golden E, Dotson CD, Maudsley S, Kim W, Jang H‐J, Mattson MP, Drucker DJ, Egan JM, Munger SD. Modulation of taste sensitivity by GLP‐1 signaling. J Neurochem 106: 455‐463, 2008. DOI: 10.1111/j.1471‐4159.2008.05397.x.
 239.Shin Y‐K, Martin B, Kim W, White CM, Ji S, Sun Y, Smith RG, Sévigny J, Tschöp MH, Maudsley S, Egan JM. Ghrelin is produced in taste cells and ghrelin receptor null mice show reduced taste responsivity to salty (NaCl) and sour (citric acid) tastants. PLoS One 5: e12729, 2010. DOI: 10.1371/journal.pone.0012729.
 240.Sigoillot M, Brockhoff A, Meyerhof W, Briand L. Sweet‐taste‐suppressing compounds: Current knowledge and perspectives of application. Appl Microbiol Biotechnol 96: 619‐630, 2012. DOI: 10.1007/s00253‐012‐4387‐3.
 241.Simon SA, Gutierrez R. TRP channels at the periphery of the taste and trigeminal systems. In: Emir TLR, editor. Neurobiology of TRP Channels. Boca Raton (FL): CRC Press/Taylor & Francis, 2017.
 242.Sinclair MS, Perea‐Martinez I, Abouyared M, St. John SJ, Chaudhari N. Oxytocin decreases sweet taste sensitivity in mice. Physiol Behav 141: 103‐110, 2015. DOI: 10.1016/j.physbeh.2014.12.048.
 243.Smith DV, Ossebaard CA. Amiloride suppression of the taste intensity of sodium chloride: Evidence from direct magnitude scaling. Physiol Behav 57: 773‐777, 1995. DOI: 10.1016/0031‐9384(94)00329‐7.
 244.Soares ES, Stapleton JR, Rodriguez A, Fitzsimmons N, Oliveira L, Nicolelis MAL, Simon SA. Behavioral and neural responses to gustatory stimuli delivered non‐contingently through intra‐oral cannulas. Physiol Behav 92: 629‐642, 2007. DOI: 10.1016/j.physbeh.2007.05.038.
 245.Spector AC. Behavioral analyses of taste function and ingestion in rodent models. Physiol Behav 152: 516‐526, 2015. DOI: 10.1016/j.physbeh.2015.04.026.
 246.Spector AC, Guagliardo NA, St. John SJ. Amiloride disrupts NaCl versus KCl discrimination performance: Implications for salt taste coding in rats. J Neurosci 16: 8115‐8122, 1996. DOI: 10.1523/JNEUROSCI.16‐24‐08115.1996.
 247.Spector AC, Klumpp PA, Kaplan JM. Analytical issues in the evaluation of food deprivation and sucrose concentration effects on the microstructure of licking behavior in the rat. Behav Neurosci 112: 678‐694, 1998. DOI: 10.1037//0735‐7044.112.3.678.
 248.Spector AC, Kopka SL. Rats fail to discriminate quinine from denatonium: Implications for the neural coding of bitter‐tasting compounds. J Neurosci 22: 1937‐1941, 2002. DOI: 10.1523/JNEUROSCI.22‐05‐01937.2002.
 249.Spector AC, Smith JC. A detailed analysis of sucrose drinking in the rat. Physiol Behav 33: 127‐136, 1984. DOI: 10.1016/0031‐9384(84)90023‐4.
 250.Spector AC, Travers SP. The representation of taste quality in the mammalian nervous system. Behav Cogn Neurosci Rev 4: 143‐191, 2005. DOI: 10.1177/1534582305280031.
 251.Stähler F, Riedel K, Demgensky S, Neumann K, Dunkel A, Täubert A, Raab B, Behrens M, Raguse J‐D, Hofmann T, Meyerhof W. A role of the epithelial sodium channel in human salt taste transduction? Chemosens Percept 1: 78‐90, 2008. DOI: 10.1007/s12078‐008‐9006‐4.
 252.Stapleton JR, Lavine ML, Wolpert RL, Nicolelis MAL, Simon SA. Rapid taste responses in the gustatory cortex during licking. J Neurosci 26: 4126‐4138, 2006. DOI: 10.1523/JNEUROSCI.0092‐06.2006.
 253.Steiner JE. The gustofacial response: Observation on normal and anencephalic newborn infants. Symp Oral Sens Percept: 254‐278, 1973.
 254.Stratford J, Larson E, Yang R, Salcedo E, Finger T. 5‐HT3a‐driven GFP delineates gustatory fibers innervating sour‐responsive taste cells: A labeled line for sour taste? J Comp Neurol 525: 2358‐2375, 2017. DOI: 10.1002/cne.24209.
 255.Stratford JM, Contreras RJ. Peripheral gustatory processing of free fatty acids. In: Montmayeur J‐P, le Coutre J, editors. Fat Detection: Taste, Texture, and Post Ingestive Effects. Boca Raton (FL): CRC Press/Taylor & Francis, 2010.
 256.Su Z, Alhadeff AL, Betley JN. Nutritive, post‐ingestive signals are the primary regulators of AgRP neuron activity. Cell Rep 21: 2724‐2736, 2017. DOI: 10.1016/j.celrep.2017.11.036.
 257.Sudo S, Sudo M, Simons CT, Dessirier JM, Carstens E. Sensitization of trigeminal caudalis neuronal responses to intraoral acid and salt stimuli and desensitization by nicotine. Pain 98: 277‐286, 2002. DOI: 10.1016/s0304‐3959(02)00025‐8.
 258.Sukumaran SK, Yee KK, Iwata S, Kotha R, Quezada‐Calvillo R, Nichols BL, Mohan S, Pinto BM, Shigemura N, Ninomiya Y, Margolskee RF. Taste cell‐expressed α‐glucosidase enzymes contribute to gustatory responses to disaccharides. Proc Natl Acad Sci 113: 6035‐6040, 2016. DOI: 10.1073/pnas.1520843113.
 259.Sundaresan S, Abumrad NA. Dietary lipids inform the gut and brain about meal arrival via CD36‐mediated signal transduction. J Nutr 145: 2195‐2200, 2015. DOI: 10.3945/jn.115.215483.
 260.Sundaresan S, Shahid R, Riehl TE, Chandra R, Nassir F, Stenson WF, Liddle RA, Abumrad NA. CD36‐dependent signaling mediates fatty acid‐induced gut release of secretin and cholecystokinin. FASEB J Off Publ Fed Am Soc Exp Biol 27: 1191‐1202, 2013. DOI: 10.1096/fj.12‐217703.
 261.Takaishi M, Fujita F, Uchida K, Yamamoto S, Shimizu MS, Uotsu CH, Shimizu M, Tominaga M. 1,8‐cineole, a TRPM8 agonist, is a novel natural antagonist of human TRPA1. Mol Pain 8: 86, 2012. DOI: 10.1186/1744‐8069‐8‐86.
 262.Tan H‐E, Sisti AC, Jin H, Vignovich M, Villavicencio M, Tsang KS, Goffer Y, Zuker CS. The gut–brain axis mediates sugar preference. Nature 580: 511‐516, 2020. DOI: 10.1038/s41586‐020‐2199‐7.
 263.Taruno A, Vingtdeux V, Ohmoto M, Ma Z, Dvoryanchikov G, Li A, Adrien L, Zhao H, Leung S, Abernethy M, Koppel J, Davies P, Civan MM, Chaudhari N, Matsumoto I, Hellekant G, Tordoff MG, Marambaud P, Foskett JK. CALHM1 ion channel mediates purinergic neurotransmission of sweet, bitter and umami tastes. Nature 495: 223‐226, 2013. DOI: 10.1038/nature11906.
 264.Tellez LA, Ferreira JG, Medina S, Land BB, DiLeone RJ, de Araujo IE. Flavor‐independent maintenance, extinction and reinstatement of fat self‐administration in mice. Biol Psychiatry 73: 851‐859, 2013. DOI: 10.1016/j.biopsych.2013.02.028.
 265.Tellez LA, Han W, Zhang X, Ferreira TL, Perez IO, Shammah‐Lagnado SJ, van den Pol AN, de Araujo IE. Separate circuitries encode the hedonic and nutritional values of sugar. Nat Neurosci 19: 465‐470, 2016. DOI: 10.1038/nn.4224.
 266.Teng B, Wilson CE, Tu Y‐H, Joshi NR, Kinnamon SC, Liman ER. Cellular and neural responses to sour stimuli require the proton channel Otop1. Curr Biol 29: 3647‐3656.e5, 2019. DOI: 10.1016/j.cub.2019.08.077.
 267.Travers JB, Dinardo LA, Karimnamazi H. Motor and premotor mechanisms of licking. Neurosci Biobehav Rev 21: 631‐647, 1997. DOI: 10.1016/S0149‐7634(96)00045‐0.
 268.Treesukosol Y, Smith KR, Spector AC. Behavioral evidence for a glucose polymer taste receptor that is independent of the T1R2+3 heterodimer in a mouse model. J Neurosci 31: 13527‐13534, 2011. DOI: 10.1523/JNEUROSCI.2179‐11.2011.
 269.Tu Y‐H, Cooper AJ, Teng B, Chang RB, Artiga DJ, Turner HN, Mulhall EM, Ye W, Smith AD, Liman ER. An evolutionarily conserved gene family encodes proton‐selective ion channels. Science 359: 1047‐1050, 2018. DOI: 10.1126/science.aao3264.
 270.Vandenbeuch A, Clapp TR, Kinnamon SC. Amiloride‐sensitive channels in type I fungiform taste cells in mouse. BMC Neurosci 9: 1, 2008. DOI: 10.1186/1471‐2202‐9‐1.
 271.Vandenbeuch A, Kinnamon SC. Why do taste cells generate action potentials? J Biol 8: 42, 2009. DOI: 10.1186/jbiol138.
 272.Verhagen JV, Giza BK, Scott TR. Responses to taste stimulation in the ventroposteromedial nucleus of the thalamus in rats. J Neurophysiol 89: 265‐275, 2003. DOI: 10.1152/jn.00870.2001.
 273.Villavicencio M, Moreno MG, Simon SA, Gutierrez R. Encoding of sucrose's palatability in the nucleus accumbens shell and its modulation by exteroceptive auditory cues. Front Neurosci 12, 2018. DOI: 10.3389/fnins.2018.00265.
 274.Vogt MB, Mistretta CM. Convergence in mammalian nucleus of solitary tract during development and functional differentiation of salt taste circuits. J Neurosci Off J Soc Neurosci 10: 3148‐3157, 1990.
 275.Voigt N, Stein J, Galindo MM, Dunkel A, Raguse J‐D, Meyerhof W, Hofmann T, Behrens M. The role of lipolysis in human orosensory fat perception. J Lipid Res 55: 870‐882, 2014. DOI: 10.1194/jlr.M046029.
 276.von Soemmerring ST. Abbildungen der menschlichen Organe des Geruches. Edition Medicina Rara, 1809.
 277.Wallroth R, Ohla K. As soon as you taste it: Evidence for sequential and parallel processing of gustatory information. eNeuro 5, 2018. DOI: 10.1523/ENEURO.0269‐18.2018.
 278.Wang GJ, Volkow ND, Logan J, Pappas NR, Wong CT, Zhu W, Netusil N, Fowler JS. Brain dopamine and obesity. Lancet Lond Engl 357: 354‐357, 2001. DOI: 10.1016/s0140‐6736(00)03643‐6.
 279.Wang L, Gillis‐Smith S, Peng Y, Zhang J, Chen X, Daniel Salzman C, Ryba NJP, Zuker CS. The coding of valence and identity in the mammalian taste system. Nature 558: 127‐131, 2018. DOI: 10.1038/s41586‐018‐0165‐4.
 280.Wang Y, Erickson RP, Simon SA. Selectivity of lingual nerve fibers to chemical stimuli. J Gen Physiol. 101: 843‐866, 1993. DOI: 10.1085/JGP.101.6.843.
 281.White BD, He B, Dean RG, Martin RJ. Low protein diets increase neuropeptide Y gene expression in the basomedial hypothalamus of rats. J Nutr 124: 1152‐1160, 1994. DOI: 10.1093/jn/124.8.1152.
 282.White TD, Boudreau JC. Taste preferences of the cat for neurophysiologically active compounds. Physiol Psychol 3: 405‐410, 1975. DOI: 10.3758/BF03326850.
 283.Williams EK, Chang RB, Strochlic DE, Umans BD, Lowell BB, Liberles SD. Sensory neurons that detect stretch and nutrients in the digestive system. Cell 166: 209‐221, 2016. DOI: 10.1016/j.cell.2016.05.011.
 284.Wilson CE, Vandenbeuch A, Kinnamon SC. Physiological and behavioral responses to optogenetic stimulation of PKD2L1+ Type III taste cells. eNeuro 6, 2019. DOI: 10.1523/ENEURO.0107‐19.2019.
 285.Witt M. Chapter 10 ‐ Anatomy and development of the human taste system. In: Doty RL, editor. Handbook of Clinical Neurology. Elsevier, 2019, p. 147‐171.
 286.Wong F, Schaefer EL, Roop BC, LaMendola JN, Johnson‐Seaton D, Shao D. Proper function of the Drosophila trp gene product during pupal development is important for normal visual transduction in the adult. Neuron 3: 81‐94, 1989. DOI: 10.1016/0896‐6273(89)90117‐7.
 287.Wu A, Dvoryanchikov G, Pereira E, Chaudhari N, Roper SD. Breadth of tuning in taste afferent neurons varies with stimulus strength. Nat Commun 6: 8171, 2015. DOI: 10.1038/ncomms9171.
 288.Yang AK, Mendoza JA, Lafferty CK, Lacroix F, Britt JP. Hippocampal input to the nucleus accumbens shell enhances food palatability. Biol Psychiatry 87: 597‐608, 2020. DOI: 10.1016/j.biopsych.2019.09.007.
 289.Yang R, Dzowo YK, Wilson CE, Russell RL, Kidd GJ, Salcedo E, Lasher RS, Kinnamon JC, Finger TE. Three‐dimensional reconstructions of mouse circumvallate taste buds using serial blockface scanning electron microscopy: I. Cell types and the apical region of the taste bud. bioRxiv 610410, 2019. DOI: 10.1101/610410.
 290.Yang R, Dzowo YK, Wilson CE, Russell RL, Kidd GJ, Salcedo E, Lasher RS, Kinnamon JC, Finger TE. Three‐dimensional reconstructions of mouse circumvallate taste buds using serial blockface scanning electron microscopy: I. Cell types and the apical region of the taste bud. J Comp Neurol 528: 756‐771, 2020. DOI: 10.1002/cne.24779.
 291.Yarmolinsky DA, Zuker CS, Ryba NJP. Common sense about taste: From mammals to insects. Cell 139: 234‐244, 2009. DOI: 10.1016/j.cell.2009.10.001.
 292.Yasumatsu K, Iwata S, Inoue M, Ninomiya Y. Fatty acid taste quality information via GPR120 in the anterior tongue of mice. Acta Physiol Oxf Engl 226: e13215, 2019. DOI: 10.1111/apha.13215.
 293.Yasumatsu K, Ohkuri T, Yoshida R, Iwata S, Margolskee RF, Ninomiya Y. Sodium‐glucose cotransporter 1 as a sugar taste sensor in mouse tongue. Acta Physiol: e13529, 2020. DOI: 10.1111/apha.13529.
 294.Ye Q, Heck GL, DeSimone JA. The anion paradox in sodium taste reception: Resolution by voltage‐clamp studies. Science 254: 724‐726, 1991. DOI: 10.1126/science.1948054.
 295.Ye W, Chang RB, Bushman JD, Tu Y‐H, Mulhall EM, Wilson CE, Cooper AJ, Chick WS, Hill‐Eubanks DC, Nelson MT, Kinnamon SC, Liman ER. The K+ channel KIR2.1 functions in tandem with proton influx to mediate sour taste transduction. Proc Natl Acad Sci 113: E229‐E238, 2016. DOI: 10.1073/pnas.1514282112.
 296.Yee KK, Sukumaran SK, Kotha R, Gilbertson TA, Margolskee RF. Glucose transporters and ATP‐gated K+ (KATP) metabolic sensors are present in type 1 taste receptor 3 (T1r3)‐expressing taste cells. Proc Natl Acad Sci 108: 5431‐5436, 2011. DOI: 10.1073/pnas.1100495108.
 297.Yokota Y, Bradley RM. Receptive field size, chemical and thermal responses, and fiber conduction velocity of rat chorda tympani geniculate ganglion neurons. J Neurophysiol 115: 3062‐3072, 2016. DOI: 10.1152/jn.00045.2016.
 298.Yoshida R, Miyauchi A, Yasuo T, Jyotaki M, Murata Y, Yasumatsu K, Shigemura N, Yanagawa Y, Obata K, Ueno H, Margolskee RF, Ninomiya Y. Discrimination of taste qualities among mouse fungiform taste bud cells. J Physiol 587: 4425‐4439, 2009. DOI: 10.1113/jphysiol.2009.175075.
 299.Yoshida R, Noguchi K, Shigemura N, Jyotaki M, Takahashi I, Margolskee RF, Ninomiya Y. Leptin suppresses mouse taste cell responses to sweet compounds. Diabetes 64: 3751‐3762, 2015. DOI: 10.2337/db14‐1462.
 300.Yoshida R, Ohkuri T, Jyotaki M, Yasuo T, Horio N, Yasumatsu K, Sanematsu K, Shigemura N, Yamamoto T, Margolskee RF, Ninomiya Y. Endocannabinoids selectively enhance sweet taste. Proc Natl Acad Sci U S A 107: 935‐939, 2010. DOI: 10.1073/pnas.0912048107.
 301.Young PT, Burright RG, Tromater LJ. Preferences of the white rat for solutions of sucrose and quinine hydrochloride. Am J Psychol 76: 205‐217, 1963.
 302.Zaidi FN, Todd K, Enquist L, Whitehead MC. Types of taste circuits synaptically linked to a few geniculate ganglion neurons. J Comp Neurol 511: 753‐772, 2008. DOI: 10.1002/cne.21869.
 303.Zhang F, Klebansky B, Fine RM, Xu H, Pronin A, Liu H, Tachdjian C, Li X. Molecular mechanism for the umami taste synergism. Proc Natl Acad Sci 105: 20930‐20934, 2008. DOI: 10.1073/pnas.0810174106.
 304.Zhang J, Jin H, Zhang W, Ding C, O'Keeffe S, Ye M, Zuker CS. Sour sensing from the tongue to the brain. Cell 179: 392‐402.e15, 2019. DOI: 10.1016/j.cell.2019.08.031.
 305.Zocchi D, Wennemuth G, Oka Y. The cellular mechanism for water detection in the mammalian taste system. Nat Neurosci 20: 927‐933, 2017. DOI: 10.1038/nn.4575.
 306.Zukerman S, Glendinning JI, Margolskee RF, Sclafani A. T1R3 taste receptor is critical for sucrose but not Polycose taste. Am J Physiol‐Regul Integr Comp Physiol 296: R866‐R876, 2009. DOI: 10.1152/ajpregu.90870.2008.
Further Reading
 1. Bigiani A. The origin of saltiness: Oral detection of NaCl. Curr Opin Physiol 19: 156‐161, 2021. DOI: 10.1016/j.cophys.2020.11.006.
 2.Davidenko O, Darcel N, Fromentin G, Tomé D. Control of protein and energy intake—brain mechanisms. Eur J Clin Nutr 67: 455‐461, 2013. DOI: 10.1038/ejcn.2013.73.
 3.Gilbertson TA, Yu T, Shah BP. Gustatory Mechanisms for Fat Detection. CRC Press/Taylor & Francis, 2010.
 4.Gutierrez R, Fonseca E, Simon SA. The neuroscience of sugars in taste, gut‐reward, feeding circuits. and obesity. Cell Mol Life Sci 77: 3469‐3502, 2020. DOI: 10.1007/s00018‐020‐03458‐2.
 5.Kaelberer MM, Rupprecht LE, Liu WW, Weng P, Bohórquez DV. Neuropod cells: The emerging biology of gut‐brain sensory transduction. Annu Rev Neurosci 43: 337‐353, 2020. DOI: 10.1146/annurev‐neuro‐091619‐022657.
 6.Lim J, Pullicin AJ. Oral carbohydrate sensing: Beyond sweet taste. Physiol Behav 202: 14‐25, 2019. DOI: 10.1016/j.physbeh.2019.01.021.
 7.Roper SD. 3.09—Microphysiology of taste buds. In: Fritzsch B, editor. The Senses: A Comprehensive Reference (2nd ed). Oxford: Elsevier, 2020, p. 187‐210.

Contact Editor

Submit a note to the editor about this article by filling in the form below.

* Required Field

How to Cite

Ranier Gutierrez, Sidney A. Simon. Physiology of Taste Processing in the Tongue, Gut, and Brain. Compr Physiol 2021, 11: 2489-2523. doi: 10.1002/cphy.c210002