Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
PLoS One ; 12(7): e0180787, 2017.
Article in English | MEDLINE | ID: mdl-28700634

ABSTRACT

Pre-exposure to taste stimuli and certain chemicals can cause water to have a taste. Here we studied further the 'sweet water taste' (SWT) perceived after exposure to the sweet taste inhibitor lactisole. Experiment 1 investigated an incidental observation that presenting lactisole in mixture with sucrose reduced the intensity of the SWT. The results confirmed this observation and also showed that rinsing with sucrose after lactisole could completely eliminate the SWT. The generalizability of these findings was investigated in experiment 2 by presenting 5 additional sweeteners before, during, or after exposure to lactisole. The results found with sucrose were replicated with fructose and cyclamate, but the 3 other sweeteners were less effective suppressors of the SWT, and the 2 sweeteners having the highest potency initially enhanced it. A third experiment investigated these interactions on the tongue tip and found that the lactisole SWT was perceived only when water was actively flowed across the tongue. The same experiment yielded evidence against the possibility that suppression of the SWT following exposure to sweeteners is an aftereffect of receptor activation while providing additional support for a role of sweetener potency. Collectively these results provide new evidence that complex inhibitory and excitatory interactions occur between lactisole and agonists of the sweet taste receptor TAS1R2-TAS1R3. Receptor mechanisms that may be responsible for these interactions are discussed in the context of the current model of the SWT and the possible contribution of allosteric modulation.


Subject(s)
Sweetening Agents/pharmacology , Adolescent , Adult , Benzene Derivatives/pharmacology , Cyclamates/pharmacology , Female , Fructose/pharmacology , Humans , Male , Middle Aged , Receptors, G-Protein-Coupled/metabolism , Sucrose/pharmacology , Taste Perception/drug effects , Young Adult
2.
J Biol Chem ; 291(29): 15358-77, 2016 Jul 15.
Article in English | MEDLINE | ID: mdl-27226572

ABSTRACT

One key to animal survival is the detection and avoidance of potentially harmful compounds by their bitter taste. Variable numbers of taste 2 receptor genes expressed in the gustatory end organs enable bony vertebrates (Euteleostomi) to recognize numerous bitter chemicals. It is believed that the receptive ranges of bitter taste receptor repertoires match the profiles of bitter chemicals that the species encounter in their diets. Human and mouse genomes contain pairs of orthologous bitter receptor genes that have been conserved throughout evolution. Moreover, expansions in both lineages generated species-specific sets of bitter taste receptor genes. It is assumed that the orthologous bitter taste receptor genes mediate the recognition of bitter toxins relevant for both species, whereas the lineage-specific receptors enable the detection of substances differently encountered by mice and humans. By challenging 34 mouse bitter taste receptors with 128 prototypical bitter substances in a heterologous expression system, we identified cognate compounds for 21 receptors, 19 of which were previously orphan receptors. We have demonstrated that mouse taste 2 receptors, like their human counterparts, vary greatly in their breadth of tuning, ranging from very broadly to extremely narrowly tuned receptors. However, when compared with humans, mice possess fewer broadly tuned receptors and an elevated number of narrowly tuned receptors, supporting the idea that a large receptor repertoire is the basis for the evolution of specialized receptors. Moreover, we have demonstrated that sequence-orthologous bitter taste receptors have distinct agonist profiles. Species-specific gene expansions have enabled further diversification of bitter substance recognition spectra.


Subject(s)
Evolution, Molecular , Receptors, G-Protein-Coupled , Animals , Humans , Mice , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Species Specificity
3.
J Nat Prod ; 77(7): 1739-43, 2014 Jul 25.
Article in English | MEDLINE | ID: mdl-24999828

ABSTRACT

In a screening of extracts of selected plants native to Ohio against the human bitterness receptor hTAS2R31, a chloroform-soluble extract of the aerial parts of Solidago canadensis (Canada goldenrod) was determined to have hTAS2R31 antagonistic activity and, thus, was fractionated for isolation of potential bitterness-masking agents. One new labdane diterpenoid, solidagol (1), and six known terpenoids, including two labdane diterpenoids (2 and 3), three clerodane diterpenoids (6ß-angeloyloxykolavenic acid, 6ß-tigloyloxykolavenic acid, and crotonic acid), and a triterpenoid (longispinogenin), were isolated. Among these compounds, 3ß-acetoxycopalic acid (2) was found to be the first member of the labdane diterpene class shown to have inhibitory activity against hTAS2R31 activation (IC50 8 µM). A homology model of hTAS2R31 was constructed, and the molecular docking of 2 to this model indicated that this diterpenoid binds well to the active site of hTAS2R31, whereas this was not the case for the closely structurally related compound 3 (sempervirenic acid). The content of 2 in the chloroform-soluble portion of the methanolic extract of S. canadensis was up to 2.24 g/100 g dry weight, as determined by HPLC.


Subject(s)
Diterpenes/isolation & purification , Diterpenes/pharmacology , Receptors, G-Protein-Coupled/drug effects , Solidago/chemistry , Terpenes/isolation & purification , Terpenes/pharmacology , Chromatography, High Pressure Liquid , Diterpenes/chemistry , Humans , Molecular Structure , Ohio , Stereoisomerism , Terpenes/chemistry
4.
J Agric Food Chem ; 59(24): 13117-21, 2011 Dec 28.
Article in English | MEDLINE | ID: mdl-22059530

ABSTRACT

The leaves of the native North American plant, Eriodictyon californicum, were once used to mask the bitter taste of pharmaceuticals, an application currently of importance. Ten flavonoids (1-10) were isolated from the leaves of E. californicum, of which the structure and absolute configuration of 6-methoxyhesperetin (8) were assigned for the first time. In addition, the absolute configurations at C-2 were established for 4'-isobutyrylhomoeriodictyol (3) and 6-methoxyhomoeriodictyol (7). Using a cell-based assay, it was determined that the 7-methoxylated flavanones, sakuranetin (2) and 6-methoxysakuranetin (9), and the flavone, jaceosidin (10), are antagonists of hTAS2R31.


Subject(s)
Eriodictyon/chemistry , Flavonoids/pharmacology , Receptors, G-Protein-Coupled/antagonists & inhibitors , Flavonoids/isolation & purification , Plant Leaves/chemistry , Taste
5.
PLoS One ; 6(8): e23165, 2011.
Article in English | MEDLINE | ID: mdl-21829714

ABSTRACT

The sense of taste is important for providing animals with valuable information about the qualities of food, such as nutritional or harmful nature. Mammals, including humans, can recognize at least five primary taste qualities: sweet, umami (savory), bitter, sour, and salty. Recent studies have identified molecules and mechanisms underlying the initial steps of tastant-triggered molecular events in taste bud cells, particularly the requirement of increased cytosolic free Ca(2+) concentration ([Ca(2+)](c)) for normal taste signal transduction and transmission. Little, however, is known about the mechanisms controlling the removal of elevated [Ca(2+)](c) from the cytosol of taste receptor cells (TRCs) and how the disruption of these mechanisms affects taste perception. To investigate the molecular mechanism of Ca(2+) clearance in TRCs, we sought the molecules involved in [Ca(2+)](c) regulation using a single-taste-cell transcriptome approach. We found that Serca3, a member of the sarco/endoplasmic reticulum Ca(2+)-ATPase (SERCA) family that sequesters cytosolic Ca(2+) into endoplasmic reticulum, is exclusively expressed in sweet/umami/bitter TRCs, which rely on intracellular Ca(2+) release for signaling. Serca3-knockout (KO) mice displayed significantly increased aversive behavioral responses and greater gustatory nerve responses to bitter taste substances but not to sweet or umami taste substances. Further studies showed that Serca2 was mainly expressed in the T1R3-expressing sweet and umami TRCs, suggesting that the loss of function of Serca3 was possibly compensated by Serca2 in these TRCs in the mutant mice. Our data demonstrate that the SERCA family members play an important role in the Ca(2+) clearance in TRCs and that mutation of these proteins may alter bitter and perhaps sweet and umami taste perception.


Subject(s)
Receptors, G-Protein-Coupled/physiology , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Taste/physiology , Animals , Immunohistochemistry , Mice , Mice, Knockout , Receptors, G-Protein-Coupled/genetics , Reverse Transcriptase Polymerase Chain Reaction
6.
Chem Senses ; 36(7): 649-58, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21511802

ABSTRACT

We presently investigated 2 novel menthol derivatives GIV1 and GIV2, which exhibit strong cooling effects. In previous human psychophysical studies, GIV1 delivered in a toothpaste medium elicited a cooling sensation that was longer lasting compared with GIV2 and menthol carboxamide (WS-3). In the current study, we investigated the molecular and cellular effects of these cooling agents. In calcium flux studies of TRPM8 expressed in HEK cells, both GIV1 and GIV2 were approximately 40- to 200-fold more potent than menthol and WS-3. GIV1 and GIV2 also activated TRPA1 but at levels that were 400 times greater than those required for TRPM8 activation. In calcium imaging studies, subpopulations of cultured rat trigeminal ganglion and dorsal root ganglion cells responded to GIV1 and/or GIV2; the majority of these were also activated by menthol and some were additionally activated by the TRPA1 agonist cinnamaldehyde and/or the TRPV1 agonist capsaicin. We also made in vivo single-unit recordings from cold-sensitive neurons in rat trigeminal subnucleus caudalis (Vc). GIV 1 and GIV2 directly excited some Vc neurons, GIV1 significantly enhanced their responses to cooling, and both GIV1 and GIV2 reduced responses to noxious heat. These novel cooling compounds provide additional molecular tools to investigate the neural processes of cold sensation.


Subject(s)
Cold Temperature , Menthol/pharmacology , Sensory Receptor Cells/drug effects , Temperature , Tongue/drug effects , Tongue/physiology , Trigeminal Ganglion/cytology , Animals , Cells, Cultured , Ganglia, Spinal/cytology , HEK293 Cells , Humans , Male , Menthol/analogs & derivatives , Menthol/chemistry , Molecular Structure , Rats , Rats, Sprague-Dawley , Sensory Receptor Cells/physiology , Stereoisomerism , Tongue/cytology
7.
Curr Top Med Chem ; 11(15): 1902-24, 2011.
Article in English | MEDLINE | ID: mdl-21470172

ABSTRACT

G protein-coupled receptors (GPCRs) share a common architecture consisting of seven transmembrane (TM) domains. Various lines of evidence suggest that this fold provides a generic binding pocket within the TM region for hosting agonists, antagonists, and allosteric modulators. Hence, an automated method was developed that allows a fast analysis and comparison of these generic ligand binding pockets across the entire GPCR family by providing the relevant information for all GPCRs in the same format. This methodology compiles amino acids lining the TM binding pocket including parts of the ECL2 loop in a so-called 1D ligand binding pocket vector and translates these 1D vectors in a second step into 3D receptor pharmacophore models. It aims to support various aspects of GPCR drug discovery in the pharmaceutical industry. Applications of pharmacophore similarity analysis of these 1D LPVs include definition of receptor subfamilies, prediction of species differences within subfamilies in regard to in vitro pharmacology and identification of nearest neighbors for GPCRs of interest to generate starting points for GPCR lead identification programs. These aspects of GPCR research are exemplified in the field of melanopsins, trace amine-associated receptors and somatostatin receptor subtype 5. In addition, it is demonstrated how 3D pharmacophore models of the LPVs can support the prediction of amino acids involved in ligand recognition, the understanding of mutational data in a 3D context and the elucidation of binding modes for GPCR ligands and their evaluation. Furthermore, guidance through 3D receptor pharmacophore modeling for the synthesis of subtype-specific GPCR ligands will be reported. Illustrative examples are taken from the GPCR family class C, metabotropic glutamate receptors 1 and 5 and sweet taste receptors, and from the GPCR class A, e.g. nicotinic acid and 5-hydroxytryptamine 5A receptor.


Subject(s)
Drug Discovery/methods , Receptors, G-Protein-Coupled/chemistry , Amino Acid Sequence , Binding Sites , Humans , Ligands , Models, Molecular , Molecular Sequence Data , Protein Conformation , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/metabolism
8.
J Neurophysiol ; 105(4): 1701-10, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21273322

ABSTRACT

Szechuan peppers contain hydroxy-α-sanshool that imparts desirable tingling, cooling, and numbing sensations. Hydroxy-α-sanshool activates a subset of sensory dorsal root ganglion (DRG) neurons by inhibiting two-pore potassium channels. We presently investigated if a tingle-evoking sanshool analog, isobutylalkenyl amide (IBA), excites rat DRG neurons and, if so, if these neurons are also activated by agonists of TRPM8, TRPA1, and/or TRPV1. Thirty-four percent of DRG neurons tested responded to IBA, with 29% of them also responding to menthol, 29% to cinnamic aldehyde, 66% to capsaicin, and subsets responding to two or more transient receptor potential (TRP) agonists. IBA-responsive cells had similar size distributions regardless of whether they responded to capsaicin or not; cells only responsive to IBA were larger. Responses to repeated application of IBA at a 5-min interstimulus interval exhibited self-desensitization (tachyphylaxis). Capsaicin did not cross-desensitize responses to IBA to any greater extent than the tachyphylaxis observed with repeated IBA applications. These findings are consistent with psychophysical observations that IBA elicits tingle sensation accompanied by pungency and cooling, with self-desensitization but little cross-desensitization by capsaicin. Intraplantar injection of IBA elicited nocifensive responses (paw licking, shaking-flinching, and guarding) in a dose-related manner similar to the effects of intraplantar capsaicin and serotonin. IBA had no effect on thermal sensitivity but enhanced mechanical sensitivity at the highest dose tested. These observations suggest that IBA elicits an unfamiliar aversive sensation that is expressed behaviorally by the limited response repertoire available to the animal.


Subject(s)
Amides/pharmacology , Behavior, Animal/drug effects , Plant Extracts/pharmacology , Sensory Receptor Cells/drug effects , Zanthoxylum , Acrolein/analogs & derivatives , Acrolein/pharmacology , Animals , Antipruritics/metabolism , Antipruritics/pharmacology , Behavior, Animal/physiology , Calcium/metabolism , Capsaicin/pharmacology , Cells, Cultured , Ganglia, Spinal/cytology , Ganglia, Spinal/drug effects , Ganglia, Spinal/physiology , Humans , Male , Menthol/pharmacology , Models, Animal , Rats , Rats, Sprague-Dawley , Sensory Receptor Cells/physiology
9.
Chem Senses ; 35(7): 579-92, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20660057

ABSTRACT

Variation in taste perception of different chemical substances is a well-known phenomenon in both humans and animals. Recent advances in the understanding of sweet taste signaling have identified a number of proteins involved in this signal transduction. We evaluated the hypothesis that sequence variations occurring in genes encoding taste signaling molecules can influence sweet taste perception in humans. Our population consisted of unrelated individuals (n = 160) of Caucasian, African-American, and Asian descent. Threshold and suprathreshold sensitivities of participants for sucrose were estimated using a sorting test and signal detection analysis that produced cumulative R-index area under the curve (AUC) scores. Genetic association analysis revealed significant correlation of sucrose AUC scores with genetic variation occurring in the GNAT3 gene (single point P = 10(-3) to 10(-4)), which encodes the taste-specific G(alpha) protein subunit gustducin. Subsequent sequencing identified additional GNAT3 variations having significant association with sucrose AUC scores. Collectively, GNAT3 polymorphisms explain 13% of the variation in sucrose perception. Our findings underscore the importance of common genetic variants influencing human taste perception.


Subject(s)
Genetic Variation , Perception/physiology , Polymorphism, Genetic , Signal Transduction/genetics , Sucrose/pharmacology , Taste/genetics , Cells, Cultured , Computational Biology , Humans
10.
Curr Biol ; 20(12): 1104-9, 2010 Jun 22.
Article in English | MEDLINE | ID: mdl-20537538

ABSTRACT

Human bitter taste is mediated by the hTAS2R family of G protein-coupled receptors. The discovery of the hTAS2Rs enables the potential to develop specific bitter receptor antagonists that could be beneficial as chemical probes to examine the role of bitter receptor function in gustatory and nongustatory tissues. In addition, they could have widespread utility in food and beverages fortified with vitamins, antioxidants, and other nutraceuticals, because many of these have unwanted bitter aftertastes. We employed a high-throughput screening approach to discover a novel bitter receptor antagonist (GIV3727) that inhibits activation of hTAS2R31 (formerly hTAS2R44) by saccharin and acesulfame K, two common artificial sweeteners. Pharmacological analyses revealed that GIV3727 likely acts as an orthosteric, insurmountable antagonist of hTAS2R31. Surprisingly, we also found that this compound could inhibit five additional hTAS2Rs, including the closely related receptor hTAS2R43. Molecular modeling and site-directed mutagenesis studies suggest that two residues in helix 7 are important for antagonist activity in hTAS2R31 and hTAS2R43. In human sensory trials, GIV3727 significantly reduced the bitterness associated with the two sulfonamide sweeteners, indicating that hTAS2R antagonists are active in vivo. Our results demonstrate that small molecule bitter receptor antagonists can effectively reduce the bitter taste qualities of foods, beverages, and pharmaceuticals.


Subject(s)
Perception , Receptors, G-Protein-Coupled/antagonists & inhibitors , Taste , Humans
11.
Curr Biol ; 19(15): 1288-93, 2009 Aug 11.
Article in English | MEDLINE | ID: mdl-19559618

ABSTRACT

Human sweet taste perception is mediated by the heterodimeric G protein-coupled receptor encoded by the TAS1R2 and TAS1R3 genes. Variation in these genes has been characterized, but the functional consequences of such variation for sweet perception are unknown. We found that two C/T single-nucleotide polymorphisms (SNPs) located at positions -1572 (rs307355) and -1266 (rs35744813) upstream of the TAS1R3 coding sequence strongly correlate with human taste sensitivity to sucrose and explain 16% of population variability in perception. By using a luciferase reporter assay, we demonstrated that the T allele of each SNP results in reduced promoter activity in comparison to the C alleles, consistent with the phenotype observed in humans carrying T alleles. We also found that the distal region of the TAS1R3 promoter harbors a composite cis-acting element that has a strong silencing effect on promoter activity. We conclude that the rs307355 and rs35744813 SNPs affect gene transcription by altering the function of this regulatory element. A worldwide population survey reveals that the T alleles of rs307355 and rs35744813 occur at lowest frequencies in European populations. We propose that inherited differences in TAS1R3 transcription account for a substantial fraction of worldwide differences in human sweet taste perception.


Subject(s)
Alleles , Polymorphism, Single Nucleotide/genetics , Receptors, G-Protein-Coupled/genetics , Taste Perception/genetics , Analysis of Variance , Area Under Curve , Asian People , Black People , Genotype , Humans , Luciferases , Promoter Regions, Genetic/genetics , Receptors, G-Protein-Coupled/metabolism , Sucrose/metabolism , Taste Perception/physiology , Transcription, Genetic/genetics , White People
12.
J Neurophysiol ; 101(4): 1742-8, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19164099

ABSTRACT

The enigmatic sensation of tingle involves the activation of primary sensory neurons by hydroxy-alpha-sanshool, a tingly agent in Szechuan peppers, by inhibiting two-pore potassium channels. Central mechanisms mediating tingle sensation are unknown. We investigated whether a stable derivative of sanshool-isobutylalkenyl amide (IBA)-excites wide-dynamic range (WDR) spinal neurons that participate in transmission of chemesthetic information from the skin. In anesthetized rats, the majority of WDR and low-threshold units responded to intradermal injection of IBA in a dose-related manner over a >5-min time course and exhibited tachyphylaxis at higher concentrations (1 and 10%). Almost all WDR and low-threshold units additionally responded to the pungent agents mustard oil (allyl isothiocyanate) and/or capsaicin, prompting reclassification of the low-threshold cells as WDR. The results are discussed in terms of the functional role of WDR neurons in mediating tingle sensation.


Subject(s)
Amides/pharmacology , Nonlinear Dynamics , Sensory Receptor Cells/drug effects , Spinal Cord/cytology , Action Potentials/drug effects , Animals , Capsaicin/pharmacology , Cold Temperature , Dose-Response Relationship, Drug , Hot Temperature , Isothiocyanates/pharmacology , Lumbosacral Region , Male , Physical Stimulation/methods , Rats , Rats, Sprague-Dawley , Sensory Receptor Cells/classification , Sensory System Agents/pharmacology , Sensory Thresholds/drug effects , Sensory Thresholds/physiology , Skin/innervation
13.
BMC Struct Biol ; 7: 66, 2007 Oct 12.
Article in English | MEDLINE | ID: mdl-17935609

ABSTRACT

BACKGROUND: Differences in sweet taste perception among species depend on structural variations of the sweet taste receptor. The commercially used isovanillyl sweetener neohesperidin dihydrochalcone activates the human but not the rat sweet receptor TAS1R2+TAS1R3. Analysis of interspecies combinations and chimeras of rat and human TAS1R2+TAS1R3 suggested that the heptahelical domain of human TAS1R3 is crucial for the activation of the sweet receptor by neohesperidin dihydrochalcone. RESULTS: By mutational analysis combined with functional studies and molecular modeling we identified a set of different amino acid residues within the heptahelical domain of human TAS1R3 that forms the neohesperidin dihydrochalcone binding pocket. Sixteen amino acid residues in the transmembrane domains 2 to 7 and one in the extracellular loop 2 of hTAS1R3 influenced the receptor's response to neohesperidin dihydrochalcone. Some of these seventeen residues are also part of the binding sites for the sweetener cyclamate or the sweet taste inhibitor lactisole. In line with this observation, lactisole inhibited activation of the sweet receptor by neohesperidin dihydrochalcone and cyclamate competitively, whereas receptor activation by aspartame, a sweetener known to bind to the N-terminal domain of TAS1R2, was allosterically inhibited. Seven of the amino acid positions crucial for activation of hTAS1R2+hTAS1R3 by neohesperidin dihydrochalcone are thought to play a role in the binding of allosteric modulators of other class C GPCRs, further supporting our model of the neohesperidin dihydrochalcone pharmacophore. CONCLUSION: From our data we conclude that we identified the neohesperidin dihydrochalcone binding site at the human sweet taste receptor, which overlaps with those for the sweetener cyclamate and the sweet taste inhibitor lactisole. This readily delivers a molecular explanation of our finding that lactisole is a competitive inhibitor of the receptor activation by neohesperidin dihydrochalcone and cyclamate. Some of the amino acid positions crucial for activation of hTAS1R2+hTAS1R3 by neohesperidin dihydrochalcone are involved in the binding of allosteric modulators in other class C GPCRs, suggesting a general role of these amino acid positions in allosterism and pointing to a common architecture of the heptahelical domains of class C GPCRs.


Subject(s)
Chalcones/chemistry , Chalcones/metabolism , Hesperidin/analogs & derivatives , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/metabolism , Amino Acid Sequence , Amino Acids/metabolism , Benzene Derivatives , Binding Sites , Hesperidin/chemistry , Hesperidin/metabolism , Humans , Models, Theoretical , Molecular Sequence Data , Mutation/genetics , Protein Structure, Tertiary , Receptors, G-Protein-Coupled/genetics , Sequence Alignment
14.
Curr Biol ; 15(4): 322-7, 2005 Feb 22.
Article in English | MEDLINE | ID: mdl-15723792

ABSTRACT

Individual differences in perception are ubiquitous within the chemical senses: taste, smell, and chemical somesthesis . A hypothesis of this fact states that polymorphisms in human sensory receptor genes could alter perception by coding for functionally distinct receptor types . We have previously reported evidence that sequence variants in a presumptive bitter receptor gene (hTAS2R38) correlate with differences in bitterness recognition of phenylthiocarbamide (PTC) . Here, we map individual psychogenomic pathways for bitter taste by testing people with a variety of psychophysical tasks and linking their individual perceptions of the compounds PTC and propylthiouracil (PROP) to the in vitro responses of their TAS2R38 receptor variants. Functional expression studies demonstrate that five different haplotypes from the hTAS2R38 gene code for operatively distinct receptors. The responses of the three haplotypes we also tested in vivo correlate strongly with individuals' psychophysical bitter sensitivities to a family of compounds. These data provide a direct molecular link between heritable variability in bitter taste perception to functional variations of a single G protein coupled receptor that responds to compounds such as PTC and PROP that contain the N-C=S moiety. The molecular mechanisms of perceived bitterness variability have therapeutic implications, such as helping patients to consume beneficial bitter-tasting compounds-for example, pharmaceuticals and selected phytochemicals.


Subject(s)
Genetic Variation , Phenylthiourea , Propylthiouracil , Receptors, Cell Surface/genetics , Taste/physiology , Animals , Blotting, Western , Cells, Cultured , DNA Mutational Analysis , DNA Primers , DNA, Complementary/genetics , Haplotypes/genetics , Humans , Immunohistochemistry , In Situ Hybridization , Least-Squares Analysis , Rats , Receptors, G-Protein-Coupled , Reverse Transcriptase Polymerase Chain Reaction , Structure-Activity Relationship
15.
J Neurosci ; 24(45): 10260-5, 2004 Nov 10.
Article in English | MEDLINE | ID: mdl-15537898

ABSTRACT

Weight-conscious subjects and diabetics use the sulfonyl amide sweeteners saccharin and acesulfame K to reduce their calorie and sugar intake. However, the intrinsic bitter aftertaste, which is caused by unknown mechanisms, limits the use of these sweeteners. Here, we show by functional expression experiments in human embryonic kidney cells that saccharin and acesulfame K activate two members of the human TAS2R family (hTAS2R43 and hTAS2R44) at concentrations known to stimulate bitter taste. These receptors are expressed in tongue taste papillae. Moreover, the sweet inhibitor lactisole did not block the responses of cells transfected with TAS2R43 and TAS2R44, whereas it did block the response of cells expressing the sweet taste receptor heteromer hTAS1R2-hTAS1R3. The two receptors were also activated by nanomolar concentrations of aristolochic acid, a purely bitter-tasting compound. Thus, hTAS2R43 and hTAS2R44 function as cognate bitter taste receptors and do not contribute to the sweet taste of saccharin and acesulfame K. Consistent with the in vitro data, cross-adaptation studies in human subjects also support the existence of common receptors for both sulfonyl amide sweeteners.


Subject(s)
Receptors, G-Protein-Coupled/physiology , Saccharin/pharmacology , Sweetening Agents/pharmacology , Taste , Thiazines/pharmacology , Aristolochic Acids/pharmacology , Benzene Derivatives/pharmacology , Benzyl Alcohols/pharmacology , Calcium Signaling/drug effects , Cell Line/drug effects , Dose-Response Relationship, Drug , GTP-Binding Protein alpha Subunits/genetics , GTP-Binding Protein alpha Subunits/physiology , Glucosides , Humans , Kidney , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/chemistry , Recombinant Fusion Proteins/physiology , Taste/physiology , Tongue/physiology , Tongue/ultrastructure , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...