TAS1R2

14,000,000 Leading Edge Experts on the ideXlab platform

Scan Science and Technology

Contact Leading Edge Experts & Companies

Scan Science and Technology

Contact Leading Edge Experts & Companies

The Experts below are selected from a list of 8060346 Experts worldwide ranked by ideXlab platform

Wolfgang Meyerhof - One of the best experts on this subject based on the ideXlab platform.

  • Amino acid sensing in hypothalamic tanycytes via umami taste receptors
    Molecular metabolism, 2017
    Co-Authors: Greta Lazutkaite, Wolfgang Meyerhof, Kristina Lossow, Alice Soldà, Nicholas Dale
    Abstract:

    Abstract Objective Hypothalamic tanycytes are glial cells that line the wall of the third ventricle and contact the cerebrospinal fluid (CSF). While they are known to detect glucose in the CSF we now show that tanycytes also detect amino acids, important nutrients that signal satiety. Methods Ca 2+ imaging and ATP biosensing were used to detect tanycyte responses to l -amino acids. The downstream pathway of the responses was determined using ATP receptor antagonists and channel blockers. The receptors were characterized using mice lacking the Tas1r1 gene, as well as an mGluR4 receptor antagonist. Results Amino acids such as Arg, Lys, and Ala evoke Ca 2+ signals in tanycytes and evoke the release of ATP via pannexin 1 and CalHM1, which amplifies the signal via a P2 receptor dependent mechanism. Tanycytes from mice lacking the Tas1r1 gene had diminished responses to lysine and arginine but not alanine. Antagonists of mGluR4 greatly reduced the responses to alanine and lysine. Conclusion Two receptors previously implicated in taste cells, the Tas1r1/Tas1r3 heterodimer and mGluR4, contribute to the detection of a range of amino acids by tanycytes in CSF.

  • A sweet taste receptor-dependent mechanism of glucosensing in hypothalamic tanycytes.
    Glia, 2017
    Co-Authors: Heather Elizabeth Benford, Matei Bolborea, Wolfgang Meyerhof, Sergey Kasparov, Eric Pollatzek, Kristina Lossow, Irm Hermans-borgmeyer, Beihui Liu, Nicholas Dale
    Abstract:

    Hypothalamic tanycytes are glial-like glucosensitive cells that contact the cerebrospinal fluid of the third ventricle, and send processes into the hypothalamic nuclei that control food intake and body weight. The mechanism of tanycyte glucosensing remains undetermined. While tanycytes express the components associated with the glucosensing of the pancreatic β cell, they respond to nonmetabolisable glucose analogues via an ATP receptor-dependent mechanism. Here, we show that tanycytes in rodents respond to non-nutritive sweeteners known to be ligands of the sweet taste (TAS1R2/Tas1r3) receptor. The initial sweet tastant-evoked response, which requires the presence of extracellular Ca2+, leads to release of ATP and a larger propagating Ca2+ response mediated by P2Y1 receptors. In TAS1R2 null mice the proportion of glucose nonresponsive tanycytes was greatly increased in these mice, but a subset of tanycytes retained an undiminished sensitivity to glucose. Our data demonstrate that the sweet taste receptor mediates glucosensing in about 60% of glucosensitive tanycytes while the remaining 40% of glucosensitive tanycytes use some other, as yet unknown mechanism.

  • Rubemamine and Rubescenamine, Two Naturally Occurring N-Cinnamoyl Phenethylamines with Umami-Taste-Modulating Properties.
    Journal of Agricultural and Food Chemistry, 2015
    Co-Authors: Michael Backes, Wolfgang Meyerhof, Katja Obst, Juliane Bojahr, Anika Thorhauer, Natacha Roudnitzky, Susanne Paetz, Katharina Reichelt, Gerhard Krammer, Jakob Ley
    Abstract:

    Sensory screening of a series of naturally occurring N-cinnamoyl derivatives of substituted phenethylamines revealed that rubemamine (9, from Chenopodium album) and rubescenamine (10, from Zanthoxylum rubsecens) elicit strong intrinsic umami taste in water at 50 and 10 ppm, respectively. Sensory tests in glutamate- and nucleotide-containing bases showed that the compounds influence the whole flavor profile of savory formulations. Both rubemamine (9) and rubescenamine (10) at 10–100 ppm dose-dependently positively modulated the umami taste of MSG (0.17–0.22%) up to threefold. Among the investigated amides, only rubemamine (9) and rubescenamine (10) are able to directly activate the TAS1R1-TAS1R3 umami taste receptor. Moreover, both compounds also synergistically modulated the activation of TAS1R1-TAS1R3 by MSG. Most remarkably, rubemamine (9) was able to further positively modulate the IMP-enhanced TAS1R1-TAS1R3 response to MSG ∼1.8-fold. Finally, armatamide (11), zanthosinamide (13), and dioxamine (14), w...

  • Oligomerization of Sweet and Bitter Taste Receptors
    Methods in cell biology, 2013
    Co-Authors: Christian Kuhn, Wolfgang Meyerhof
    Abstract:

    The superfamily of G protein-coupled receptors (GPCRs) mediates numerous physiological processes, including neurotransmission, cell differentiation and metabolism, and sensory perception. In recent years, it became evident that these receptors might function not only as monomeric receptors but also as homo- or heteromeric receptor complexes. The family of TAS1R taste receptors are prominent examples of GPCR dimerization as they act as obligate functional heteromers: TAS1R1 and TAS1R3 combine to form an umami taste receptor, while the combination of TAS1R2 and TAS1R3 is a sweet taste receptor. So far, TAS2Rs, a second family of ~25 taste receptors in humans that mediates responses to bitter compounds, have been shown to function on their own, but if they do so as receptor monomers or as homomeric receptors still remains unknown. Using two different experimental approaches, we have recently shown that TAS2Rs can indeed form both homomeric and heteromeric receptor complexes. The employed techniques, coimmunoprecipitations and bioluminescence resonance energy transfer (BRET), are based on different principles and complement each other well and therefore provided compelling evidences for TAS2R oligomerization. Furthermore, we have adapted the protocols to include a number of controls and for higher throughput to accommodate the investigation of a large number of receptors and receptor combinations. Here, we present the protocols in detail.

  • sweet taste receptor interacting protein cib1 is a general inhibitor of insp3 dependent ca2 release in vivo
    Journal of Neurochemistry, 2008
    Co-Authors: Jan K Hennigs, Nicole Burhenne, Frauke Stähler, Marcel Winnig, Bettina Walter, Wolfgang Meyerhof, Hartwig Schmale
    Abstract:

    In a search for sweet taste receptor interacting proteins, we have identified the calcium- and integrin-binding protein 1 (CIB1) as specific binding partner of the intracellular carboxyterminal domain of the rat sweet taste receptor subunit TAS1R2. In heterologous human embryonic kidney 293 (HEK293) cells, the G protein chimeras Gα16gust44 and Gα15i3 link the sweet taste receptor dimer TAS1R2/TAS1R3 to an inositol 1,4,5-trisphosphate (InsP3)-dependent Ca2+ release pathway. To demonstrate the influence of CIB1 on the cytosolic Ca2+ concentration, we used sweet and umami compounds as well as other InsP3-generating ligands in FURA-2-based Ca2+ assays in wild-type HEK293 cells and HEK293 cells expressing functional human sweet and umami taste receptor dimers. Stable and transient depletion of CIB1 by short-hairpin RNA increased the Ca2+ response of HEK293 cells to the InsP3-generating ligands ATP, UTP and carbachol. Over-expression of CIB1 had the opposite effect as shown for the sweet ligand saccharin, the umami receptor ligand monosodium glutamate and UTP. The CIB1 effect was dependent on the thapsigargin-sensitive Ca2+ store of the endoplasmic reticulum (ER) and independent of extracellular Ca2+. The function of CIB1 on InsP3-evoked Ca2+ release from the ER is most likely mediated by its interaction with the InsP3 receptor. Thus, CIB1 seems to be an inhibitor of InsP3-dependent Ca2+ release in vivo.

Barry G. Green - One of the best experts on this subject based on the ideXlab platform.

  • sweet thermal taste perceptual characteristics in water and dependence on TAS1R2 tas1r3
    Chemical Senses, 2020
    Co-Authors: Danielle Nachtigal, Barry G. Green
    Abstract:

    The initial objective of this study was to determine if activation of the sweet taste receptor TAS1R2/TAS1R3 is necessary for perception of sweet thermal taste (swTT). Our approach was to inhibit the receptor with the inverse agonist lactisole using a temperature-controlled flow gustometer. Because all prior studies of thermal taste (TT) used metal thermodes to heat the tongue tip, we first investigated whether it could be generated in heated water. Experiment 1 showed that sweetness could be evoked when deionized water was heated from 20 to 35 °C, and testing with static temperatures between 20 and 35 °C demonstrated the importance of heating from a cool temperature. As in previous studies, thermal sweetness was reported by only a subset of participants, and replicate measurements found variability in reports of sweetness across trials and between sessions. Experiment 2 then showed that exposure to 8 mM lactisole blocked perception of swTT. Confirmation of the involvement of TAS1R2/TAS1R3 led to an investigation of possible sensory and cognitive interactions between thermal and chemical sweetness. Using sucrose as a sweet stimulus and quinine as a nonsweet control, we found that dynamic heating capable of producing thermal sweetness did not increase the sweetness of sucrose compared with static heating at 35 °C. However, swTT was disrupted if trials containing sucrose (but not quinine) were interspersed among heating-only trials. These findings provide new information relevant to understanding the perceptual processes and receptor mechanisms of swTT, as well as the heat sensitivity of sweet taste in general.

  • Sweet Thermal Taste: Perceptual Characteristics in Water and Dependence on TAS1R2/TAS1R3.
    Chemical senses, 2020
    Co-Authors: Danielle Nachtigal, Barry G. Green
    Abstract:

    The initial objective of this study was to determine if activation of the sweet taste receptor TAS1R2/TAS1R3 is necessary for perception of sweet thermal taste (swTT). Our approach was to inhibit the receptor with the inverse agonist lactisole using a temperature-controlled flow gustometer. Because all prior studies of thermal taste (TT) used metal thermodes to heat the tongue tip, we first investigated whether it could be generated in heated water. Experiment 1 showed that sweetness could be evoked when deionized water was heated from 20 to 35 °C, and testing with static temperatures between 20 and 35 °C demonstrated the importance of heating from a cool temperature. As in previous studies, thermal sweetness was reported by only a subset of participants, and replicate measurements found variability in reports of sweetness across trials and between sessions. Experiment 2 then showed that exposure to 8 mM lactisole blocked perception of swTT. Confirmation of the involvement of TAS1R2/TAS1R3 led to an investigation of possible sensory and cognitive interactions between thermal and chemical sweetness. Using sucrose as a sweet stimulus and quinine as a nonsweet control, we found that dynamic heating capable of producing thermal sweetness did not increase the sweetness of sucrose compared with static heating at 35 °C. However, swTT was disrupted if trials containing sucrose (but not quinine) were interspersed among heating-only trials. These findings provide new information relevant to understanding the perceptual processes and receptor mechanisms of swTT, as well as the heat sensitivity of sweet taste in general.

  • Differential modulation of the lactisole 'Sweet Water Taste' by sweeteners.
    PloS one, 2017
    Co-Authors: Cynthia Alvarado, Jay Patrick Slack, Danielle Nachtigal, Barry G. Green
    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.

Danielle Nachtigal - One of the best experts on this subject based on the ideXlab platform.

  • sweet thermal taste perceptual characteristics in water and dependence on TAS1R2 tas1r3
    Chemical Senses, 2020
    Co-Authors: Danielle Nachtigal, Barry G. Green
    Abstract:

    The initial objective of this study was to determine if activation of the sweet taste receptor TAS1R2/TAS1R3 is necessary for perception of sweet thermal taste (swTT). Our approach was to inhibit the receptor with the inverse agonist lactisole using a temperature-controlled flow gustometer. Because all prior studies of thermal taste (TT) used metal thermodes to heat the tongue tip, we first investigated whether it could be generated in heated water. Experiment 1 showed that sweetness could be evoked when deionized water was heated from 20 to 35 °C, and testing with static temperatures between 20 and 35 °C demonstrated the importance of heating from a cool temperature. As in previous studies, thermal sweetness was reported by only a subset of participants, and replicate measurements found variability in reports of sweetness across trials and between sessions. Experiment 2 then showed that exposure to 8 mM lactisole blocked perception of swTT. Confirmation of the involvement of TAS1R2/TAS1R3 led to an investigation of possible sensory and cognitive interactions between thermal and chemical sweetness. Using sucrose as a sweet stimulus and quinine as a nonsweet control, we found that dynamic heating capable of producing thermal sweetness did not increase the sweetness of sucrose compared with static heating at 35 °C. However, swTT was disrupted if trials containing sucrose (but not quinine) were interspersed among heating-only trials. These findings provide new information relevant to understanding the perceptual processes and receptor mechanisms of swTT, as well as the heat sensitivity of sweet taste in general.

  • Sweet Thermal Taste: Perceptual Characteristics in Water and Dependence on TAS1R2/TAS1R3.
    Chemical senses, 2020
    Co-Authors: Danielle Nachtigal, Barry G. Green
    Abstract:

    The initial objective of this study was to determine if activation of the sweet taste receptor TAS1R2/TAS1R3 is necessary for perception of sweet thermal taste (swTT). Our approach was to inhibit the receptor with the inverse agonist lactisole using a temperature-controlled flow gustometer. Because all prior studies of thermal taste (TT) used metal thermodes to heat the tongue tip, we first investigated whether it could be generated in heated water. Experiment 1 showed that sweetness could be evoked when deionized water was heated from 20 to 35 °C, and testing with static temperatures between 20 and 35 °C demonstrated the importance of heating from a cool temperature. As in previous studies, thermal sweetness was reported by only a subset of participants, and replicate measurements found variability in reports of sweetness across trials and between sessions. Experiment 2 then showed that exposure to 8 mM lactisole blocked perception of swTT. Confirmation of the involvement of TAS1R2/TAS1R3 led to an investigation of possible sensory and cognitive interactions between thermal and chemical sweetness. Using sucrose as a sweet stimulus and quinine as a nonsweet control, we found that dynamic heating capable of producing thermal sweetness did not increase the sweetness of sucrose compared with static heating at 35 °C. However, swTT was disrupted if trials containing sucrose (but not quinine) were interspersed among heating-only trials. These findings provide new information relevant to understanding the perceptual processes and receptor mechanisms of swTT, as well as the heat sensitivity of sweet taste in general.

  • Differential modulation of the lactisole 'Sweet Water Taste' by sweeteners.
    PloS one, 2017
    Co-Authors: Cynthia Alvarado, Jay Patrick Slack, Danielle Nachtigal, Barry G. Green
    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.

Marcel Winnig - One of the best experts on this subject based on the ideXlab platform.

  • sweet taste receptor interacting protein cib1 is a general inhibitor of insp3 dependent ca2 release in vivo
    Journal of Neurochemistry, 2008
    Co-Authors: Jan K Hennigs, Nicole Burhenne, Frauke Stähler, Marcel Winnig, Bettina Walter, Wolfgang Meyerhof, Hartwig Schmale
    Abstract:

    In a search for sweet taste receptor interacting proteins, we have identified the calcium- and integrin-binding protein 1 (CIB1) as specific binding partner of the intracellular carboxyterminal domain of the rat sweet taste receptor subunit TAS1R2. In heterologous human embryonic kidney 293 (HEK293) cells, the G protein chimeras Gα16gust44 and Gα15i3 link the sweet taste receptor dimer TAS1R2/TAS1R3 to an inositol 1,4,5-trisphosphate (InsP3)-dependent Ca2+ release pathway. To demonstrate the influence of CIB1 on the cytosolic Ca2+ concentration, we used sweet and umami compounds as well as other InsP3-generating ligands in FURA-2-based Ca2+ assays in wild-type HEK293 cells and HEK293 cells expressing functional human sweet and umami taste receptor dimers. Stable and transient depletion of CIB1 by short-hairpin RNA increased the Ca2+ response of HEK293 cells to the InsP3-generating ligands ATP, UTP and carbachol. Over-expression of CIB1 had the opposite effect as shown for the sweet ligand saccharin, the umami receptor ligand monosodium glutamate and UTP. The CIB1 effect was dependent on the thapsigargin-sensitive Ca2+ store of the endoplasmic reticulum (ER) and independent of extracellular Ca2+. The function of CIB1 on InsP3-evoked Ca2+ release from the ER is most likely mediated by its interaction with the InsP3 receptor. Thus, CIB1 seems to be an inhibitor of InsP3-dependent Ca2+ release in vivo.

  • sweet taste receptor interacting protein cib1 is a general inhibitor of insp3 dependent ca2 release in vivo
    Journal of Neurochemistry, 2008
    Co-Authors: Jan K Hennigs, Nicole Burhenne, Frauke Stähler, Marcel Winnig, Bettina Walter, Wolfgang Meyerhof, Hartwig Schmale
    Abstract:

    In a search for sweet taste receptor interacting proteins, we have identified the calcium- and integrin-binding protein 1 (CIB1) as specific binding partner of the intracellular carboxyterminal domain of the rat sweet taste receptor subunit TAS1R2. In heterologous human embryonic kidney 293 (HEK293) cells, the G protein chimeras Gα16gust44 and Gα15i3 link the sweet taste receptor dimer TAS1R2/TAS1R3 to an inositol 1,4,5-trisphosphate (InsP3)-dependent Ca2+ release pathway. To demonstrate the influence of CIB1 on the cytosolic Ca2+ concentration, we used sweet and umami compounds as well as other InsP3-generating ligands in FURA-2-based Ca2+ assays in wild-type HEK293 cells and HEK293 cells expressing functional human sweet and umami taste receptor dimers. Stable and transient depletion of CIB1 by short-hairpin RNA increased the Ca2+ response of HEK293 cells to the InsP3-generating ligands ATP, UTP and carbachol. Over-expression of CIB1 had the opposite effect as shown for the sweet ligand saccharin, the umami receptor ligand monosodium glutamate and UTP. The CIB1 effect was dependent on the thapsigargin-sensitive Ca2+ store of the endoplasmic reticulum (ER) and independent of extracellular Ca2+. The function of CIB1 on InsP3-evoked Ca2+ release from the ER is most likely mediated by its interaction with the InsP3 receptor. Thus, CIB1 seems to be an inhibitor of InsP3-dependent Ca2+ release in vivo.

  • the binding site for neohesperidin dihydrochalcone at the human sweet taste receptor
    BMC Structural Biology, 2007
    Co-Authors: Marcel Winnig, Bernd Bufe, Jay Patrick Slack, Nicole A Kratochwil, Wolfgang Meyerhof
    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.

  • The binding site for neohesperidin dihydrochalcone at the human sweet taste receptor
    BMC Structural Biology, 2007
    Co-Authors: Marcel Winnig, Bernd Bufe, Jay Patrick Slack, Nicole A Kratochwil, Wolfgang Meyerhof
    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.

  • A TAS1R receptor-based explanation of sweet ‘water-taste’
    Nature, 2006
    Co-Authors: Veronica Galindo-cuspinera, Marcel Winnig, Bernd Bufe, Wolfgang Meyerhof, Paul A. S. Breslin
    Abstract:

    Paradoxically, artificial sweeteners such as sodium saccharin and acesulfame-K taken in high concentrations are not sweet: they can even seem bitter. And if the mouth is then rinsed out with water, it takes on a sweet taste. These observations have led to new insights into the action of the TAS1R taste receptor. As well as causing the ‘sweet water’ aftertaste, saccharin at high concentrations masks the effect of other sweeteners tasted at the same time. What emerges at the molecular level is a two-site system. Saccharin and acesulfame-K elicit a perception of sweetness when they bind to a high-affinity binding site; at high concentrations they bind to a second low-affinity inhibitory site. When the sweet taste inhibitors are washed away, the sweet receptor re-activates and the perception of sweetness returns. Sweet inhibitors are used in the food industry to offset the high sweetness that results from replacing fats with sweet carbohydrates in some reduced-fat products: the sweet water taste may be a useful predictor for sweet inhibitor activity. ‘Water-tastes’ are gustatory after-impressions elicited by water following the removal of a chemical solution from the mouth, akin to colour after-images appearing on ‘white’ paper after fixation on coloured images. Unlike colour after-images, gustatory after-effects are poorly understood1. One theory posits that ‘water-tastes’ are adaptation phenomena, in which adaptation to one taste solution causes the water presented subsequently to act as a taste stimulus2,3. An alternative hypothesis is that removal of the stimulus upon rinsing generates a receptor-based, positive, off-response in taste-receptor cells, ultimately inducing a gustatory perception4. Here we show that a sweet ‘water-taste’ is elicited when sweet-taste inhibitors are rinsed away. Responses of cultured cells expressing the human sweetener receptor directly parallel the psychophysical responses—water rinses remove the inhibitor from the heteromeric sweetener receptor TAS1R2–TAS1R3, which activates cells and results in the perception of strong sweetness from pure water. This ‘rebound’ activity occurs when equilibrium forces on the two-state allosteric sweet receptors result in their coordinated shift to the activated state upon being released from inhibition by rinsing5,6,7.

Bernd Bufe - One of the best experts on this subject based on the ideXlab platform.

  • the binding site for neohesperidin dihydrochalcone at the human sweet taste receptor
    BMC Structural Biology, 2007
    Co-Authors: Marcel Winnig, Bernd Bufe, Jay Patrick Slack, Nicole A Kratochwil, Wolfgang Meyerhof
    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.

  • The binding site for neohesperidin dihydrochalcone at the human sweet taste receptor
    BMC Structural Biology, 2007
    Co-Authors: Marcel Winnig, Bernd Bufe, Jay Patrick Slack, Nicole A Kratochwil, Wolfgang Meyerhof
    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.

  • A TAS1R receptor-based explanation of sweet ‘water-taste’
    Nature, 2006
    Co-Authors: Veronica Galindo-cuspinera, Marcel Winnig, Bernd Bufe, Wolfgang Meyerhof, Paul A. S. Breslin
    Abstract:

    Paradoxically, artificial sweeteners such as sodium saccharin and acesulfame-K taken in high concentrations are not sweet: they can even seem bitter. And if the mouth is then rinsed out with water, it takes on a sweet taste. These observations have led to new insights into the action of the TAS1R taste receptor. As well as causing the ‘sweet water’ aftertaste, saccharin at high concentrations masks the effect of other sweeteners tasted at the same time. What emerges at the molecular level is a two-site system. Saccharin and acesulfame-K elicit a perception of sweetness when they bind to a high-affinity binding site; at high concentrations they bind to a second low-affinity inhibitory site. When the sweet taste inhibitors are washed away, the sweet receptor re-activates and the perception of sweetness returns. Sweet inhibitors are used in the food industry to offset the high sweetness that results from replacing fats with sweet carbohydrates in some reduced-fat products: the sweet water taste may be a useful predictor for sweet inhibitor activity. ‘Water-tastes’ are gustatory after-impressions elicited by water following the removal of a chemical solution from the mouth, akin to colour after-images appearing on ‘white’ paper after fixation on coloured images. Unlike colour after-images, gustatory after-effects are poorly understood1. One theory posits that ‘water-tastes’ are adaptation phenomena, in which adaptation to one taste solution causes the water presented subsequently to act as a taste stimulus2,3. An alternative hypothesis is that removal of the stimulus upon rinsing generates a receptor-based, positive, off-response in taste-receptor cells, ultimately inducing a gustatory perception4. Here we show that a sweet ‘water-taste’ is elicited when sweet-taste inhibitors are rinsed away. Responses of cultured cells expressing the human sweetener receptor directly parallel the psychophysical responses—water rinses remove the inhibitor from the heteromeric sweetener receptor TAS1R2–TAS1R3, which activates cells and results in the perception of strong sweetness from pure water. This ‘rebound’ activity occurs when equilibrium forces on the two-state allosteric sweet receptors result in their coordinated shift to the activated state upon being released from inhibition by rinsing5,6,7.

  • Valine 738 and lysine 735 in the fifth transmembrane domain of rTas1r3 mediate insensitivity towards lactisole of the rat sweet taste receptor
    BMC Neuroscience, 2005
    Co-Authors: Marcel Winnig, Bernd Bufe, Wolfgang Meyerhof
    Abstract:

    Background The sweet taste inhibitor lactisole acts on the human sweet taste receptor heteromer TAS1R2-TAS1R3 but not on its rodent counterpart. Recently, it was shown that the lactisole sensitivity of the human sweet taste receptor involves the part of TAS1R3 encompassing the seven transmembrane regions but not the huge N-terminal domain. Using mutational analysis we investigated which amino acid residues distinguish lactisole insensitive rat from sensitive human T1R3 receptors.

  • Valine 738 and lysine 735 in the fifth transmembrane domain of rTas1r3 mediate insensitivity towards lactisole of the rat sweet taste receptor
    BMC Neuroscience, 2005
    Co-Authors: Marcel Winnig, Bernd Bufe, Wolfgang Meyerhof
    Abstract:

    Background The sweet taste inhibitor lactisole acts on the human sweet taste receptor heteromer TAS1R2-TAS1R3 but not on its rodent counterpart. Recently, it was shown that the lactisole sensitivity of the human sweet taste receptor involves the part of TAS1R3 encompassing the seven transmembrane regions but not the huge N-terminal domain. Using mutational analysis we investigated which amino acid residues distinguish lactisole insensitive rat from sensitive human T1R3 receptors. Results The functional analysis of specific receptor mutants in HEK293T cells revealed that the exchange of valine 738 in the fifth transmembrane domain of rTas1r3 by an alanine is sufficient to confer lactisole sensitivity to the rat sweet taste receptor. The sensitivity of this receptor mutant is ~2 fold lower than the sensitivity of the human sweet taste receptor. Additional substitution of lysine 735 by phenylalanine in rTas1r3 results in a rat sweet taste receptor that is as sensitive to lactisole as its human counterpart. The exchange of valine 738 to alanine was accompanied by a ~50% reduction in receptor efficacy. This effect was seen with all six different sweet compounds examined. Conclusion The lactisole insensitivity of rat sweet taste receptor is caused by only two amino acids in transmembrane region five, which is critical for the interaction of lactisole with the sweet taste receptor. The observation that the mutant receptor simultaneously displays a generally reduced sensitivity towards all agonists suggests that the lactisole insensitivity of the rodent receptor might be more likely caused by the inaccessibility of the lactisole binding site rather then by its direct disruption.