Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Front Pharmacol ; 13: 832529, 2022.
Article in English | MEDLINE | ID: mdl-35250580

ABSTRACT

The sweet taste receptor is rather unique, recognizing a diverse repertoire of natural or synthetic ligands, with a surprisingly large structural diversity, and with potencies stretching over more than six orders of magnitude. Yet, it is not clear if different cell-based assays can faithfully report the relative potencies and efficacies of these molecules. Indeed, up to now, sweet taste receptor agonists have been almost exclusively characterized using cell-based assays developed with overexpressed and promiscuous G proteins. This non-physiological coupling has allowed the quantification of receptor activity via phospholipase C activation and calcium mobilization measurements in heterologous cells on a FLIPR system, for example. Here, we developed a novel assay for the human sweet taste receptor where endogenous G proteins and signaling pathways are recruited by the activated receptor. The effects of several sweet taste receptor agonists and other types of modulators were recorded by measuring changes in dynamic mass redistribution (DMR) using an Epic® reader. Potency and efficacy values obtained in the DMR assay were compared to those results obtained with the classical FLIPR assay. Results demonstrate that for some ligands, the two assay systems provide similar information. However, a clear bias for the FLIPR assay was observed for one third of the agonists evaluated, suggesting that the use of non-physiological coupling may influence the potency and efficacy of sweet taste receptor ligands. Replacing the promiscuous G protein with a chimeric G protein containing the C-terminal tail 25 residues of the physiologically relevant G protein subunit Gαgustducin reduced or abrogated bias.

2.
J Med Chem ; 63(9): 4957-4977, 2020 05 14.
Article in English | MEDLINE | ID: mdl-32330040

ABSTRACT

In humans, bitter taste is mediated by 25 TAS2Rs. Many compounds, including certain active pharmaceutical ingredients, excipients, and nutraceuticals, impart their bitter taste (or in part) through TAS2R8 activation. However, effective TAS2R8 blockers that can either suppress or reduce the bitterness of these compounds have not been described. We are hereby reporting a series of novel 3-(pyrazol-4-yl) imidazolidine-2,4-diones as potent and selective TAS2R8 antagonists. In human sensory tests, S6821 and S7958, two of the most potent analogues from the series, demonstrated efficacy in blocking TAS2R8-mediated bitterness and were selected for development. Following data evaluation by expert panels of a number of national and multinational regulatory bodies, including the US, the EU, and Japan, S6821 and S7958 were approved as safe under conditions of intended use as bitter taste blockers.


Subject(s)
Hydantoins/pharmacology , Pyrazoles/pharmacology , Receptors, Cell Surface/antagonists & inhibitors , Receptors, G-Protein-Coupled/antagonists & inhibitors , Taste/drug effects , Animals , Coffee/chemistry , Drug Discovery , Drug Stability , Humans , Hydantoins/chemical synthesis , Hydantoins/toxicity , Molecular Structure , Pyrazoles/chemical synthesis , Pyrazoles/toxicity , Rats , Structure-Activity Relationship
3.
Toxicol Rep ; 4: 507-520, 2017.
Article in English | MEDLINE | ID: mdl-28959681

ABSTRACT

A toxicological evaluation of N-(1-((4-amino-2,2-dioxido-1H-benzo[c][1,2,6]thiadiazin-5-yl)oxy)-2-methylpropan-2-yl)-2,6-dimethylisonicotinamide (S2218; CAS 1622458-34-7), a flavour with modifying properties, was completed for the purpose of assessing its safety for use in food and beverage applications. S2218 exhibited minimal oxidative metabolism in vitro, and in rat pharmacokinetic studies, the compound was poorly orally bioavailable and rapidly eliminated. S2218 was not found to be mutagenic in an in vitro bacterial reverse mutation assay, and was found to be neither clastogenic nor aneugenic in an in vitro mammalian cell micronucleus assay. In subchronic oral toxicity studies in male and female rats, the NOAEL was 140 mg/kg bw/day (highest dose tested) for S2218 sulfate salt (S8069) when administered as a food ad-mix for 13 consecutive weeks. Furthermore, S2218 sulfate salt demonstrated a lack of maternal toxicity, as well as adverse effects on fetal morphology at the highest dose tested, providing a NOAEL of 1000 mg/kg bw/day for both maternal toxicity and embryo/fetal development when administered orally during gestation to pregnant rats.

4.
Trends Pharmacol Sci ; 32(11): 631-6, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21807420

ABSTRACT

A diet low in carbohydrates helps to reduce the amount of ingested calories and to maintain a healthy weight. With this in mind, food and beverage companies have reformulated a large number of their products, replacing sugar or high fructose corn syrup with several different types of zero-calorie sweeteners to decrease or even totally eliminate their caloric content. A challenge remains, however, with the level of acceptance of some of these products in the market-place. Many consumers believe that zero-calorie sweeteners simply do not taste like sugar. A recent breakthrough reveals that positive allosteric modulators of the human sweet taste receptor, small molecules that enhance the receptor activity and sweetness perception, could be more effective than other reported taste enhancers at reducing calories in consumer products without compromising on the true taste of sugar. A unique mechanism of action at the receptor level could explain the robust synergy achieved with these new modulators.


Subject(s)
Allosteric Site/physiology , Sweetening Agents/metabolism , Taste Perception/physiology , Taste/physiology , Allosteric Regulation/drug effects , Carbohydrates/physiology , Energy Intake/physiology , Humans , Receptors, G-Protein-Coupled/metabolism , Sweetening Agents/chemistry , Taste Buds
5.
Biomol Concepts ; 2(4): 327-32, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-25962040

ABSTRACT

Excess sugar intake posts several health problems. Artificial sweeteners have been used for years to reduce dietary sugar content, but they are not ideal substitutes for sugar owing to their off-taste. A new strategy focused on allosteric modulation of the sweet taste receptor led to identification of sweet taste 'enhancers' for the first time. The enhancer molecules do not taste sweet, but greatly potentiate the sweet taste of sucrose and sucralose selectively. Following a similar mechanism as the natural umami taste enhancers, the sweet enhancer molecules cooperatively bind with the sweeteners to the Venus flytrap domain of the human sweet taste receptor and stabilize the active conformation. Now that the approach has proven successful, enhancers for other sweeteners and details of the molecular mechanism for the enhancement are being actively pursued.

6.
Proc Natl Acad Sci U S A ; 107(10): 4746-51, 2010 Mar 09.
Article in English | MEDLINE | ID: mdl-20173092

ABSTRACT

To identify molecules that could enhance sweetness perception, we undertook the screening of a compound library using a cell-based assay for the human sweet taste receptor and a panel of selected sweeteners. In one of these screens we found a hit, SE-1, which significantly enhanced the activity of sucralose in the assay. At 50 microM, SE-1 increased the sucralose potency by >20-fold. On the other hand, SE-1 exhibited little or no agonist activity on its own. SE-1 effects were strikingly selective for sucralose. Other popular sweeteners such as aspartame, cyclamate, and saccharin were not enhanced by SE-1 whereas sucrose and neotame potency were increased only by 1.3- to 2.5-fold at 50 microM. Further assay-guided chemical optimization of the initial hit SE-1 led to the discovery of SE-2 and SE-3, selective enhancers of sucralose and sucrose, respectively. SE-2 (50 microM) and SE-3 (200 microM) increased sucralose and sucrose potencies in the assay by 24- and 4.7-fold, respectively. In human taste tests, 100 microM of SE-1 and SE-2 allowed for a reduction of 50% to >80% in the concentration of sucralose, respectively, while maintaining the sweetness intensity, and 100 microM SE-3 allowed for a reduction of 33% in the concentration of sucrose while maintaining the sweetness intensity. These enhancers did not exhibit any sweetness when tasted on their own. Positive allosteric modulators of the human sweet taste receptor could help reduce the caloric content in food and beverages while maintaining the desired taste.


Subject(s)
Receptors, G-Protein-Coupled/metabolism , Sweetening Agents/pharmacology , Taste Buds/drug effects , Taste/drug effects , Allosteric Regulation , Cell Line , Dose-Response Relationship, Drug , Humans , Molecular Structure , Pyrimidines/chemistry , Pyrimidines/pharmacology , Receptors, G-Protein-Coupled/genetics , Sucrose/administration & dosage , Sucrose/analogs & derivatives , Sucrose/chemistry , Sucrose/pharmacology , Sweetening Agents/administration & dosage , Sweetening Agents/chemistry , Taste Buds/metabolism , Thiophenes/chemistry , Thiophenes/pharmacology
7.
Proc Natl Acad Sci U S A ; 107(10): 4752-7, 2010 Mar 09.
Article in English | MEDLINE | ID: mdl-20173095

ABSTRACT

Positive allosteric modulators of the human sweet taste receptor have been developed as a new way of reducing dietary sugar intake. Besides their potential health benefit, the sweet taste enhancers are also valuable tool molecules to study the general mechanism of positive allosteric modulations of T1R taste receptors. Using chimeric receptors, mutagenesis, and molecular modeling, we reveal how these sweet enhancers work at the molecular level. Our data argue that the sweet enhancers follow a similar mechanism as the natural umami taste enhancer molecules. Whereas the sweeteners bind to the hinge region and induce the closure of the Venus flytrap domain of T1R2, the enhancers bind close to the opening and further stabilize the closed and active conformation of the receptor.


Subject(s)
Models, Molecular , Receptors, G-Protein-Coupled/chemistry , Sweetening Agents/chemistry , Allosteric Regulation , Amino Acid Sequence , Animals , Binding Sites/genetics , Cell Line , Dose-Response Relationship, Drug , Humans , Molecular Sequence Data , Mutation , Protein Binding/drug effects , Protein Structure, Tertiary , Rats , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Sequence Homology, Amino Acid , Sucrose/analogs & derivatives , Sucrose/chemistry , Sucrose/metabolism , Sweetening Agents/metabolism , Sweetening Agents/pharmacology , Transfection
8.
Proc Natl Acad Sci U S A ; 105(52): 20930-4, 2008 Dec 30.
Article in English | MEDLINE | ID: mdl-19104071

ABSTRACT

Umami is one of the 5 basic taste qualities. The umami taste of L-glutamate can be drastically enhanced by 5' ribonucleotides and the synergy is a hallmark of this taste quality. The umami taste receptor is a heteromeric complex of 2 class C G-protein-coupled receptors, T1R1 and T1R3. Here we elucidate the molecular mechanism of the synergy using chimeric T1R receptors, site-directed mutagenesis, and molecular modeling. We propose a cooperative ligand-binding model involving the Venus flytrap domain of T1R1, where L-glutamate binds close to the hinge region, and 5' ribonucleotides bind to an adjacent site close to the opening of the flytrap to further stabilize the closed conformation. This unique mechanism may apply to other class C G-protein-coupled receptors.


Subject(s)
Glutamic Acid/chemistry , Models, Molecular , Multiprotein Complexes/chemistry , Receptors, G-Protein-Coupled/chemistry , Ribonucleotides/chemistry , Taste/physiology , Binding Sites/physiology , Cell Line , Glutamic Acid/genetics , Glutamic Acid/metabolism , Humans , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Mutagenesis, Site-Directed , Protein Binding/physiology , Protein Structure, Quaternary/physiology , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Ribonucleotides/genetics , Ribonucleotides/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...