Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Pharmacol Res ; 204: 107211, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38744400

ABSTRACT

Several non-caloric sweeteners exhibit a delay in sweetness onset and a sweetness linger after sampling. These temporal properties are thought to be the result of non-specific interactions with cell membranes and proteins in the oral cavity. Data and analysis presented in this report also support the potential involvement of receptor affinity and binding kinetics to this phenomenon. In general, affected sweeteners exhibit distinctly higher binding affinity compared to carbohydrate sweeteners, which do not have temporal issues. In addition, binding kinetic simulations illustrate much slower receptor binding association and dissociation kinetics for a set of non-caloric sweeteners presenting temporal issues, in comparison to carbohydrate sweeteners. So, the higher affinity of some non-caloric sweeteners, dictating lower use levels, and affecting binding kinetics, could contribute to their delay and linger in sweetness perception. Simple pharmacology principles could explain, at least in part, some of the temporal issues of sweeteners.


Subject(s)
Sweetening Agents , Taste Perception , Animals , Humans , Kinetics , Receptors, G-Protein-Coupled/metabolism , Sweetening Agents/metabolism , Sweetening Agents/pharmacology , Taste
2.
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.

3.
Handb Exp Pharmacol ; 275: 109-136, 2022.
Article in English | MEDLINE | ID: mdl-33580387

ABSTRACT

Umami, the fifth taste, has been recognized as a legitimate taste modality only recently relative to the other tastes. Dozens of compounds from vastly different chemical classes elicit a savory (also called umami) taste. The prototypical umami substance glutamic acid or its salt monosodium glutamate (MSG) is present in numerous savory food sources or ingredients such as kombu (edible kelp), beans, soy sauce, tomatoes, cheeses, mushrooms, and certain meats and fish. Derivatives of glutamate (Glu), other amino acids, nucleotides, and small peptides can also elicit or modulate umami taste. In addition, many potent umami tasting compounds structurally unrelated to amino acids, nucleotides, and MSG have been either synthesized or discovered as naturally occurring in plants and other substances. Over the last 20 years several receptors have been suggested to mediate umami taste, including members of the metabotropic and ionotropic Glu receptor families, and more recently, the heterodimeric G protein-coupled receptor, T1R1/T1R3. Careful assessment of representative umami tasting molecules from several different chemical classes shows activation of T1R1/T1R3 with the expected rank order of potency in cell-based assays. Moreover, 5'-ribonucleotides, molecules known to enhance the savory note of Glu, considerably enhance the effect of MSG on T1R1/T1R3 in vitro. Binding sites are found on at least 4 distinct locations on T1R1/T1R3, explaining the propensity of the receptor to being activated or modulated by many structurally distinct compounds and these binding sites allosterically interact to modulate receptor activity. Activation of T1R1/T1R3 by all known umami substances evaluated and the receptor's pharmacological properties are sufficient to explain the basic human sensory experience of savory taste and it is therefore unlikely that other receptors are involved.


Subject(s)
Sodium Glutamate , Taste , Animals , Binding Sites , Humans , Nucleotides/pharmacology , Receptors, G-Protein-Coupled/metabolism , Sodium Glutamate/pharmacology , Taste/physiology
4.
Adv Pharmacol ; 88: 59-82, 2020.
Article in English | MEDLINE | ID: mdl-32416872

ABSTRACT

Humans perceive sweet taste via activation of a specific taste receptor expressed at the surface of taste receptor cells located on the tongue and soft palate papillae. The sweet taste receptor functions as an obligate heterodimer, comprising two different class C GPCR subunits. This receptor is unique in that it is activated or modulated by a plethora of ligands from highly diverse chemical classes, from small molecules to peptides and proteins and interacting with topologically distinct sites on each of its subunits. Modulators acting at separate functional domains of the sweet taste receptor can behave as full agonists. However, contrary to observations made with other class C GPCRs such as the metabotropic glutamate receptors and the γ-aminobutyric acid type B receptor (GABAB) receptor, modulators interacting within the allosteric sites in the transmembrane domains of the sweet taste receptor only exert a relatively small effect on the affinity and efficacy of the agonist interacting at the orthosteric binding site located within the Venus fly trap domain (VFD). Newly identified potent and efficacious positive allosteric modulators (PAM)s of the sweet taste receptor rather interact at a site in close proximity to the agonist, within the VFD, display significant probe dependence, and markedly increase the affinity of the orthosteric ligand. Several sweet taste receptor inhibitors have also been characterized. Functional studies reveal a complex relationship between different ligands. Whether the antagonist will be surmountable or insurmountable and will act competitively or non-competitively largely depends on the agonist being studied and the location of its interaction site on the receptor.


Subject(s)
Receptors, G-Protein-Coupled/metabolism , Taste/physiology , Allosteric Regulation , Allosteric Site , Animals , Binding Sites , Humans , Receptors, G-Protein-Coupled/antagonists & inhibitors , Signal Transduction
5.
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
6.
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.

7.
Bioorg Med Chem Lett ; 27(16): 3931-3938, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28662813

ABSTRACT

The paper presents the activity trends for a novel series of phenoxyacetyl amides as human TRPM8 receptor agonists. This series encompasses in vitro activity values ranging from the micromolar to the picomolar levels. Sensory evaluation of these molecules highlights their relevance as cooling agents for oral applications. The positive outcome of the complete evaluation of N-(1H-pyrazol-3-yl)-N-(thiophen-2-ylmethyl)-2-(p-tolyloxy)acetamide resulted in its approval for Generally Recognized As Safe (GRAS) status by the Flavor & Extract Manufacturer Association (FEMA) as FEMA 4809.


Subject(s)
Amides/pharmacology , Cryoprotective Agents/pharmacology , Drug Discovery , TRPM Cation Channels/agonists , Amides/chemical synthesis , Amides/chemistry , Cryoprotective Agents/chemical synthesis , Cryoprotective Agents/chemistry , Dose-Response Relationship, Drug , Humans , Molecular Structure , Structure-Activity Relationship , TRPM Cation Channels/metabolism
8.
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
9.
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.

10.
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
11.
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
12.
Eur J Pharmacol ; 489(3): 139-49, 2004 Apr 12.
Article in English | MEDLINE | ID: mdl-15087236

ABSTRACT

Taste receptors are thought to couple to the G protein Galpha-gustducin to initiate signal transduction cascades leading to taste perception. To further characterize the G protein-coupling selectivity of these receptors, we expressed them in HEK293 cells and monitored the modulation of different signaling pathways upon stimulation. We found that the bitter compound cycloheximide induces phosphorylation of extracellular signal-regulated kinases1 and 2 (ERK 1/2) and inhibits cAMP accumulation in HEK293 cells expressing the mouse bitter T2R(5) receptor. These effects are totally abolished upon treatment with pertussis toxin. On the other hand, sweeteners and monosodium glutamate induce phosphorylation of ERK1/2 and inhibit cAMP accumulation in HEK293 cells expressing the human sweet T1R(2)/T1R(3) receptor and the human umami T1R(1)/T1R(3) receptor, respectively. The effects of these taste modalities are also prevented by treatment with pertussis toxin. Collectively, our results show that taste receptors can functionally couple to Galpha(i/o) proteins to transmit intracellular signals.


Subject(s)
GTP-Binding Protein alpha Subunits, Gi-Go/physiology , MAP Kinase Signaling System/physiology , Receptors, G-Protein-Coupled/metabolism , Taste Buds/physiology , Taste/drug effects , Adenylyl Cyclase Inhibitors , Adenylyl Cyclases/metabolism , Animals , Aspartame/pharmacology , Cell Line , Colforsin/antagonists & inhibitors , Colforsin/pharmacology , Cyclamates/pharmacology , Cyclic AMP/antagonists & inhibitors , Cyclic AMP/chemistry , Cyclic AMP/metabolism , Cycloheximide/metabolism , Cycloheximide/pharmacology , Dose-Response Relationship, Drug , Fructose/pharmacology , Humans , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Pertussis Toxin/pharmacology , Phosphorylation/drug effects , Receptors, G-Protein-Coupled/drug effects , Receptors, G-Protein-Coupled/genetics , Saccharin/metabolism , Saccharin/pharmacology , Sodium Glutamate/metabolism , Sodium Glutamate/pharmacology , Sucrose/metabolism , Sucrose/pharmacology , Taste/genetics , Taste Buds/drug effects , Transducin/physiology , Tryptophan/metabolism , Tryptophan/pharmacology
13.
Nat Cell Biol ; 4(7): 513-8, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12080345

ABSTRACT

In gradients of external chemo-attractant, mammalian neutrophilic leukocytes (neutrophils) and Dictyostelium discoideum amoebae adopt a polarized morphology and selectively accumulate lipid products of phosphatidylinositol-3-OH kinases (PI(3)Ks), including PtdIns(3,4,5)P(3), at their up-gradient edges; the internal PtdIns(3,4,5)P(3) gradient substantially exceeds that of the external attractant. An accompanying report presents evidence for a positive feedback loop that amplifies the gradient of internal signal: PtdIns(3,4,5)P(3) at the leading edge stimulates its own accumulation by inducing activation of one or more Rho GTPases (Rac, Cdc42, and/or Rho), which in turn increase PtdIns(3,4,5)P(3) accumulation. Here we show that interruption of this feedback by treatment with PI(3)K inhibitors reduces the size and stability of pseudopods and causes cells to migrate in jerky trajectories that deviate more from the up-gradient direction than do those of controls. Moreover, amplification of the internal PtdIns(3,4,5)P(3) gradient is markedly impaired by latrunculin or jasplakinolide, toxins that inhibit polymerization or depolymerization of actin, respectively. Thus reciprocal interplay between PtdIns(3,4,5)P(3) and polymerized actin initiates and maintains the asymmetry of intracellular signals responsible for cell polarity and directed motility.


Subject(s)
Cell Movement , Cell Polarity , Depsipeptides , Neutrophils/cytology , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol Phosphates/metabolism , Animals , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Chemotaxis/drug effects , Dictyostelium , Enzyme Inhibitors/pharmacology , Feedback, Physiological , HL-60 Cells , Humans , Marine Toxins/pharmacology , Peptides, Cyclic/pharmacology , Pseudopodia/drug effects , Thiazoles/pharmacology , Thiazolidines
SELECTION OF CITATIONS
SEARCH DETAIL
...