Hostname: page-component-848d4c4894-4hhp2 Total loading time: 0 Render date: 2024-05-15T00:03:55.668Z Has data issue: false hasContentIssue false

Insights on modulators in perception of taste modalities: a review

Published online by Cambridge University Press:  08 July 2019

Shanmugamprema Deepankumar
Affiliation:
Department of Biochemistry, Bharathiar University, Marudhamalai Road, Coimbatore, Tamil Nadu 641046, India
Muthuswamy Karthi
Affiliation:
Department of Biochemistry, Bharathiar University, Marudhamalai Road, Coimbatore, Tamil Nadu 641046, India
Krishnan Vasanth
Affiliation:
Department of Botany, Bharathiar University, Marudhamalai Road, Coimbatore, Tamil Nadu 641046, India
Subramaniam Selvakumar*
Affiliation:
Department of Biochemistry, Bharathiar University, Marudhamalai Road, Coimbatore, Tamil Nadu 641046, India
*
*Corresponding author: Dr S. Selvakumar, email selvs20@yahoo.com
Rights & Permissions [Opens in a new window]

Abstract

A major challenge in taste research is to overcome the flavour imperfections in food products and to build nutritious strategies to combat against obesity as well as other related metabolic syndromes. The field of molecular taste research and chemical senses has contributed to an enormous development in understanding the taste receptors and mechanisms of taste perception. Accordingly, the development of taste-modifying compounds or taste modulators that alter the perception of basic taste modalities has gained significant prominence in the recent past. The beneficial aspects of these substances are overwhelming while considering their potential taste-modifying properties. The objective of the present review is to provide an impression about the taste-modulating compounds and their distinctive taste-modifying properties with reference to their targets and proposed mechanisms of action. The present review also makes an effort to discuss the basic mechanism involved in oro-gustatory taste perception as well as on the effector molecules involved in signal transduction downstream to the activation of taste receptors.

Type
Review Article
Copyright
© The Authors 2019 

Introduction

Taste perception plays a significant role in food preference and determines the intake of foods with pleasurable taste while evading those that are unpleasant. Taste perception is mediated through binding of tastants with their specific receptors and this process is altered by the molecules that interrupt their interaction. Processing of taste information is crucial for facilitating food preference and creating familiar dietetic habits(Reference Kral and Rauh1). Taste modalities such as umami and sweet contribute to an evolutionary role in nutrition as a selective supplier of proteins and energy-rich diet, respectively. Sour and bitter tastes are involved in the evasion of detrimental or spoiled foods(Reference Breslin and Spector2). Salty taste ensures proper dietary electrolyte balance. In addition, recent compelling evidence demonstrates that lipids can be perceived by definite receptors in taste cells and, hence, fat taste is slowly attaining the status of a sixth taste modality(Reference Gaillard, Laugerette and Darcel3Reference Subramaniam, Ozdener and Abdoul-Azize5).

The incidence of obesity and obesity-associated disorders such as type 2 diabetes and the metabolic syndrome has increased pointedly in the previous decades, attained epidemic levels and consequently is becoming a most important worldwide health issue(Reference Ng, Fleming and Robinson6). Increasing concerns regarding the health and quality of life have inspired individuals to prevent the consumption of foods that are rich in fat or sugar(Reference Romagny, Ginon and Salles7, Reference Brug8). Hence, identifying taste in food choice and analysing consumption behaviour will help in understanding the fundamentals involved in body weight maintenance and the development of obesity.

A taste mimetic is a substance that possesses the distinctive organoleptic qualities of a food product. On the other hand, taste modulators are substances that could either enhance or inhibit the perception of a specific taste modality(Reference DeSimone, DuBois, Lyall and Doty9). Modulation of taste responses may occur at numerous phases throughout the course of taste perception, which includes the interaction of tastant molecules with saliva(Reference Matsuo10). Both taste mimetics and taste modulators are being discovered as tools for basic taste research and have some absolute applications in the food industry as well as in the pharmacological industry. Drug discoveries for various medical conditions including asthma and respiratory tract infections(Reference Lee, Xiong and Kofonow11Reference Deshpande, Wang and McIlmoyle13), CVD(Reference Foster, Porrello and Purdue14) and metabolic disorders(Reference Dotson, Zhang and Xu15) are based on taste modulators. In the present review, we focus on both the taste mimetics and taste modulators that reliably modify various taste responses at the receptor level.

Sweet taste

The taste receptors T1R2 and T1R3 (human taste receptor type 1 members 2 and 3) are the principal receptors for sweet taste. These receptors can sense numerous, chemically varied sweetened compounds including both the natural and artificial non-energetic sweeteners, sweet-tasting proteins, natural sugars and certain specific D-amino acids(Reference Behrens, Meyerhof and Hellfritsch16Reference Liman, Zhang and Montell18). In the absence of T1R2, homodimers of T1R3 respond to both monosaccharides as well as to disaccharides. These receptors belong to the class C G protein-coupled receptors (GPCR) family that has definite structures including an N-terminal extracellular domain linked to the seventh transmembrane domain through a sharp cysteine-rich domain(Reference Pin, Galvez and Prezeau19, Reference Vigues, Dotson and Munger20). This family of GPCR contain a bilobate domain with two lobes being separated by a cleft(Reference Liu, Ha and Meng21, Reference Assadi-Porter, Maillet and Radek22). The stimulation of sweet taste receptors, irrespective of their expression pattern either in the taste bud cells of the tongue or in other areas of the oral cavity, results in the activation of α-gustducin. Phospholipase C β2 is subsequently stimulated, leading to the release of stored intracellular Ca2+ and activation of transient receptor potential melastatin 5 (TRPM5) (Fig. 1). This sequence results in membrane depolarisation and the release of neurotransmitters, which can then activate afferent sensory neurons that send signals to brain centres involved in taste perception(Reference Zhang, Hoon and Chandrashekar23).

Fig. 1. Mechanisms of sweet, umami, bitter and fat taste perception by taste bud cells. The taste qualities of sweet, umami or bitter are sensed by the G protein-coupled receptor (GPCR) expressed on type II taste bud cells. Whereas, fat taste perception is mediated by both GPCR and cluster of differentiation 36 (CD36). Upon activation of GPCR, the signal is transmitted through βγ subunits Gβγ and α-gustducin to the downstream effectors phospholipase C β2 (PLC) and adenylate cyclase (AC), respectively. PLC β2 activation generates two second messengers, namely, diacylglycerol (DAG) and inositol 1,4,5-trisphosphate (IP3). IP3 acts on IP3 receptors on endoplasmic reticulum (ER) and increase the intracellular calcium levels. Whereas the AC mediates cyclic AMP (cAMP) production, which block the potassium channels on the plasma membrane. Both these pathways ultimately lead to the depolarisation of the taste receptor cells which lead to the opening of the transient receptor potential melastatin 5 (TRPM5) ion channel. Consequently, ATP is released through the Panx1 hemichannel into the extracellular space, which stimulates multiple targets including the gustatory afferent nerve fibres or the adjacent presynaptic cells which releases the neurotransmitters including serotonin (5-hydroxytryptamine; 5-HT) and noradrenaline into the synaptic cleft. PIP2, phosphatidylinositol 4,5-bisphosphate; T1R, taste receptor type 1; PDE, phosphodiesterase; PKA, protein kinase A; MEK, MAPK/ERK; MAPK, mitogen-activated-protein kinase; ERK, extracellular signal–regulated kinase; PKC, protein kinase C; DRK, delayed rectifying potassium; IP3R, IP3 receptor; SERCA, sarcoendoplasmic reticulum calcium transport ATPase. For a colour figure, see the online version of the article.

Sweet tastants and sweet taste modifiers

Sweeteners or sweet inducers are extensively used as dietary supplements, food additives and pharmaceuticals(Reference Mooradian, Smith and Tokuda24). Sweeteners can be classified into distinctive types according to their intrinsic properties, origin (artificial or natural), nutritional value (energy-containing or non-energy-containing), sweetness potency and their stability(Reference Carocho, Morales and Ferreira25).

The most common natural sweeteners are neohesperidine, steviol glycosides, thaumatin and dihydrochalcone. A bioengineered synthesis pathway concerning glucosyltransferase optimisation using Saccharomyces cerevisiae has led to a new generation of stevia sweeteners(Reference Olsson, Carlsen and Semmler26). Among steviol glucosides, two molecules, namely, rebaudioside A and stevioside, have been established as standard sweeteners(Reference Aranda-Gonzalez, Barbosa-Martin and Toraya-Aviles27, Reference Samuel, Ayoob and Magnuson28). In addition, cycloartane-type saponins, namely, abrusosides from the leaves of Camellia sinensis and Abrus precatorius L., could serve as effective sweet tastants(Reference Stavrianidi, Stekolshchikova and Rodin29). Accordingly, the sweet-tasting proteins can also be perceived as promising natural sweeteners due to their high sweet effectiveness and sensory properties. The plant proteins, namely, brazzein, thaumatin, neoculin, monellin and miraculin, have been recently identified as sweet-tasting molecules(Reference Kant30). Among these, miraculin and neoculin have the uncommon, rare property of adapting sourness into sweetness. Moreover, sensory analysis has revealed that miraculin could represent a natural sugar mimetic used in ancillary beverages(Reference Rodrigues, Andrade and Bastos31). However, the leading problem deterring in the use of sweet-tasting proteins in diet applications is the obstacle of gaining proteins from their natural source.

In addition to the natural sweeteners, artificial sweeteners are also currently in use and the prominent ones are acesulfame K, cyclamates, aspartame, saccharin, neotame and sucralose(Reference Godshall32). Among these, sucralose and neotame are chlorinated derivatives of sucrose and aspartame, respectively (Table 1). In addition, a recently developed dipeptide, advantame, is an excellent heat-stable sweetener that is 20 000 times sweeter than sucrose(Reference Bishay, Bursey and Nabors33).

Table 1. Characteristics of candidate sweet and bitter taste modifiers/tastants and their associated mechanism of chemoreception

TRPM5, transient receptor potential melastatin 5; MCL, miraculin; T1R2, taste receptor type 1 member 2; NCL, neoculin; ACK, acesulfame K; ASP, aspartame; GPCR, G protein-coupled receptor; GIV3727, 4-(2,2,3-trimethylcyclopentyl) butanoic acid; T2R, taste receptor type 2 (bitter taste receptor); GABA, γ-aminobutyric acid; BCML, Nα, Nα-bis (carboxymethyl)-l-lysine; ABA, abscisic acid.

Sweet taste receptors are subjected to both positive and negative allosteric modulation. Screening of the compound library for molecules that positively regulate the in vitro responses of the heterologous sweet taste receptors led to the identification of a sweet taste-enhancing molecule(Reference Servant, Tachdjian and Tang34, Reference Zhang, Klebansky and Fine35). This substance, termed as ‘SE-1’, predominantly enhanced the stimulation of the sweet taste receptor if intensified with sucralose; however, it displayed inadequate or negative modulation when tested with other sweet taste substances(Reference Servant, Tachdjian and Tang34). The structural variation of SE-1 developed by molecular modelling and site-directed mutagenesis gave rise to strong derivatives, which are labelled as SE-2 and SE-3, that are slightly increased in sweetness compared with sucrose(Reference Zhang, Klebansky and Fine35). In addition, polyols are the larger consumed fraction of sweet enhancers due to their lack of cariogenic properties(Reference O’Brien Nabors and Nabors36, Reference Evrendilek, Varzakas, Labropoulos and Anestis37). Therefore, polyols have been used as food additives that effect the hydrogenation of reducing sugars which are found naturally in vegetables and fruits. Many of the polyols including sorbitol, maltitol, mannitol, isomaltose, lactitol, erythritol and xylitol act as effective sweet modifiers.

In contrast, some other compounds inhibit the activity of sweet taste receptors (Table 1). Lactisole is one such compound that inhibits the human sweet taste receptor but not that of the rodent(Reference Sclafani and Pérez38). It was shown that this substance does not bind to the sweet taste receptor subunit T1R2, but instead interacts with T1R3(Reference Jiang, Cui and Zhao39). In addition, compounds isolated from the leaves of the plant Gymnema sylvestre, namely, gymnemic acid and gurmarin peptide, are also sweetness-suppressing molecules that act specifically on rodent sweet taste receptors(Reference Kamei, Takano and Miyasaka40, Reference Glaser, Hellekant and Brouwer41).

Bitter taste

Bitter taste is normally considered to be an unfavourable taste attribute in most food products and elicits a stereotypical innate response by mammals to avert consumption of harmful food constituents. Bitter taste is identified by the receptors that are encoded by the Tas2r gene family expressed in type II taste bud cells and have a sequence length of about 300–330 amino acids with a short extracellular N-terminus. Bitter taste receptors (taste receptors type 2; T2R) belong to class A GPCR and have ligand-binding sites in their transmembrane sections(Reference Adler, Hoon and Mueller42). In taste bud cells, T1R (which sense umami and sweet tastes) and T2R are generally expressed in a non-overlapping array(Reference Meyerhof, Batram and Kuhn43), implying a partition of receptor cells that detect appetitive v. aversive stimuli. In contrast to T1R, the T2R are commonly believed to act as monomers; nevertheless, current evidence proposes that they may also form heterodimers(Reference Kuhn, Bufe and Batram44). The broad and overlapping range of ligand sensitivities of T2R assures that this family of receptors responds to an enormous range of bitter-tasting chemicals.

Bitter taste signalling is initiated by the interaction of the ligand with its cognate T2R that brings a conformational change in the receptor and triggers the heterotrimeric intracellular G-protein complex(Reference Chaudhari and Roper45). When activated, Gα-gustducin becomes relieved from the complex and activates phospholipase C β2 (PLC β2)(Reference Caicedo, Pereira and Margolskee46) (Fig. 1). PLC β2 further acts on phosphatidylinositol-4,5-diphosphate to generate diacylglycerol and inositol 1,4,5-trisphosphate (IP3). The generated IP3 releases Ca2+ ions from the endoplasmic reticulum and opens the TRPM5 which leads to membrane depolarisation and subsequent neurotransmitter release(Reference Chaudhari and Roper45, Reference Hofmann, Chubanov and Gudermann47).

Bitter tastants/modifiers

A widespread variety of structurally diverse compounds can trigger T2R; however, the efficacy of most of them remains to be pharmacologically determined(Reference Devillier, Naline and Grassin-Delyle48, Reference Wiener, Shudler and Levit49) (Table 1). A recent assessment of a bitter compound library comprising both synthetic and natural bitter substances gave rise to the classification of twenty-eight T2R46 agonists, thirty-three T2R14 agonists, and thirty-two T2R10 agonists. The collective activity of all the three receptors is sufficient to perceive half of the tested substances, signifying that these receptors work as ‘generalists’ in the detection of most bitter substances(Reference Meyerhof, Batram and Kuhn43).

A recent study analysed both the promiscuity and selectivity of bitter ligands for human T2R(Reference Di Pizio and Niv50). In this report, the authors proposed that promiscuous bitter compounds activate all the selective T2R while both selective and promiscuous compounds can activate promiscuous T2R. However, no compound is known to activate all twenty-five T2R or no unique compound towards a selective T2R(Reference Meyerhof, Batram and Kuhn43, Reference Di Pizio and Niv50).

4-(2,2,3-Trimethylcyclopentyl) butanoic acid (GIV3727) was the first T2R inhibitor discovered by employing the high-throughput screening approach of 17 854 compounds, which specifically acts as a competitive inhibitor for T2R31 against acesulfame K(Reference Slack, Brockhoff and Batram51). The site-directed mutagenesis and molecular modelling revealed that the binding site of the bitter taste receptors is overlaid with that of the bitter agonists and therefore, this finding describes the competitive mechanism of action and the perceived selectivity of GIV3727 for an individual subset of the human bitter taste receptors(Reference Brockhoff, Behrens and Roudnitzky52). Additionally, probenecid, which was formerly known as an anion transporter channel inhibitor, mostly used as a uricosuric drug(Reference Stamp, O’Donnell and Chapman53), was suggested as a bitter taste blocker(Reference Brockhoff, Behrens and Roudnitzky52, Reference Greene, Alarcon and Thomas54). Moreover, enterodiol, a T2R inhibitor that has appeared to conceal the bitter taste of caffeine, inhibits numerous human bitter taste receptors(Reference Ley, Dessoy and Paetz55). This molecule was recognised by the examination of compounds structurally related to the known bitter masking molecule homoeriodictyol. In addition, 3β-hydroxydihydrocostunolide (3HDC) and 3β-hydroxypelenolide (3HP), natural sesquiterpene lactones from edible plants, were recognised as bitter taste receptor blockers which block the responses of T2R46(Reference Brockhoff, Behrens and Roudnitzky52). Ley et al. (Reference Ley, Dessoy and Paetz55) in their in silico docking experimentations proposed that the attachment of enterodiol to T2R10 may make an impact on the observed bitter masking effect. Recently, Nα,Nα-bis (carboxymethyl)-l-lysine (BCML), γ-aminobutyric acid and (+)-S-abscisic acid are suggested as competitive inhibitors of activated T2R4(Reference Pydi, Sobotkiewicz and Billakanti56). Among them, BCML was considered as a highly effective T2R antagonist reported until now. Besides, studies classified various effective bitter taste inhibitors such as 5’-AMP, sodium acetate, monosodium glutamate (MSG), and sodium gluconate to inhibit whey protein hydrolysate and offer insights on potential bitter taste inhibitors for the product applications associated with whey protein hydrolysate.

More recently, Kim et al. (Reference Kim, Kim and Jin Son57) reported on the active umami fraction (F05) of modernised Korean soya sauce and its bitter masking effect on human bitter-taste sensory receptor-expressing cells. This active umami fraction (F05) reduced the human-perceived bitterness along with efficient repression of the intracellular Ca2+ response induced by caffeine in the hT2R46 and hT2R43 bitter taste receptor-expressing cells. Both Glu-enriched oligopeptides and free amino acids are proposed to be vital in the effect of F05 on bitter taste receptors; F05 was also mixed with other bitter components like magnesium chloride and Gly-Leu that partly modulate the action of human bitter taste receptors.

In contrast, bitter-tasting tri-peptides are shown to be more effective in T2R1 stimulation. Among the peptides examined, the bitter tri-peptide Phe-Phe-Phe is the most potent in activating T2R1 with a half maximal effective concentration (EC50) value in the micromolar range(Reference Upadhyaya, Pydi and Singh58). In addition, Melis & Tomassini Barbarossa(Reference Melis and Tomassini Barbarossa59) examined the taste perception of bitter, sweet and umami with the modifications triggered by l-arginine (l-Arg) supplementation. The outcome proposes that l-Arg could be used as a strategic tool to modify taste responses that are related to eating behaviours. l-Arg can enrich the bitterness intensity of 6-n-propylthiouracil (PROP), whereas other reports(Reference Ahijevych, Tepper and Graham60, Reference Leksrisompong, Gerard and Lopetcharat61) have established a suppression of quinine’s bitterness.

Umami taste

The term ‘umami’ was coined in 1909 by Japanese chemist Kikunae Ikeda, and means ‘delicious savoury taste’(Reference Ikeda62). Binding of the umami tastant, such as free glutamate, in foods to the oral umami taste receptor triggers the umami taste sensation. The typical model of the umami receptor T1R1+T1R3 was reported by Temussi(Reference Temussi63) and Chandrashekar et al. (Reference Chandrashekar, Hoon and Ryba17); it was stated that T1R1 is considered critical for sensing umami taste(Reference Mouritsen and Khandelia64). The heterodimeric GPCR complex of T1R1 and T1R3 elicits the umami taste when interacting with amino acids, typically MSG, and this interaction occurs synergistically with the 5′-ribonucleotides GMP, IMP and AMP(Reference Nelson, Chandrashekar and Hoon65). Furthermore, mGluR1 and mGluR4 were also be identified as the probable receptor candidates for umami taste.

Ligand binding to the T1R1/T1R3 receptor activates Gβ3γ13, which in turn activates PLC β2 that catalyses the production of the second messengers IP3 and diacylglycerol. IP3 binds with IP3 receptor IP3R3 to induce the release of Ca2+ from intracellular stores (Fig. 1). The increase in intracellular Ca2+ subsequently activates TRPM5, which results in taste cell depolarisation and release of ATP that activates ionotropic purinergic receptors on gustatory afferent nerve fibres(Reference Roper66).

Brain mechanisms underlying the oral perception of umami taste have been well documented. Animal studies revealed that the facial (chorda tympani and greater superficial petrosal), glossopharyngeal and vagus (superior laryngeal) nerves, which make synapses with taste cells, convey umami taste information to the first relay nucleus, the rostral part of the nucleus of the solitary tract, and then the taste information is finally transferred to the insular cortex(Reference Sakai, Uneyama, Chavasit and Grumezescu67). The umami tastants such as MSG and IMP activate the same regions of the insular cortex that is known as a primary taste cortex in humans, suggesting that both umami tastants could be similarly recognised(Reference de Araujo, Rolls and Kringelbach68).

Umami tastant/modifiers

Umami tastants are very important for food seasoning and are widely used in food production. They show many health benefits, including reduction in fat deposition, inhibition of weight gain, and decrease in plasma leptin levels in rats(Reference Kondoh and Torii69, Reference Nakamura, Sanematsu and Ohta70). Umami tastants were also found to regulate gastrointestinal functions and to decrease the risk of stroke and CHD in adults by reducing Na intake in their diets(Reference Aburto, Hanson and Gutierrez71, Reference Bera, Kar and Yadav72). The umami taste preference is native and associated with protein-rich food uptake(Reference Chandrashekar, Hoon and Ryba17, Reference Chaudhari and Roper45). Several recent efforts motivated further studies to evaluate the taste properties of umami ingredients and to find new umami substances(Reference Kunishima, Shimada and Tsuji73, Reference Masic and Yeomans74) (Table 2).

Table 2. Characteristics of candidate umami and salt taste modifiers/tastants and their associated mechanism of chemoreception

MSG, monosodium glutamate; T1R, taste receptor type 1; TMD, transmembrane domain; NGCC, N-geranylcyclo propylcarboxamide; TRPV1t, transient receptor potential vanilloid 1; CNS, central nervous system; ENaC, epithelial Na channel.

MSG was the first molecule reported to have umami taste(Reference Ault75). Later, certain ribonucleotides such as IMP and GMP were discovered to have synergistic properties with MSG(Reference Yamaguchi76, Reference Zhang, Venkitasamy and Pan77). In addition, theanine, gallic acid, theogallin(Reference Kaneko, Kumazawa and Masuda78), N-acetylglycine(Reference Amado and Schlichtherle-Cerny79), pyroglutamyl peptides(Reference Martin, Hedwig and Michael80), glycopeptides(Reference Frerot and Benzi81) and succinoyl amides of amino acids(Reference Iwasaki, Miyamura and Kuroda82) were all reported to have umami taste. Alapyridaine(Reference Soldo, Blank and Hofmann83), which is a product of the Maillard reaction, and morelid(Reference Rotzoll, Dunkel and Hofmann84) found in morel mushrooms were also found to enhance umami taste.

Cairoli et al. (Reference Cairoli, Pieraccini and Sironi85) evaluated umami taste enhancement by the positive effect of sulfur substitution, where the umami taste enrichment was amplified if the methylene function of the alkyl chain linked to the exocyclic amino function of 5’-GMP was replaced by a sulfur atom. Furthermore, the umami-enhancing potential was declined if there was an oxidation of sulfur atoms to consequent sulfoxides(Reference Morelli, Manitto and Speranza86).

Moreover, along with these natural and synthesised umami taste-enhancing compounds(Reference Kondoh and Torii69, Reference Nakamura, Sanematsu and Ohta70), several other investigations demonstrated that a few peptide molecules produced from hydrolysates of fish protein, beef bouillon, or other foods, have umami taste(Reference Arai, Yamashita and Noguchi87Reference Winkel, de Klerk and Visser89). Recently, Rhyu & Kim(Reference Rhyu and Kim90) found that low-molecular-weight acidic peptides (F-IV; 1000> MWP500) were the constituent that contributed to the umami taste of doenjang water extract. Further, Su et al. (Reference Su, Cui and Zheng91) found two novel umami taste-enhancing peptides, an octapeptide and an undecapeptide, from groundnut hydrolysate. Bagnasco et al. (Reference Bagnasco, Pappalardo and Meregaglia92) reported that medium-to-small size polypeptides contributed to the umami taste of hydrolysate of rice middlings.

Fat taste

Recently, there has been a massive upsurge of information and evidence on the oro-gustatory perception of fat taste. Reports indicate that improper oral fat detection may be associated with several complications, including obesity-induced lipotoxicity, diabetes, arterial hypertension, atherosclerosis, etc.(Reference Gurevich-Panigrahi, Panigrahi and Wiechec93). The detection of fat stimuli was thought to depend mostly on olfactory, textural and post-ingestive cues(Reference Besnard, Passilly-Degrace and Khan94). However, recently, research conducted predominantly on rodent models exposed an additional gustatory element for the detection of long-chain fatty acids(Reference Besnard, Passilly-Degrace and Khan94, Reference Gilbertson and Khan95). In mammals, oro-gustatory sensing of dietary fat is facilitated by fat taste receptors, namely, cluster of differentiation 36 (CD36) and GPR120/40, which are expressed in taste bud cells on circumvallate papillae, fungiform papillae and foliate papillae of the tongue epithelium(Reference Ozdener, Subramaniam and Sundaresan4, Reference Subramaniam, Ozdener and Abdoul-Azize5). Fat taste perception involves Ca signalling downstream to the activation of CD36 and GPR120/40, stromal interaction molecule 1-mediated opening of store-operated Ca channels, the release of neurotransmitters from taste bud cells and, finally, stimulation of afferent nerve fibres that transmit the signals to the brain(Reference Ozdener, Subramaniam and Sundaresan4, Reference Abdoul-Azize, Selvakumar and Sadou96). Our recent report suggests that the activation of extracellular signal-regulated kinase signalling cascade downstream to the activation of CD36 by fatty acids regulates Ca homeostasis modulator 1 (CALHM1)-mediated Ca signalling in both human and mouse taste bud cells(Reference Subramaniam, Ozdener and Abdoul-Azize5). TRPM5, a monovalent, non-selective cation channel, is reported to be a probable contributor of fat taste signalling(Reference Liu, Shah and Croasdell97). In addition, the delayed rectifying K channels expressed in taste bud cells are inhibited by PUFA in the diet and this supports their involvement in fat taste perception (Fig. 1). Moreover, toll-like receptor 4 signalling has recently been reported to stimulate consumption of obesogenic foods that are rich in fat and sugar(Reference Camandola and Mattson98).

Fat tastants/modifiers

Like sweet tastants, fat tastants are molecules that may be synthesised in the laboratory or purified from plants that imitate the purpose of fat taste by binding to fat taste receptors (online Supplementary Table S1). Recent research had foreseen that the fatty acid-activated CD36 and GPCR might be the potential target of plant-driven tastants eliciting the fat taste sensation devoid of having any energy value(Reference Dramane, Akpona and Simonin99). CD36 and GPR120 agonism with grifolic acid (GA) has been shown to elicit intracellular Ca signalling in both human and mouse taste bud cells(Reference Ozdener, Subramaniam and Sundaresan4). Several selective ligands for free fatty acid (FFA) receptors have subsequently advanced as plausible remedies for type 2 diabetes(Reference Vangaveti, Shashidhar and Jarrod100Reference Zhang and Leung102). Therefore, many enduring academic programmes and industries are driven by the aim of improving selective and potent agonists for FFA receptors. Godinot et al.(Reference Godinot, Yasumatsu and Barcos103) synthesised effective agonists for the fat taste receptors GPR120 and GPR40 in mice, which trigger the glossopharyngeal nerve through binding to the receptor. In humans, various reports derived from triangle tests and two-alternative forced choice, and sensory profiling demonstrate that GPR40 agonists were perceived in sip-and-spit tests and bring out a taste similar to that of linoleic acid(Reference Godinot, Yasumatsu and Barcos103). A vastly convincing FFA1 agonist TUG-770, with its promising pharmacokinetic and physico-chemical properties, displayed increased glucose tolerance in diet-induced obese mice(Reference Christiansen, Hansen and Urban104). Further, through mutational and modelling efforts, the dual synthetic agonists of GPR120/40, including GW9508, NCG21 and NCG46, are emerging as novel ligands with improved pharmacological properties for the fat receptors.

In 2018, Melis et al.(Reference Melis, Mastinu and Arca105) showed that the alterations of oleic acid perception stimulated by the administration of l-Arg are associated with the PROP taster status of subjects and common variants in CD36. Moreover, the low concentration supplementation of l-Arg governed an upsurge in perceived intensity of oleic acid, mostly in medium tasters and PROP non-tasters. Sihag & Jones(Reference Sihag and Jones106) found that oleoylethanolamide, which acts as an effective agonist of PPAR-α, potentially modifies the expression of CD36 and thereby alters fat taste perception. Consequently, a certain group of hybrid composites of thiazolidinedione PPARγ agonists also revealed therapeutic prospective beyond antidiabetic activity(Reference Thangavel, Al Bratty and Akhtar Javed107). In addition, Sasaki et al. (Reference Sasaki, Yasoshima and Matsui108) identified that both intraperitoneal (IP) and orally administered d-serine affect feeding behaviour; especially, IP-injected d-serine prevented the acquisition of a preference for high-fat diets. Furthermore, Murtaza et al.(Reference Murtaza, Berrichi and Bennamar109) presented the first evidence for the modulation of fat taste perception in human taste bud cells (hTBC) by ziziphin, purified from the edible fruit of Zizyphus lotus, indicating the possibility of using these compounds as a fat taste modifier/fat taste mimetic.

Salt taste

Salty taste in mammals is triggered by two different pathways termed as the amiloride-sensitive (AS) pathway and the amiloride-insensitive (AI), or high-salt, pathway. The former selectively reacts to Na and Li salts, which is facilitated by the epithelial Na channel (ENaC)(Reference Heck, Mierson and DeSimone110Reference Roper112). However, the latter reacts to a wide scale of Na and non-Na salts(Reference Ninomiya and Funakoshi113, Reference Halpern114). In rodents, about 65 % of fungiform papillae cells and 35 % of foliate papillae cells displayed efficient amiloride-sensitive Na+ currents, whereas the circumvallate papillae cells are absolutely insensitive to amiloride even though amiloride-sensitive Na+ channel proteins and ENaCα mRNA have been spotted in those cells(Reference Shigemura, Islam and Sadamitsu115). The AI pathway responses have been described in both a subpopulation of type II bitter taste cells and polycystic kidney disease 2-like 1 protein (PKD2L1)-expressing type III taste cells, which is essential for sour taste perception(Reference Oka, Butnaru and von Buchholtz116).

The degree of co-occurrence between sour and AI salt responses in type III taste cells is still undefined due to the frequency of cell-to-cell signal transmission in the taste bud(Reference Roper117). Earlier reports have shown that the type III taste cells are essential for both AI salt taste and sour taste(Reference Oka, Butnaru and von Buchholtz116, Reference Huang, Spielmeyer and Lagudah118Reference Yoshida, Miyauchi and Yasuo120); nevertheless, it was uncertain whether the expression of these taste receptors was exhibited by distinct or similar populations of taste cells. Furthermore, previous studies have shown that the AI salt taste responses originated in a subset of both sour-responsive taste cells and bitter-sensitive type II cells(Reference Oka, Butnaru and von Buchholtz116, Reference Lewandowski, Sukumaran and Margolskee121). These findings suggest that the AI salt taste response perception depends mostly on the collective stimulus of different taste cell populations which determine further distinct bitter or sour taste qualities.

Several reports indicated that NaCl-mediated induction and its relative contributions depend on the concentration of Na(Reference Tordoff, Aleman and McCaughey122, Reference Taruno, Vingtdeux and Ohmoto123). Accordingly, at lower Na concentrations, there is no direct association of CALHM1 but there is an infusion of Na within ENaC located in taste bud cells(Reference Ishiwatari and Bachmanov124, Reference Cherukuri, Bachmanov and McCaughey125) (Fig. 2). At higher Na concentrations, CALHM1 participates in an even more vital role in the neural response. However, further studies will be required to ascertain whether this is due to the stimulation of elevated thresholds of salt receptors in type II cells or due to other Na-receptive cell types that cooperate with type II cells.

Fig. 2. Mechanisms of sour and salt taste perception by taste bud cells. Sour taste is triggered in type III cells by the intracellular proton concentration change triggered by protonated acids. In addition, several channels, including polycystin 2 like 1 (PKD2L1; transient receptor potential cation channel) and PKD1L3 have also been associated with sour taste. For salt taste, the putative candidate is the epithelial-type sodium channel (ENaC). The principal salt stimulus (sodium ion; Na+) can permeate through these cation channels on the apical surface of taste bud cells and trigger depolarisation. ΔVm, membrane potential change; TRPP3, transient receptor potential polycystic 3. For a colour figure, see the online version of the article.

Salt tastants/modifiers

The extreme consumption of salt in the diet is a universal health issue. Various efforts have been made to focus on this issue, involving the evolution of salt substitutes (Table 2) and mounting strategies to lower salt intake(Reference Henney, Taylor and Boon126). Salty taste is chiefly prompted by Na+, which is the simple cation known to elicit a pure salt taste transduction in humans. Other than sodium chloride, various mineral and organic salts provoke a salty taste but to a minor extent(Reference van der Klaauw and Smith127). Potassium chloride is a promising candidate that acts as a substitute for sodium chloride in low-salt foods(Reference Barat, Allende and Aliño128Reference Toldrá and Barat130). However, it has a vulnerable salty taste compared with sodium chloride and, in addition, when used in excessive quantities, it is often linked with bitterness. Other substitutes such as sodium gluconate and ammonium chloride are also recommended but reveal the similar complication of association with bitterness when reacting with KCl, which limits their usage.

Nakagawa et al. (Reference Nakagawa, Kohori and Koike131) reported that sodium aspartate is a potent enhancer of salt taste perception. They proposed a research model based on their study results that the sodium aspartate-induced conformational change on ionic channels enhance NaCl and KCl perception. The enhancement of salt taste transduction by sodium aspartate was also confirmed by human sensory assessments.

The compound (+)-(S)-alapyridaine has universal taste-enhancing properties. When alapyridaine was introduced in the sensory triangle test, the threshold concentrations for the umami taste of MSG and GMP, for the sweet taste of sucrose and glucose, along with the salty taste of NaCl, were considerably decreased. However, on the other hand, the bitter taste perception of l-phenylalanine and caffeine, along with the sour taste of citric acid, was unaltered(Reference Soldo, Blank and Hofmann83). Hence, the taste-enhancing properties of alapyridaine in umami and saltiness prompt developments in the production of low-Na foods for hypertensive patients. Furthermore, Kim et al. (Reference Kim, Son and Kim132) proposed a unique synthetic compound (N-geranylcyclopropylcarboxamide), which modulates amiloride-insensitive Na+-opening pathways, thereby characterised as a salt taste enhancer. More importantly, as per their report, the N-geranylcyclopropylcarboxamide concentration at which it greatly enhanced the benzamil-insensitive Na+ chorda tympani response in rodents also improved the taste perception of NaCl solutions (60–80 mm) in human subjects. Additionally, choline chloride is also proposed to be a salt taste enhancer(Reference Locke and Fielding133) and several choline-containing compounds were synthesised to be used as salt taste enhancers(Reference Fielding and Locke134). Nevertheless, chlorhexidin (an antiseptic compound) has been revealed to impede salt taste perception generated by the chloride salts of Na, K, ammonium and Li.

Taste-enriching peptides are also used to enhance salt taste perception and hence diminish the usage of sodium chloride content in foods. Kino & Kino(Reference Kino and Kino135) synthesised an effective salty taste-enriching dipeptide, Met-Gly, using l-amino acid ligase (Lal) of BL00235 (Lal from Bacillus licheniformis) or TabS (Lal from Pseudomonas syringae) by site-directed mutagenesis based on the perceived crystal structure.

Sour taste

Weak organic acids tend to diffuse through the plasma membrane as neutral molecules in sour taste transduction and dissociate inside the cytoplasm, which causes intracellular acidification(Reference Lyall, Alam and Phan136). However, strong acids depolarise the sour taste receptor cells either by the inhibition of K+ channels or by the stimulation of voltage-gated Na+ channels(Reference Chaudhari and Roper45) (Fig. 2). However, both these mechanisms might result in the opening of voltage-gated Ca2+ channels and stimulate the discharge of neurotransmitters against adjacent afferent nerves.

Sour tastants/modifiers

In sour taste transduction, several ion channels have been predicted to function as mediators despite the fact that the genetics of sour taste perception are inadequately recognised. Although protons signify a dominant part of the sour stimulus, the precise nature of the sour taste stimulus is still under dispute. It was established that weak organic acids in their undissociated form can competently pervade the taste cell plasma membrane and possibly will reduce the intracellular pH values near the cell surface(Reference Huang, Maruyama and Stimac137). Recently, Ohishi et al. (Reference Ohishi, Nishida and Miyamoto138) examined the potentials of the bortezomib-induced taste condition in mice and reported that the sensitivity of sour taste was drastically increased by the administration of bortezomib (online Supplementary Table S1). Moreover, PKD2L1 expression was increased in bortezomib-administered mice, and on cessation of its administration its effect reverted to the control level. Hence, these effects propose that the increase in PKD2L1 protein expression develops sour taste sensitivity in bortezomib-administered mice, and this modification is reversed on termination of its administration. Alternatively, Ishii et al.(Reference Ishii, Kurokawa and Kishi139) suggested that capsaicin can be exploited as an inhibitor of PKD2L1 and PKD1L3, which signifies that in a medical condition, pre-treatment with capsaicin will probably lessen sour taste sensitivity.

Conclusions and future perspectives

Our diets differ based on numerous influences such as environment, our culture and health. At the molecular level, individuals perceive different taste modalities with the help of a range of specified tissues that direct sensory receptors to control nutritious value. In general, the interplay between trigeminal, olfactory and gustatory sensation is interpreted as taste perception like sweet, bitter, umami, salty and sour. Apart from these five principal taste behaviours, the taste system perceives certain non-canonical senses of orosensory taste stimuli such as fat, kokumi, complex carbohydrates and water that prove the existence of additional taste modalities. Current investigations foretold the plant-driven taste modulators which possibly target fatty acid taste receptors (CD36 and GPCR) and provoke a taste sensation devoid of any energy value that may be used against dietary-induced obesity.

Moreover, while the molecular characterisation of modulators and activators for the umami and sweet taste receptors has been carried out, the precise relationship among the T1R heteromeric subunits has yet to be characterised. In the near future, to combat against obesity and other related metabolic syndromes, rigorous strategies should be developed to construct libraries of plant-derived or chemical taste modifiers that would adhere to taste receptors and prompt a façade gustatory sense. In order to achieve this goal, we should attain a comprehensive understanding of the entire oro-gustatory receptors and their respective plant-derived/chemical compounds that are able to activate the gustatory system at the cellular and neurological levels. Likewise, the further understanding of combined biochemical properties of tastants and their inter-species transformations in chemosensory signal detection could be improved.

Furthermore, there is a need for modern contemporary analytical technologies to accelerate the detection of unidentified active chemosensory molecules present in nature and to recognise their physico-chemical relations on a molecular level with food matrix ingredients. The existing complication to gain sensors with suitable sensitivity and selectivity for the examination of taste modulators is directed to the concept of electronic tongues. For example, the electronic bio-mimetic tongue is capable of validating the projected taste intensity of unidentified taste modulators.

Advancement in examination and screening of specific taste-modulating compounds may offer novel platforms for checking the taste of drug candidates and in food quality control. Nowadays, a major challenge in taste research is to overcome the flavour defects in nutritious food products and production of natural or biosynthetic, non-energy-containing fat/sugar mimetics as well as bitter maskers. Moreover, thorough understanding is needed in distinctive age-dependent chemosensory genotypic and phenotypic variances in sensory preference for aversion against food flavours. Further research is essential to enhance our understanding towards the molecular adaptations of salivary composition upon stimulation with tastants along with the receptor proteins included in perceiving tastants. This knowledge will help in developing methodological questions regarding the sequential profile of taste modulators and their biased agonism, which may lead to recognise broadly adjusted enhancers like positive allosteric modulators or inhibitors such as negative allosteric modulators for industrial or commercial applications.

Author ORCIDs

Shanmugamprema Deepankumar 0000-0002-0043-3864, Subramaniam Selvakumar 0000-0003-3798-2899

Supplementary material

The supplementary material for this article can be found at https://doi.org/10.1017/S0954422419000118

Acknowledgements

Financial support provided by the DST-SERB (ECR/20l6/001101) and UGC (BSR startup grant no. F.30-354/2017) India is greatly acknowledged.

S. D. and S. S. planned the graphical abstract and wrote the article. M. K. and K. V. wrote selected parts of the review and facilitated in the formulation of table.

The authors declare no conflicts of interests.

References

Kral, TV & Rauh, EM (2010) Eating behaviors of children in the context of their family environment. Physiol Behav 100, 567573.CrossRefGoogle ScholarPubMed
Breslin, PA & Spector, AC (2008) Mammalian taste perception. Curr Biol 18, R148R155.CrossRefGoogle ScholarPubMed
Gaillard, D, Laugerette, F, Darcel, N, et al. (2008) The gustatory pathway is involved in CD36-mediated orosensory perception of long-chain fatty acids in the mouse. FASEB J 22, 14581468.CrossRefGoogle ScholarPubMed
Ozdener, MH, Subramaniam, S, Sundaresan, S, et al. (2014) CD36- and GPR120-mediated Ca2+ signaling in human taste bud cells mediates differential responses to fatty acids and is altered in obese mice. Gastroenterology 146, 9951005.CrossRefGoogle Scholar
Subramaniam, S, Ozdener, MH, Abdoul-Azize, S, et al. (2016) ERK1/2 activation in human taste bud cells regulates fatty acid signaling and gustatory perception of fat in mice and humans. FASEB J 30, 34893500.CrossRefGoogle ScholarPubMed
Ng, M, Fleming, T, Robinson, M, et al. (2014) Global, regional, and national prevalence of overweight and obesity in children and adults during 1980–2013: a systematic analysis for the Global Burden of Disease Study 2013. Lancet 384, 766781.CrossRefGoogle ScholarPubMed
Romagny, S, Ginon, E & Salles, C (2017) Impact of reducing fat, salt and sugar in commercial foods on consumer acceptability and willingness to pay in real tasting conditions: a home experiment. Food Qual Prefer 56, 164172.CrossRefGoogle Scholar
Brug, J (2008) Determinants of healthy eating: motivation, abilities and environmental opportunities. Fam Pract 25, Suppl. 1, i50i55.CrossRefGoogle ScholarPubMed
DeSimone, JA, DuBois, GE & Lyall, V (2015) Modulators of taste. In Handbook of Olfaction and Gustation, 3rd ed., pp. 667685 [Doty, RL, editor]. Hoboken, NJ: Wiley-Blackwell.Google Scholar
Matsuo, R (2000) Role of saliva in the maintenance of taste sensitivity. Crit Rev Oral Biol Med 11, 216229.CrossRefGoogle ScholarPubMed
Lee, RJ, Xiong, G, Kofonow, JM, et al. (2012) T2R38 taste receptor polymorphisms underlie susceptibility to upper respiratory infection. J Clin Invest 122, 41454159.CrossRefGoogle ScholarPubMed
Tizzano, M, Gulbransen, BD, Vandenbeuch, A, et al. (2010) Nasal chemosensory cells use bitter taste signaling to detect irritants and bacterial signals. Proc Natl Acad Sci U S A 107, 32103215.CrossRefGoogle ScholarPubMed
Deshpande, DA, Wang, WCH, McIlmoyle, EL, et al. (2010) Bitter taste receptors on airway smooth muscle bronchodilate by localized calcium signaling and reverse obstruction. Nat Med 16, 12991304.CrossRefGoogle ScholarPubMed
Foster, SR, Porrello, ER, Purdue, B, et al. (2013) Expression, regulation and putative nutrient-sensing function of taste GPCRs in the heart. PloS ONE 8, e64579.CrossRefGoogle ScholarPubMed
Dotson, CD, Zhang, L, Xu, H, et al. (2008) Bitter taste receptors influence glucose homeostasis. PloS ONE 3, e3974.CrossRefGoogle ScholarPubMed
Behrens, M, Meyerhof, W, Hellfritsch, C, et al. (2011) Sweet and umami taste: natural products, their chemosensory targets, and beyond. Angew Chem Int Ed Engl 50, 22202242.CrossRefGoogle ScholarPubMed
Chandrashekar, J, Hoon, MA, Ryba, NJ, et al. (2006) The receptors and cells for mammalian taste. Nature 444, 288294.CrossRefGoogle ScholarPubMed
Liman, ER, Zhang, YV & Montell, C (2014) Peripheral coding of taste. Neuron 81, 9841000.CrossRefGoogle ScholarPubMed
Pin, JP, Galvez, T & Prezeau, L (2003) Evolution, structure, and activation mechanism of family 3/C G-protein-coupled receptors. Pharmacol Ther 98, 325354.CrossRefGoogle ScholarPubMed
Vigues, S, Dotson, CD & Munger, SD (2009) The receptor basis of sweet taste in mammals. Results Probl Cell Differ 47, 187202.Google ScholarPubMed
Liu, B, Ha, M, Meng, XY, et al. (2012) Functional characterization of the heterodimeric sweet taste receptor T1R2 and T1R3 from a New World monkey species (squirrel monkey) and its response to sweet-tasting proteins. Biochem Biophys Res Commun 427, 431437.CrossRefGoogle ScholarPubMed
Assadi-Porter, FM, Maillet, EL, Radek, JT, et al. (2010) Key amino acid residues involved in multi-point binding interactions between brazzein, a sweet protein, and the T1R2–T1R3 human sweet receptor. J Mol Biol 398, 584599.CrossRefGoogle ScholarPubMed
Zhang, Y, Hoon, MA, Chandrashekar, J, et al. (2003) Coding of sweet, bitter, and umami tastes: different receptor cells sharing similar signaling pathways. Cell 112, 293301.CrossRefGoogle ScholarPubMed
Mooradian, AD, Smith, M & Tokuda, M (2017) The role of artificial and natural sweeteners in reducing the consumption of table sugar: a narrative review. Clin Nutr ESPEN 18, 18.CrossRefGoogle ScholarPubMed
Carocho, M, Morales, P & Ferreira, ICFR (2017) Sweeteners as food additives in the XXI century: a review of what is known, and what is to come. Food Chem Toxicol 107, 302317.CrossRefGoogle ScholarPubMed
Olsson, K, Carlsen, S, Semmler, A, et al. (2016) Microbial production of next-generation stevia sweeteners. Microb Cell Fact 15, 207.CrossRefGoogle ScholarPubMed
Aranda-Gonzalez, I, Barbosa-Martin, E, Toraya-Aviles, R, et al. (2014) Safety assessment of Stevia rebaudiana Bertoni grown in southeastern Mexico as food sweetener (article in Spanish). Nutr Hosp 30, 594601.Google Scholar
Samuel, P, Ayoob, KT, Magnuson, BA, et al. (2018) Stevia leaf to stevia sweetener: exploring its science, benefits, and future potential. J Nutr 148, 1186 S1205 S.CrossRefGoogle ScholarPubMed
Stavrianidi, A, Stekolshchikova, E, Rodin, I, et al. (2018) Structure elucidation of sweet-tasting cycloartane-type saponins from ginseng oolong tea and Abrus precatorius L. leaves. Nat Prod Res 32, 24902493.CrossRefGoogle ScholarPubMed
Kant, R (2005) Sweet proteins – potential replacement for artificial low calorie sweeteners. Nutr J 4, 5.CrossRefGoogle ScholarPubMed
Rodrigues, JF, Andrade, RDS, Bastos, SC, et al. (2016) Miracle fruit: an alternative sugar substitute in sour beverages. Appetite 107, 645653.CrossRefGoogle ScholarPubMed
Godshall, MA (2007) The expanding world of nutritive and non-nutritive sweeteners. Sugar J 69, 1220.Google Scholar
Bishay, IE & Bursey, RG (2012) Advantame. In Alternative Sweeteners, 4th ed., pp. 3145 [Nabors, L O’Brien, editor]. Boca Raton, FL: CRC Press.Google Scholar
Servant, G, Tachdjian, C, Tang, XQ, et al. (2010) Positive allosteric modulators of the human sweet taste receptor enhance sweet taste. Proc Natl Acad Sci U S A 107, 47464751.CrossRefGoogle ScholarPubMed
Zhang, F, Klebansky, B, Fine, RM, et al. (2010) Molecular mechanism of the sweet taste enhancers. Proc Natl Acad Sci U S A 107, 47524757.CrossRefGoogle ScholarPubMed
O’Brien Nabors, L (2011) Alternative sweeteners: a preview. In Alternative Sweeteners, 4th ed., pp. 110 [Nabors, L O’Brien, editor]. Boca Raton, FL: CRC Press.Google Scholar
Evrendilek, GA (2012) Sugar alcohols (polyols). In Sweeteners: Nutritional Aspects, Applications, and Production Technology, pp. 5660 [Varzakas, T, Labropoulos, A and Anestis, S editors]. Boca Raton, FL: CRC Press.Google Scholar
Sclafani, A & Pérez, C (1997) Cypha™ [propionic acid, 2-(4-methoxyphenol) salt] inhibits sweet taste in humans, but not in rats. Physiol Behav 61, 2529.CrossRefGoogle Scholar
Jiang, P, Cui, M, Zhao, B, et al. (2005) Lactisole interacts with the transmembrane domains of human T1R3 to inhibit sweet taste. J Biol Chem 280, 1523815246.CrossRefGoogle ScholarPubMed
Kamei, K, Takano, R, Miyasaka, A, et al. (1992) Amino acid sequence of sweet-taste-suppressing peptide (gurmarin) from the leaves of Gymnema sylvestre. J Biochem 111, 109112.CrossRefGoogle ScholarPubMed
Glaser, D, Hellekant, G, Brouwer, JN, et al. (1984) Effects of gymnemic acid on sweet taste perception in primates. Chem Senses 8, 367374.CrossRefGoogle Scholar
Adler, E, Hoon, MA, Mueller, KL, et al. (2000) A novel family of mammalian taste receptors. Cell 100, 693702.CrossRefGoogle ScholarPubMed
Meyerhof, W, Batram, C, Kuhn, C, et al. (2010) The molecular receptive ranges of human TAS2R bitter taste receptors. Chem Senses 35, 157170.CrossRefGoogle ScholarPubMed
Kuhn, C, Bufe, B, Batram, C, et al. (2010) Oligomerization of TAS2R bitter taste receptors. Chem Senses 35, 395406.CrossRefGoogle ScholarPubMed
Chaudhari, N & Roper, SD (2010) The cell biology of taste. J Cell Biol 190, 285296.CrossRefGoogle ScholarPubMed
Caicedo, A, Pereira, E, Margolskee, RF, et al. (2003) Role of the G-protein subunit α-gustducin in taste cell responses to bitter stimuli. J Neurosci 23, 99479952.CrossRefGoogle ScholarPubMed
Hofmann, T, Chubanov, V, Gudermann, T, et al. (2003) TRPM5 is a voltage-modulated and Ca2+-activated monovalent selective cation channel. Curr Biol 13, 11531158.CrossRefGoogle Scholar
Devillier, P, Naline, E & Grassin-Delyle, S (2015) The pharmacology of bitter taste receptors and their role in human airways. Pharmacol Ther 155, 1121.CrossRefGoogle ScholarPubMed
Wiener, A, Shudler, M, Levit, A, et al. (2012) BitterDB: a database of bitter compounds. Nucleic Acids Res 40, D413D419.CrossRefGoogle ScholarPubMed
Di Pizio, A, Niv, M (2015) Promiscuity and selectivity of bitter molecules and their receptors. Bioorg Med Chem 23, 40824091.CrossRefGoogle ScholarPubMed
Slack, JP, Brockhoff, A, Batram, C, et al. (2010) Modulation of bitter taste perception by a small molecule hTAS2R antagonist. Curr Biol 20, 11041109.CrossRefGoogle ScholarPubMed
Brockhoff, A, Behrens, M, Roudnitzky, N, et al. (2011) Receptor agonism and antagonism of dietary bitter compounds. J Neurosci 31, 1477514782.CrossRefGoogle ScholarPubMed
Stamp, LK, O’Donnell, JL & Chapman, PT (2007) Emerging therapies in the long-term management of hyperuricaemia and gout. Intern Med J 37, 258266.CrossRefGoogle ScholarPubMed
Greene, TA, Alarcon, S, Thomas, A, et al. (2011) Probenecid inhibits the human bitter taste receptor TAS2R16 and suppresses bitter perception of salicin. PLoS ONE 6, e20123.CrossRefGoogle ScholarPubMed
Ley, JP, Dessoy, M, Paetz, S, et al. (2012) Identification of enterodiol as a masker for caffeine bitterness by using a pharmacophore model based on structural analogues of homoeriodictyol. J Agric Food Chem 60, 63036311.CrossRefGoogle ScholarPubMed
Pydi, SP, Sobotkiewicz, T, Billakanti, R, et al. (2014) Amino acid derivatives as bitter taste receptor (T2R) blockers. J Biol Chem 289, 2505425066.CrossRefGoogle ScholarPubMed
Kim, Y, Kim, E-Y, Jin Son, H, et al. (2017) Identification of a key umami-active fraction in modernized Korean soy sauce and the impact thereof on bitter-masking. Food Chem 233, 256262.CrossRefGoogle ScholarPubMed
Upadhyaya, J, Pydi, Sp, Singh, N, et al. (2010) Bitter taste receptor T2R1 is activated by dipeptides and tripeptides. Biochem Biophys Res Commun 398, 331335.CrossRefGoogle ScholarPubMed
Melis, M & Tomassini Barbarossa, I (2017) Taste perception of sweet, sour, salty, bitter, and umami and changes due to l-arginine supplementation, as a function of genetic ability to taste 6-n-propylthiouracil. Nutrients 9, E541.CrossRefGoogle ScholarPubMed
Ahijevych, K, Tepper, BJ, Graham, MC, et al. (2015) Relationships of PROP taste phenotype, taste receptor genotype, and oral nicotine replacement use. Nicotine Tob Res 17, 11491155.CrossRefGoogle ScholarPubMed
Leksrisompong, P, Gerard, P, Lopetcharat, K, et al. (2012) Bitter taste inhibiting agents for whey protein hydrolysate and whey protein hydrolysate beverages. J Food Sci 77, S282S287.CrossRefGoogle ScholarPubMed
Ikeda, K (2002) New seasonings. Chem Senses 27, 847849.CrossRefGoogle ScholarPubMed
Temussi, PA (2012) The good taste of peptides. J Pept Sci 18, 7382.CrossRefGoogle ScholarPubMed
Mouritsen, OG & Khandelia, H (2012) Molecular mechanism of the allosteric enhancement of the umami taste sensation. FEBS J 279, 31123120.CrossRefGoogle ScholarPubMed
Nelson, G, Chandrashekar, J, Hoon, MA, et al. (2002) An amino-acid taste receptor. Nature 416, 199202.CrossRefGoogle Scholar
Roper, SD (2009) Parallel processing in mammalian taste buds? Physiol Behav 97, 604608.CrossRefGoogle ScholarPubMed
Sakai, N, Uneyama, H & Chavasit, V (2016) Psychological and physiological bases of umami taste perception as related to nutrition. In Novel Approaches of Nanotechnology in Food, pp. 697723 [Grumezescu, AM, editor]. Cambridge, MA: Academic Press.CrossRefGoogle Scholar
de Araujo, IE, Rolls, ET, Kringelbach, ML, et al. (2003) Taste-olfactory convergence, and the representation of the pleasantness of flavour, in the human brain. Eur J Neurosci 18, 20592068.CrossRefGoogle ScholarPubMed
Kondoh, T & Torii, K (2008) MSG intake suppresses weight gain, fat deposition, and plasma leptin levels in male Sprague–Dawley rats. Physiol Behav 95, 135144.CrossRefGoogle ScholarPubMed
Nakamura, Y, Sanematsu, K, Ohta, R, et al. (2008) Diurnal variation of human sweet taste recognition thresholds is correlated with plasma leptin levels. Diabetes 57, 26612665.CrossRefGoogle ScholarPubMed
Aburto, NJ, Hanson, S, Gutierrez, H, et al. (2013) Effect of increased potassium intake on cardiovascular risk factors and disease: systematic review and meta-analyses. Br Med J 346, f1378.CrossRefGoogle ScholarPubMed
Bera, T, Kar, SK, Yadav, PK, et al. (2017) Effects of monosodium glutamate (MSG) on human health: a systematic review. World J Pharm Sci 5, 139144.Google Scholar
Kunishima, N, Shimada, Y, Tsuji, Y, et al. (2000) Structural basis of glutamate recognition by a dimeric metabotropic glutamate receptor. Nature 407, 971977.CrossRefGoogle ScholarPubMed
Masic, U & Yeomans, MR (2014) Monosodium glutamate delivered in a protein-rich soup improves subsequent energy compensation. J Nutr Sci 3, e15.CrossRefGoogle Scholar
Ault, A (2004) The monosodium glutamate story: the commercial production of MSG and other amino acids. J Chem Educ 81, 347.CrossRefGoogle Scholar
Yamaguchi, S (1967) The synergistic taste effect of monosodium glutamate and disodium 5′-inosinate. J Food Sci 32, 473478.CrossRefGoogle Scholar
Zhang, Y, Venkitasamy, C, Pan, Z, et al. (2013) Recent developments on umami ingredients of edible mushrooms – a review. Trends Food Sci Technol 33, 7892.CrossRefGoogle Scholar
Kaneko, S, Kumazawa, K, Masuda, H, et al. (2006) Molecular and sensory studies on the umami taste of Japanese green tea. J Agric Food Chem 54, 26882694.CrossRefGoogle ScholarPubMed
Amado, R & Schlichtherle-Cerny, H (2003) Flavoring compositions. Google Patents EP1312 268A1.Google Scholar
Martin, G, Hedwig, SC & Michael, A (2003) Flavouring compositions containing N-acetylglysine. Google Patents WO2003088768A1.Google Scholar
Frerot, E & Benzi, F (2004) Amino acid derivatives of dicarboxylic acids as flavor ingredients. Google Patents WO2004075663A1.Google Scholar
Iwasaki, T, Miyamura, N, Kuroda, M, et al. (2004) Novel glycopeptide or peptide capable of imparting rich taste and method of imparting rich taste to food therewith. Google Patents EP1619201A4.Google Scholar
Soldo, T, Blank, I & Hofmann, T (2003) (+)-(S)-alapyridaine – a general taste enhancer? Chem Senses 28, 371379.CrossRefGoogle ScholarPubMed
Rotzoll, N, Dunkel, A & Hofmann, T (2005) Activity-guided identification of (S)-malic acid 1-O-d-glucopyranoside (morelid) and γ-aminobutyric acid as contributors to umami taste and mouth-drying oral sensation of morel mushrooms (Morchella deliciosa Fr.). J Agric Food Chem 53, 41494156.CrossRefGoogle Scholar
Cairoli, P, Pieraccini, S, Sironi, M, et al. (2008) Studies on umami taste. Synthesis of new guanosine 5’-phosphate derivatives and their synergistic effect with monosodium glutamate. J Agric Food Chem 56, 10431050.CrossRefGoogle ScholarPubMed
Morelli, C, Manitto, P & Speranza, G (2011) Study on umami taste: the MSG taste-enhancing activity of N 2-alkyl and N 2-alkanoyl-5′-guanylic acids having a sulfoxide group inside the N 2-substituent. Flavour Fragr J 26, 279281.CrossRefGoogle Scholar
Arai, S, Yamashita, M & Noguchi, M (1973) Tastes of l-glutamyl oligopeptides in relation to their chromatographic properties. Agric Biol Chem 37, 151156.CrossRefGoogle Scholar
Tamura, M, Seki, T, Kawasaki, Y, et al. (1989) An enhancing effect on the saltiness of sodium chloride of added amino acids and their esters. Agric Biol Chem 53, 16251633.Google Scholar
Winkel, C, de Klerk, A, Visser, J, et al. (2008) New developments in umami (enhancing) molecules. Chem Biodivers 5, 11951203.CrossRefGoogle ScholarPubMed
Rhyu, MR & Kim, EY (2011) Umami taste characteristics of water extract of Doenjang, a Korean soybean paste: low-molecular acidic peptides may be a possible clue to the taste. Food Chem 127, 12101215.CrossRefGoogle ScholarPubMed
Su, G, Cui, C, Zheng, L, et al. (2012) Isolation and identification of two novel umami and umami-enhancing peptides from peanut hydrolysate by consecutive chromatography and MALDI-TOF/TOF MS. Food Chem 135, 479485.CrossRefGoogle ScholarPubMed
Bagnasco, L, Pappalardo, V, Meregaglia, A, et al. (2013) Use of food-grade proteases to recover umami protein–peptide mixtures from rice middlings. Food Res Int 50, 420427.CrossRefGoogle Scholar
Gurevich-Panigrahi, T, Panigrahi, S, Wiechec, E, et al. (2009) Obesity: pathophysiology and clinical management. Curr Med Chem 16, 506521.CrossRefGoogle ScholarPubMed
Besnard, P, Passilly-Degrace, P & Khan, NA (2016) Taste of fat: a sixth taste modality? Physiol Rev 96, 151176.CrossRefGoogle ScholarPubMed
Gilbertson, TA & Khan, NA (2014) Cell signaling mechanisms of oro-gustatory detection of dietary fat: advances and challenges. Prog Lipid Res 53, 8292.CrossRefGoogle ScholarPubMed
Abdoul-Azize, S, Selvakumar, S, Sadou, H, et al. (2014) Ca2+ signaling in taste bud cells and spontaneous preference for fat: unresolved roles of CD36 and GPR120. Biochimie 96, 813.CrossRefGoogle ScholarPubMed
Liu, P, Shah, BP, Croasdell, S, et al. (2011) Transient receptor potential channel type M5 is essential for fat taste. J Neurosci 31, 86348642.CrossRefGoogle ScholarPubMed
Camandola, S & Mattson, MP (2017) Brain metabolism in health, aging, and neurodegeneration. EMBO J 36, 14741492.CrossRefGoogle ScholarPubMed
Dramane, G, Akpona, S, Simonin, AM, et al. (2011) Cell signaling mechanisms of gustatory perception of lipids: can the taste cells be the target of anti-obesity agents? Curr Med Chem 18, 34173422.CrossRefGoogle ScholarPubMed
Vangaveti, V, Shashidhar, V, Jarrod, G, et al. (2010) Free fatty acid receptors: emerging targets for treatment of diabetes and its complications. Ther Adv Endocrinol Metab 1, 165175.Google ScholarPubMed
Ichimura, A, Hasegawa, S, Kasubuchi, M, et al. (2014) Free fatty acid receptors as therapeutic targets for the treatment of diabetes. Front Pharmacol 5, 236.CrossRefGoogle ScholarPubMed
Zhang, D & Leung, PS (2014) Potential roles of GPR120 and its agonists in the management of diabetes. Drug Des Devel Ther 8, 10131027.Google ScholarPubMed
Godinot, N, Yasumatsu, K, Barcos, ME, et al. (2013) Activation of tongue-expressed GPR40 and GPR120 by non-caloric agonists is not sufficient to drive preference in mice. Neuroscience 250, 2030.CrossRefGoogle Scholar
Christiansen, E, Hansen, SV, Urban, C, et al. (2013) Discovery of TUG-770: a highly potent free fatty acid receptor 1 (FFA1/GPR40) agonist for treatment of type 2 diabetes. ACS Med Chem Lett 4, 441445.CrossRefGoogle ScholarPubMed
Melis, M, Mastinu, M, Arca, M, et al. (2018) Effect of chemical interaction between oleic acid and l-arginine on oral perception, as a function of polymorphisms of CD36 and OBPIIa and genetic ability to taste 6-n-propylthiouracil. PLOS ONE 13, e0194953.CrossRefGoogle ScholarPubMed
Sihag, J & Jones, PJH (2018) Oleoylethanolamide: the role of a bioactive lipid amide in modulating eating behaviour. Obes Rev 19, 178197.CrossRefGoogle ScholarPubMed
Thangavel, N, Al Bratty, M, Akhtar Javed, S, et al. (2017) Targeting peroxisome proliferator-activated receptors using thiazolidinediones: strategy for design of novel antidiabetic drugs. Int J Med Chem 2017, 1069718.Google ScholarPubMed
Sasaki, T, Yasoshima, Y, Matsui, S, et al. (2017) Intraperitoneal injection of d-serine inhibits high-fat diet intake and preference in male mice. Appetite 118, 120128.CrossRefGoogle ScholarPubMed
Murtaza, B, Berrichi, M, Bennamar, C, et al. (2017) Zizyphin modulates calcium signalling in human taste bud cells and fat taste perception in the mouse. Fundam Clin Pharmacol 31, 486494.CrossRefGoogle ScholarPubMed
Heck, GL, Mierson, S & DeSimone, JA (1984) Salt taste transduction occurs through an amiloride-sensitive sodium transport pathway. Science 223, 403405.CrossRefGoogle ScholarPubMed
Chandrashekar, J, Kuhn, C, Oka, Y, et al. (2010) The cells and peripheral representation of sodium taste in mice. Nature 464, 297301.CrossRefGoogle ScholarPubMed
Roper, SD (2015) The taste of table salt. Pflugers Arch 467, 457463.CrossRefGoogle ScholarPubMed
Ninomiya, Y & Funakoshi, M (1988) Amiloride inhibition of responses of rat single chorda tympani fibers to chemical and electrical tongue stimulations. Brain Res 451, 319325.CrossRefGoogle ScholarPubMed
Halpern, BP (1998) Amiloride and vertebrate gustatory responses to NaCl. Neurosci Biobehav Rev 23, 547.CrossRefGoogle ScholarPubMed
Shigemura, N, Islam, AA, Sadamitsu, C, et al. (2005) Expression of amiloride-sensitive epithelial sodium channels in mouse taste cells after chorda tympani nerve crush. Chem Senses 30, 531538.CrossRefGoogle ScholarPubMed
Oka, Y, Butnaru, M, von Buchholtz, L, et al. (2013) High salt recruits aversive taste pathways. Nature 494, 472475.CrossRefGoogle ScholarPubMed
Roper, SD (2013) Taste buds as peripheral chemosensory processors. Semin Cell Dev Biol 24, 7179.CrossRefGoogle ScholarPubMed
Huang, S, Spielmeyer, W, Lagudah, ES, et al. (2006) A sodium transporter (HKT7) is a candidate for Nax1, a gene for salt tolerance in durum wheat. Plant Physiol 142, 17181727.CrossRefGoogle ScholarPubMed
Tomchik, SM, Berg, S, Kim, JW, et al. (2007) Breadth of tuning and taste coding in mammalian taste buds. J Neurosci 27, 1084010848.CrossRefGoogle ScholarPubMed
Yoshida, R, Miyauchi, A, Yasuo, T, et al. (2009) Discrimination of taste qualities among mouse fungiform taste bud cells. J Physiol 587, 44254439.CrossRefGoogle ScholarPubMed
Lewandowski, BC, Sukumaran, SK, Margolskee, RF, et al. (2016) Amiloride-insensitive salt taste is mediated by two populations of type III taste cells with distinct transduction mechanisms. J Neurosci 36, 19421953.CrossRefGoogle ScholarPubMed
Tordoff, MG, Aleman, TR & McCaughey, SA (2015) Heightened avidity for trisodium pyrophosphate in mice lacking Tas1r3. Chem Senses 40, 5359.CrossRefGoogle ScholarPubMed
Taruno, A, Vingtdeux, V, Ohmoto, M, et al. (2013) CALHM1 ion channel mediates purinergic neurotransmission of sweet, bitter and umami tastes. Nature 495, 223226.CrossRefGoogle ScholarPubMed
Ishiwatari, Y & Bachmanov, AA (2012) NaCl taste thresholds in 13 inbred mouse strains. Chem Senses 37, 497508.CrossRefGoogle ScholarPubMed
Cherukuri, CM, Bachmanov, AA & McCaughey, SA (2013) A/J and C57BL/6J mice differ in chorda tympani responses to NaCl. Neurosci Res 75, 283288.CrossRefGoogle ScholarPubMed
Henney, JE, Taylor, CL & Boon, CS (2010) Strategies to Reduce Sodium Intake in the United States. Washington, DC: The National Academies Press.Google Scholar
van der Klaauw, NJ & Smith, DV (1995) Taste quality profiles for fifteen organic and inorganic salts. Physiol Behav 58, 295306.CrossRefGoogle ScholarPubMed
Barat, J, Allende, D, Aliño, M, et al. (2011) Kinetics studies during NaCl and KCl pork meat brining. J Food Eng 106, 102110.CrossRefGoogle Scholar
Hooge, S & Chambers, D (2010) A comparison of basic taste modalities, using a descriptive analysis technique, for varying levels of sodium and KCl in two model soup systems. J Sens Stud 25, 521535.Google Scholar
Toldrá, F & Barat, JM (2009) Recent patents for sodium reduction in foods. Recent Pat Food Nutr Agric 1, 8086.CrossRefGoogle ScholarPubMed
Nakagawa, T, Kohori, J, Koike, S, et al. (2014) Sodium aspartate as a specific enhancer of salty taste perception-sodium aspartate is a possible candidate to decrease excessive intake of dietary salt. Chem Senses 39, 781786.CrossRefGoogle ScholarPubMed
Kim, MJ, Son, HJ, Kim, Y, et al. (2014) Selective activation of hTRPV1 by N-geranyl cyclopropylcarboxamide, an amiloride-insensitive salt taste enhancer. PLOS ONE 9, e89062.CrossRefGoogle ScholarPubMed
Locke, KW & Fielding, S (1994) Enhancement of salt intake by choline chloride. Physiol Behav 55, 10391046.CrossRefGoogle ScholarPubMed
Fielding, S & Locke, KW (1993) Choline-containing compositions as salt substitutes and enhancers and a method of preparation. Interneuron Pharmaceuticals Inc US5206049A.Google Scholar
Kino, H & Kino, K (2015) Alteration of the substrate specificity of l-amino acid ligase and selective synthesis of Met-Gly as a salt taste enhancer. Biosci Biotechnol Biochem 79, 18271832.CrossRefGoogle ScholarPubMed
Lyall, V, Alam, RI, Phan, DQ, et al. (2001) Decrease in rat taste receptor cell intracellular pH is the proximate stimulus in sour taste transduction. Am J Physiol Cell Physiol 281, C1005C1013.CrossRefGoogle ScholarPubMed
Huang, YA, Maruyama, Y, Stimac, R, et al. (2008) Presynaptic (type III) cells in mouse taste buds sense sour (acid) taste. J Physiol 586, 29032912.CrossRefGoogle ScholarPubMed
Ohishi, A, Nishida, K, Miyamoto, K, et al. (2017) Bortezomib alters sour taste sensitivity in mice. Toxicol Rep 4, 172180.CrossRefGoogle ScholarPubMed
Ishii, S, Kurokawa, A, Kishi, M, et al. (2012) The response of PKD1L3/PKD2L1 to acid stimuli is inhibited by capsaicin and its pungent analogs. FEBS J 279, 18571870.CrossRefGoogle ScholarPubMed
Philippaert, K, Pironet, A, Mesuere, M, et al. (2017) Steviol glycosides enhance pancreatic β-cell function and taste sensation by potentiation of TRPM5 channel activity. Nat Commun 8, 14733.CrossRefGoogle ScholarPubMed
Anton, SD, Martin, CK, Han, H, et al. (2010) Effects of stevia, aspartame, and sucrose on food intake, satiety, and postprandial glucose and insulin levels. Appetite 55, 3743.CrossRefGoogle ScholarPubMed
Sanematsu, K, Kitagawa, M, Yoshida, R, et al. (2016) Intracellular acidification is required for full activation of the sweet taste receptor by miraculin. Sci Rep 6, 22807.CrossRefGoogle ScholarPubMed
Misaka, T (2013) Molecular mechanisms of the action of miraculin, a taste-modifying protein. Semin Cell Dev Biol 24, 222225.CrossRefGoogle ScholarPubMed
Koizumi, T, Terada, T, Nakajima, K, et al. (2015) Identification of key neoculin residues responsible for the binding and activation of the sweet taste receptor. Sci Rep 5, 12947.CrossRefGoogle ScholarPubMed
Nakajima, K-I, Koizumi, A, Iizuka, K, et al. (2011) Non-acidic compounds induce the intense sweet taste of neoculin, a taste-modifying protein. Biosci Biotechnol Biochem 75, 16001602.CrossRefGoogle ScholarPubMed
Li, X, Staszewski, L, Xu, H, et al. (2002) Human receptors for sweet and umami taste. Proc Natl Acad Sci U S A 99, 46924696.CrossRefGoogle ScholarPubMed
Sanematsu, K, Kusakabe, Y, Shigemura, N, et al. (2014) Molecular mechanisms for sweet-suppressing effect of gymnemic acids. J Biol Chem 289, 2571125720.CrossRefGoogle ScholarPubMed
Pydi, SP, Jaggupilli, A, Nelson, KM, et al. (2015) Abscisic acid acts as a blocker of the bitter taste G protein-coupled receptor T2R4. Biochemistry 54, 26222631.CrossRefGoogle ScholarPubMed
Rachid, O, Simons, FER, Rawas-Qalaji, M, et al. (2010) An electronic tongue: evaluation of the masking efficacy of sweetening and/or flavoring agents on the bitter taste of epinephrine. AAPS PharmSciTech 11, 550557.CrossRefGoogle ScholarPubMed
Keast, RS & Breslin, PA (2005) Bitterness suppression with zinc sulfate and Na-cyclamate: a model of combined peripheral and central neural approaches to flavor modification. Pharm Res 22, 19701977.CrossRefGoogle ScholarPubMed
Szejtli, J & Szente, L (2005) Elimination of bitter, disgusting tastes of drugs and foods by cyclodextrins. Eur J Pharm Biopharm 61, 115125.CrossRefGoogle ScholarPubMed
Kim, MJ, Son, HJ, Kim, Y, et al. (2015) Umami-bitter interactions: the suppression of bitterness by umami peptides via human bitter taste receptor. Biochem Biophys Res Commun 456, 586590.CrossRefGoogle ScholarPubMed
Chaudhari, N, Yang, H, Lamp, C, et al. (1996) The taste of monosodium glutamate: membrane receptors in taste buds. J Neurosci 16, 38173826.CrossRefGoogle ScholarPubMed
Kurihara, K (2015) Umami the fifth basic taste: history of studies on receptor mechanisms and role as a food flavor. Biomed Res Int 2015, 189402.CrossRefGoogle ScholarPubMed
Zhang, F, Klebansky, B, Fine, RM, et al. (2008) Molecular mechanism for the umami taste synergism. Proc Natl Acad Sci U S A 105, 2093020934.CrossRefGoogle ScholarPubMed
Backes, M, Obst, K, Bojahr, J, et al. (2015) Rubemamine and rubescenamine, two naturally occurring N-cinnamoyl phenethylamines with umami-taste-modulating properties. J Agric Food Chem 63, 86948704.CrossRefGoogle ScholarPubMed
Ottinger, H & Hofmann, T (2003) Identification of the taste enhancer alapyridaine in beef broth and evaluation of its sensory impact by taste reconstitution experiments. J Agric Food Chem 51, 67916796.CrossRefGoogle ScholarPubMed
Zhang, Y, Venkitasamy, C, Pan, Z, et al. (2017) Novel umami ingredients: umami peptides and their taste. J Food Sci 82, 1623.CrossRefGoogle ScholarPubMed
Xu, H, Staszewski, L, Tang, H, et al. (2004) Different functional roles of T1R subunits in the heteromeric taste receptors. Proc Natl Acad Sci U S A 101, 1425814263.CrossRefGoogle ScholarPubMed
Kochem, M & Breslin, PAS (2017) Clofibrate inhibits the umami-savory taste of glutamate. PLOS ONE 12, e0172534.CrossRefGoogle ScholarPubMed
Dewis, ML, Phan, TH, Ren, Z, et al. (2013) N-geranyl cyclopropyl-carboximide modulates salty and umami taste in humans and animal models. J Neurophysiol 109, 10781090.CrossRefGoogle ScholarPubMed
Katsumata, T, Nakakuki, H, Tokunaga, C, et al. (2008) Effect of Maillard reacted peptides on human salt taste and the amiloride-insensitive salt taste receptor (TRPV1t). Chem Senses 33, 665680.CrossRefGoogle Scholar
Schindler, A, Dunkel, A, Stahler, F, et al. (2011) Discovery of salt taste enhancing arginyl dipeptides in protein digests and fermented fish sauces by means of a sensomics approach. J Agric Food Chem 59, 1257812588.CrossRefGoogle ScholarPubMed
Triki, M, Khemakhem, I, Trigui, I, et al. (2017) Free-sodium salts mixture and AlgySalt® use as NaCl substitutes in fresh and cooked meat products intended for the hypertensive population. Meat Sci 133, 194203.CrossRefGoogle ScholarPubMed
van Buren, L, Dotsch-Klerk, M, Seewi, G, et al. (2016) Dietary impact of adding potassium chloride to foods as a sodium reduction technique. Nutrients 8, 235.CrossRefGoogle ScholarPubMed
Nagai, T, Nii, D & Takeuchi, H (2001) Amiloride blocks salt taste transduction of the glossopharyngeal nerve in metamorphosed salamanders. Chem Senses 26, 965969.CrossRefGoogle ScholarPubMed
Figure 0

Fig. 1. Mechanisms of sweet, umami, bitter and fat taste perception by taste bud cells. The taste qualities of sweet, umami or bitter are sensed by the G protein-coupled receptor (GPCR) expressed on type II taste bud cells. Whereas, fat taste perception is mediated by both GPCR and cluster of differentiation 36 (CD36). Upon activation of GPCR, the signal is transmitted through βγ subunits Gβγ and α-gustducin to the downstream effectors phospholipase C β2 (PLC) and adenylate cyclase (AC), respectively. PLC β2 activation generates two second messengers, namely, diacylglycerol (DAG) and inositol 1,4,5-trisphosphate (IP3). IP3 acts on IP3 receptors on endoplasmic reticulum (ER) and increase the intracellular calcium levels. Whereas the AC mediates cyclic AMP (cAMP) production, which block the potassium channels on the plasma membrane. Both these pathways ultimately lead to the depolarisation of the taste receptor cells which lead to the opening of the transient receptor potential melastatin 5 (TRPM5) ion channel. Consequently, ATP is released through the Panx1 hemichannel into the extracellular space, which stimulates multiple targets including the gustatory afferent nerve fibres or the adjacent presynaptic cells which releases the neurotransmitters including serotonin (5-hydroxytryptamine; 5-HT) and noradrenaline into the synaptic cleft. PIP2, phosphatidylinositol 4,5-bisphosphate; T1R, taste receptor type 1; PDE, phosphodiesterase; PKA, protein kinase A; MEK, MAPK/ERK; MAPK, mitogen-activated-protein kinase; ERK, extracellular signal–regulated kinase; PKC, protein kinase C; DRK, delayed rectifying potassium; IP3R, IP3 receptor; SERCA, sarcoendoplasmic reticulum calcium transport ATPase. For a colour figure, see the online version of the article.

Figure 1

Table 1. Characteristics of candidate sweet and bitter taste modifiers/tastants and their associated mechanism of chemoreception

Figure 2

Table 2. Characteristics of candidate umami and salt taste modifiers/tastants and their associated mechanism of chemoreception

Figure 3

Fig. 2. Mechanisms of sour and salt taste perception by taste bud cells. Sour taste is triggered in type III cells by the intracellular proton concentration change triggered by protonated acids. In addition, several channels, including polycystin 2 like 1 (PKD2L1; transient receptor potential cation channel) and PKD1L3 have also been associated with sour taste. For salt taste, the putative candidate is the epithelial-type sodium channel (ENaC). The principal salt stimulus (sodium ion; Na+) can permeate through these cation channels on the apical surface of taste bud cells and trigger depolarisation. ΔVm, membrane potential change; TRPP3, transient receptor potential polycystic 3. For a colour figure, see the online version of the article.

Supplementary material: File

Deepankumar et al. supplementary material

Deepankumar et al. supplementary material 1

Download Deepankumar et al. supplementary material(File)
File 25.6 KB