Skip to main content

Behavioral genetics and taste

Abstract

This review focuses on behavioral genetic studies of sweet, umami, bitter and salt taste responses in mammals. Studies involving mouse inbred strain comparisons and genetic analyses, and their impact on elucidation of taste receptors and transduction mechanisms are discussed. Finally, the effect of genetic variation in taste responsiveness on complex traits such as drug intake is considered. Recent advances in development of genomic resources make behavioral genetics a powerful approach for understanding mechanisms of taste.

Introduction

The chemical senses, gustation and olfaction, have provided a candidate media for investigation of the effects of major genes on behavior, owing to considerable natural variation for both senses among individuals or strains of mice and rats. Genetic analysis of behavioural responses to bitter and sweet taste stimuli in mice facilitated the recent discovery of receptor gene families for bitter, sweet and amino acid taste (for recent reviews, see [1–4]). The function and specificity of taste receptors continues to be elucidated based on not only in vitro expression assays and transgenic approaches, but also based on studies of gene variation and evolution (e.g. [5–8]).

Taste, beyond its purely sensory function, is also inextricably linked to a larger set of behaviors. The gustatory system is anatomically located at the beginning of the alimentary canal, and as such is a key player in homeostatic systems dealing with nutrient and fluid intake. Taste and feeding are linked in terms of reciprocal connectivity between brain centers (e.g. [9, 10]). The consumption of sweeteners, for example, often involves a complex integration of the peripheral sensory system, the central nervous system and post-ingestive events. Sweet taste detection in mammals has been shown to be heavily modulated by post-ingestive feedback, including neural or hormonal factors (e.g. [11–13]). This feedback extends to the level of the taste receptor cells and afferent nerves [14, 15]. Nutritive sweeteners provide post-ingestive stimulation [16], probably due to their calories. These interactive mechanisms of consummatory responses to sweeteners imply that ingestion is a complex behavior, and is likely to be determined by multiple genes.

Although studies of non-mammalian organisms such as fruit flies hold tremendous potential for elucidating genetic basis of taste behavior and physiology [17, 18] this review will focus on studies of taste genetics in mammals, primarily in mice, and to a lesser extent in rats and humans. In particular, we will concentrate on the characterization of phenotypic diversity among inbred strains of mice, as this naturally-occurring variation has proved crucial for genetic analysis and the eventual identification of G-protein coupled taste receptors. Hence, the review is arranged by taste quality, including those characterized by humans as tasting sweet, umami (savory), bitter and salty. Although candidate sour taste receptor molecules have been recently identified [19, 20], less progress has been made regarding genetic approaches to sour (acid) taste, and this quality is not discussed here.

Measuring taste behavior

Taste perception involves several aspects, such as intensity, quality and hedonic value of the taste sensation. In humans, these aspects of taste perception are usually assessed using verbal information, e.g., by plotting sensation intensity on a scale with verbal descriptors or by reporting a difference between samples. Early studies of human genetic variation of taste sensitivity concentrated on either responses to a single concentration of stimulus (i.e., tasters vs. non-tasters of phenylthiocarbamide (PTC)) or measurement of absolute thresholds [21]. A more expansive view of taste ability, and therefore phenotypic differences, may be gained from analyses of the perceived taste intensity [22, 23].

Assessment of taste perception in non-human animals relies on recording of behavior elicited by taste stimuli using a number of different techniques [24]. Each of these techniques characterizes some, but not all aspects of taste perception. For example, hedonics of taste sensation can be assessed based on consummatory responses in naive animals, but assessing taste quality involves conditioning of animals. Many of these techniques have been adapted for use in mice and can be used in genetic experiments (Table 1).

Table 1 Methods to characterize taste in non-human animals

Traditionally, behavior genetic studies of taste in the mouse have been conducted using two-bottle intake tests, e.g. [25–28]. These tests simply measure the consumption of a taste stimulus over a 24- or 48-h period, relative to the consumption of water. Sweet-tasting stimuli are generally preferred to water, whereas bitter-tasting stimuli are avoided. Such tests, while amenable for testing large cohorts of mice required for genetic analysis, do not easily allow for the dissociation of sensory ability from post-ingestive factors such as satiety or toxicity. Brief-access tests minimize post-ingestive cues by restricting trial lengths to short durations (< 30 s). The dependent measure is number of licks taken from a particular stimulus, typically expressed as a ratio (licks to stimulus/licks to water). Such tests have been effectively used for high-throughput screening of taste function in mouse populations, especially for compounds with an aversive taste [29, 30].

Other tests of taste function in the mouse include measurements of threshold detection or the ability to discriminate between two stimuli [31, 32]. Conditioned taste aversion (CTA) is a commonly used paradigm to estimate perceptual similarity or dissimilarity between stimuli. The presentation of a novel taste stimulus is paired with a stimulus that produces temporary gastric distress, usually an intraperitoneal dose of LiCl. After pairing, the animal will avoid consumption of the conditioned taste stimulus, and this CTA will generalize to stimuli with similar perceptual qualities.

Finally, electrophysiological recordings of activity in the afferent gustatory nerves often corroborate behavioral taste tests in non-human animals. The major gustatory nerves are the chorda tympani (CT) branch of the facial (VII) nerve and the glossopharyngeal (IX) nerve. The CT predominantly innervates taste buds on the anterior tongue, whereas the IXth nerve innervates taste buds contained in trenches on the posterior tongue. Studies of gustatory nerve activity help to elucidate whether genetic effects on taste perception have peripheral or central origin.

Genetic analysis of taste behavior

In genetic terms, the taste response of an individual represents a phenotype, i.e., an observable property of an organism produced by genetic and environmental factors. A typical goal of genetic studies is to characterize the genetic factors contributing to the phenotype. This is usually achieved by analyzing populations of related individuals, such as twins or families in humans, or crosses between inbred strains of mice or rats. An initial goal of the genetic analysis is to establish whether the phenotypical variation among individuals has a genetic component (i.e., is heritable). Once heritability is confirmed, chromosomal localization of genes responsible for the heritable component of the phenotype can be found using linkage analysis. The linkage analysis involves assessment of association between genetic markers distributed throughout the genome and the phenotype. Some linkage studies in rodents use recombinant inbred strains, which are produced by intercrossing two parental inbred strains, obtaining the second generation of hybrids (F2), and subsequent inbreeding (brother × sister mating) of multiple strains. After a genetic locus affecting a phenotype is mapped to a particular chromosome (Chr), subsequent studies can identify the molecular nature of the corresponding gene. Because gene identification is based primarily on the chromosomal position of the gene, this approach is called positional cloning.

Sweet and umami taste

Genetic variation in sweet taste responsiveness

Taste sensitivity to sweeteners varies among individual humans, and this variation may be determined genetically [33–42]. Prominent genetic differences in taste responses to sweeteners also exist among inbred strains of mice. These differences were shown using different experimental techniques and a variety of sweeteners (such as sucrose, glucose, dulcin, saccharin, acesulfame, glycine, D-phenylalanine and L-glutamine). Mice from different strains vary in taste responses to sweeteners assessed using long-term preference tests [26, 27, 43–53], single-bottle tests [54], brief-access tests based on lick recording [55, 56], taste detection thresholds [31], CTA generalization [57], and responses of gustatory nerves [58–60]. These studies have shown that responses to many of these sweeteners (e.g., sucrose, glucose, dulcin, saccharin and acesulfame) closely correlate among mouse strains, suggesting a common genetic basis for sweet taste. However, responses to some sweet-tasting amino acids display somewhat different patterns of strain differences. Strain differences in consummatory responses to sweeteners have also been reported for rats [61–63] and hamsters [64].

A few mouse strains with large differences in sweet taste responses were used to produce crosses for analyses of genetic and physiological mechanisms underlying behavioral responses to sweeteners. The most detailed physiological analysis was conducted using mice from the C57BL/6 (B6) strain with high sweetener preferences and mice from the 129 strains with low sweetener preferences. Compared with 129 mice, B6 mice had higher preferences for a large number of sweeteners, including sugars (sucrose and maltose), sweet-tasting amino acids (glycine, D-phenylalanine, D-tryptophan, L-proline and L-glutamine), and several but not all non-caloric sweeteners (saccharin, acesulfame, dulcin, sucralose and SC-45647) [46, 50–52, 65]. This phenotypic difference is specific to sweet-taste processing, and is not due to a generalized difference in taste responsiveness or differences in appetite [66, 67].

Differences between B6 and 129 mice in preference for a sweet-tasting amino acid glycine [65] appear to depend on mechanisms distinct from those affecting responses to many other sweeteners. Both B6 and 129 mice generalized a CTA between glycine and several other sweeteners, demonstrating that they perceive the sucrose-like taste of glycine. Thus, the lack of a strong glycine preference by 129 mice cannot be explained by their inability to perceive its sweetness [68]. Despite differences in glycine intakes and preferences, CT responses to glycine are similar in mice from both strains [58]. Neither behavioral nor neural responses to glycine are influenced by the Tas1r3 genotype [31, 69], suggesting that variation in taste responses to glycine depends on other genes.

Some genetic analyses of sweetener consumption by mice yielded evidence that it is influenced by a single locus, named Sac (saccharin preference) [46, 50, 70, 71], whereas other experiments indicated that more than one gene is involved [47, 50, 51, 72, 73]. The apparent discrepancy on whether single-gene or multi-gene model better describes genetic variation in sweetener preferences is likely due to use of different progenitor strains and types of mapping panels, different sweetener solutions tested, and different quantitative analyses used in these studies.

The saccharin preference (Sac) locus and the Tas1r3gene

Using long-term two-bottle tests, Fuller [70] demonstrated that differences in saccharin preferences between the B6 and DBA/2J inbred strains largely depend on a single locus, Sac, with a dominant Sacballele present in the B6 strain associated with higher saccharin preference, and a recessive Sacdallele present in the DBA/2J strain associated with lower saccharin preference. Subsequent studies confirmed this finding in the BXD recombinant inbred strains, and in crosses between the B6 and DBA/2 or between the B6 and 129 strains [46, 50, 71, 72, 74, 75]. In addition to sweetener preferences, the Sac genotype influences the afferent responses of gustatory nerves to sweeteners [75, 76], which indicated that the Sac gene is involved in peripheral taste transduction and may encode a sweet taste receptor.

The Sac locus has been mapped to the subtelomeric region of mouse Chr 4 [50, 72, 74–76] (see also Figure 1). A positional cloning study at the Monell Chemical Senses Center showed that the Sac locus corresponds to the Tas1r3 gene [77, 78]. This study involved a high-resolution linkage analysis of a B6 × 129 F2 intercross, the marker-assisted selection of a 129.B6-Sac congenic strain, physical mapping that involved construction of a contig of bacterial artificial chromosome (BAC) clones, BAC sequencing, and sequence analysis of candidate genes. One of the genes within the critical interval of the Sac locus was a G protein-coupled receptor gene, Tas1r3 (taste receptor, type 1, member 3). Based on the effects of the Sac genotype on peripheral sweet taste responsiveness [75, 76], and on involvement of a G protein-coupled mechanism in sweet taste transduction [79], Tas1r3 was selected as the most likely candidate for the Sac locus (Figure 1).

Figure 1
figure 1

Positional identification of the Sac (saccharin preference) locus. a. Linkage map of mouse distal Chr 4 based on data from the B6 × 129 F2 intercross. The X axis shows distances between markers in recombination units (cM). The Y axis shows the logarithm of the odds ratio (LOD) scores for sucrose and saccharin consumption. The LOD score peaks (indicated by black triangles) and confidence intervals (solid horizontal line for sucrose, 4.5 cM, and dotted horizontal line for saccharin, 5.3 cM) define the genomic region of the Sac locus. b. Average daily 17 mM saccharin consumption by mice from parental 129 and B6 strains (left), B6 × 129 F2 hybrids (center), and congenic 129.B6-Sac mice (right) in 96-hr two-bottle tests with water (means ± SE). Tas1r3 genotypes of the F2 and congenic mice and mouse numbers are indicated on the bars. Differences between parental strains and among the F2 and congenic genotypes were significant (p < 0.0001, ANOVA). F2 and congenic B6 homozygotes and heterozygotes for Tas1r3 did not differ from each other, and had higher saccharin intakes compared with 129 homozygotes (p < 0.0001, post hoc tests). c. Linkage map of the Sac-containing region defined based on the size of the donor fragment in the 129.B6-Sac congenic strain (black box). Distances between markers were estimated based on the B6 × 129 F2 intercross (see panel a). d. A contig of bacterial artificial chromosome (BAC) clones and physical map of the Sac region. BAC clones are represented by horizontal lines. Dots indicate marker content of the BAC clones. e. Genes within the Sac-containing interval. Filled areas indicate predicted genes. Arrows indicate the predicted direction of transcription. Figure reproduced with permission from [253].

The Tas1r3 gene contains 6 coding exons and is translated into an 858-amino acid protein, T1R3, with a predicted secondary structure that includes seven transmembrane domains and a large hydrophilic extracellular N-terminus. This structure is typical of the class C G protein-coupled receptor family, which includes the metabotropic glutamate and extracellular calcium-sensing receptors. T1R3 belongs to a small family of G protein coupled receptors, T1R, which also includes the T1R1 and T1R2 proteins. The three mouse T1R genes are located on distal Chr 4 in the order: Tas1r2 (70.0 cM, or 139 Mb, NCBI Build 36) – Tas1r1 (81.5 cM, or 151 Mb) – Tas1r3 (83.0 cM, or 155 Mb). The Tas1r1 and Tas1r2 genes were excluded as candidates for the Sac locus based on their more proximal chromosomal location [76, 80, 81]. All three T1R members are expressed in taste receptor cells [53, 80–85]. When T1R3 is co-expressed in a heterologous system with T1R2, it functions as a sweet receptor [85–87], but T1R3 may also function as a low-affinity sugar receptor alone, probably as a homodimer [88]. Tas1r3 has a human ortholog, TAS1R3, residing in a region of conserved synteny in the short arm of human Chr 1 (1p36) [89].

If Tas1r3 is identical to Sac, substitution of Tas1r3 alleles must result in phenotypical changes attributed to the Sac locus. Introgression of the 194-kb chromosomal fragment containing the Tas1r3 allele from the high-sweetener preferring B6 strain onto the genetic background of the 129 strain fully rescued its low sweetener preference phenotype: sweetener intake of the congenic mice was as high as that of mice from the donor B6 strain [77] (Figure 1). Equivalent phenotype rescue results were obtained in a transgenic experiment [85]. These data demonstrate that substitution of Tas1r3 alleles results in behavioral changes attributed to the Sac locus and therefore provides a proof that Tas1r3 is identical to Sac, and that the T1R3 receptor responds to sweeteners. Further evidence for identity of the Sac locus and Tas1r3 gene was obtained in studies of mice with targeted mutations of the Tas1r3 gene, which were found to be deficient in taste responses to sweeteners [88, 90]

Identity of Sac and Tas1r3 implies that there must be Tas1r3 polymorphisms, resulting in variation of sweet taste responses attributed to allelic variants of the Sac locus. Although several candidate functional polymorphisms were proposed in studies that identified the Tas1r3 gene [80, 81, 83–85], these studies lacked a proper quantitative analyses of gene-phenotype associations.

Reed et al [53] conducted a comprehensive quantitative analysis of the Tas1r3 sequence variants associated with saccharin preference using 30 genealogically diverse inbred mouse strains. Genomic sequences including Tas1r3 exons, introns, upstream and downstream regions were examined, so that polymorphisms affecting amino acid composition or potential regulatory regions could be detected. The strongest association with saccharin preference was found for a haplotype including three sites: nucleotide (nt) -791 (3 bp insertion/deletion), nt +135 (Ser45Ser), and nt +179 (Ile60Thr) (Figure 2). Lack of differences in the Tas1r3 gene expression in the taste tissues of mice with different Tas1r3 haplotypes suggested that the polymorphisms that do not change amino acid sequence of the T1R3 protein (nt -791 and nt +135) are unlikely to affect receptor function. Therefore, the amino acid substitution of isoleucine to threonine at position 60 (Ile60Thr) was predicted to be a functional polymorphism [53]. This prediction was subsequently confirmed in an in vitro study showing that a corresponding site-directed mutation changes binding affinity of the T1R3 protein to several sweeteners [91].

Figure 2
figure 2

Preference for 1.6 mM saccharin by mice from inbred strains with different Tas1r3 genotypes at the T/C variant site at nucleotide position +179 (relative to the first nucleotide in the ATG start codon of the Tas1r3 gene). This polymorphism results in amino acid substitution of isoleucine to threonine at position 60 (I60T), in the extracellular N-terminus of the predicted T1R3 protein. Closed circles denote means for C57BL/6, C57L/J, CAST/Ei, CE/J, FVB/NJ, I/LnJ, IS/Cam, KK/HlJ, NOD/LtJ, NZB/BlNJ, P/J, RBF/DnJ, SEA/GnJ, SJL/J, SM/J, SPRET/Ei, ST/bJ and SWR/J strains with +179 T genotype. Mice from these strains strongly preferred saccharin (average preference score 88 ± 2%, Mean ± SE; n = 18). Open circles show means for 129P3/J, A/J, AKR/J, BALB/cByJ, BUB/BnJ, C3H/HeJ, CBA/J, DBA/2J, LP/J, PL/J, RF/J and RIIIS/J strains with +179 C genotype. Mice from these strains were indifferent to or only weakly preferred saccharin (average preference score 59 ± 3%, n = 12; p = 0.00000000012, t-test). Despite the strong phenotypical effect of the Tas1r3 genotype, there is also substantial variation in saccharin preference within each genotype group. As a result, Tas1r3 genotype explains only 78% of genetic variation in saccharin preferences among the inbred strains; the remaining 22% of genetic variance is attributed to the effect of other genes. Adapted with permission from [53].

Ligand specificity of the T1R3 receptor

In vitro studies of T1R proteins have indicated that they function as broad-spectrum sweet and umami receptors (Table 2) [85, 87, 92]. Several aspects of these in vitro studies emphasize importance of an in vivo approach to characterize ligand-receptor interactions. First, discrepancies between results obtained using different expression systems leave open a question of whether responsiveness or unresponsiveness to a particular sweetener reflects in vivo sensitivity of the receptor, or is an artifact of the in vitro system. Second, responses of the heterologously expressed mouse receptors to amino acids are not always consistent with mouse behavioral responses to these stimuli. For example, sweet L-proline and L-threonine did not activate the T1R2 and T1R3 combination, but instead activated the T1R1 and T1R3 combination, which also responded to some umami-tasting and bitter (e.g., L-phenylalanine) compounds [87].

Table 2 Deorphanized taste receptors

The in vivo approach to characterize ligand specificity of the T1R3 receptor is based on an assumption that if a response to a compound is affected by Tas1r3 genotype, then this compound activates a receptor involving T1R3. Several earlier genetic mapping studies have shown that in addition to saccharin preferences, the Sac locus also affects consumption of sucrose, acesulfame [46, 50, 74, 75], and ethanol, which has a sweet taste component [93], and also chorda tympani responses to saccharin and sucrose [75, 76]. Inoue et al. [69] examined behavioral and neural responses to a larger set of sweeteners in the F2 hybrids between the B6 and 129 strains. They found that the Tas1r3 genotype affected consumption of the sweeteners sucrose, saccharin and D-phenylalanine, but not glycine. For CT responses, significant linkages to Tas1r3 were found for the sweeteners sucrose, saccharin, D-phenylalanine, D-tryptophan and SC-45647, but not glycine, L-proline, L-alanine or L-glutamine. No linkages to the Tas1r3 chromosomal region were detected for behavioral or neural responses to non-sweet quinine, citric acid, HCl, NaCl, KCl, monosodium glutamate (MSG), inosine 5'-monophosphate (IMP) or ammonium glutamate. Thus, allelic variation of the Tas1r3 gene affects taste responses to many but not all sweeteners, suggesting that a wide variety of sweeteners can activate a receptor involving T1R3. Lack of the effect of the Tas1r3 genotype on taste responses to some sweeteners, such as glycine, can be explained by several possible mechanisms: (i) sweetener binding to the T1R3 receptor at a site that is not affected by the polymorphic variants; (ii) binding to the T1R2 receptor; (iii) existence of another sweet taste receptor binding these sweeteners. These mechanisms can be examined using mice with targeted mutations of the Tas1r genes, but there are currently no data on taste responses to glycine in Tas1r knockout mice.

Other genes involved in sweet taste responses

Multigenic inheritance of sweetener preferences was shown in a number of studies [47, 50, 51, 72, 73]. Accordingly, several lines of evidence indicated that allelic variation of the mouse Tas1r3 locus does not account for all the genetically determined differences in sweetener preferences. Analysis of multiple inbred mouse strains has shown that the Tas1r3 genotype explains only 78% of genetic variation in saccharin preference [53] (Figure 2). In the B6 × 129 F2 cross, the Tas1r3 genotype explained 64 – 96% of genetic variation in preference scores for different sweeteners, 10 – 35% of genetic variation in sweetener intakes, and 37 – 92% of genetic variation in CT responses to sweeteners [69, 75]. Responses to sweeteners in brief-access tests differ among mouse strains but do not seem to be associated with Tas1r3 alleles [55]. Thus, a substantial part of the genetic variation in taste responses to sweeteners among mouse strains is attributed to loci other than Tas1r3. Taste responses to glycine provide a remarkable example: although there are substantial differences among mouse strains in responses to glycine [50, 65], this variation is not attributed to the Tas1r3 genotypes [31, 69]. Consistent with the mouse work suggesting effects of genes other than Tas1r3, variation in saccharin preferences in rats is not associated with sequence variants of the rat Tas1r3 gene [94], and therefore it must be attributed to the effects of other genes (see also [95]).

Because a sweet taste receptor appears to be a heterodimer of T1R2 and T1R3 proteins, sequence variants of the Tas1r2 gene potentially can also result in variation in sweet taste responses. There is no evidence that Tas1r2 variation contributes to mouse strain differences in responses to sweeteners. However, sequence variation among the Tas1r2 orthologs underlies some species differences in sweet taste perception. Inability of rodents to perceive sweetness of several compounds that taste sweet to humans is attributed to variation between human and rodent orthologs of Tas1r2 [87, 96]. Lack of preference for sugars and some other sweeteners in cats (Felidae) [97, 98] is due to pseudogenization of the Tas1r2 gene, and thus inability to produce a T1R2 + T1R3 heterodimeric sweet taste receptor [8].

One of the genetic loci affecting sweet taste responses is dpa (D-phenylalanine aversion), which affects ability of mice to generalize a CTA between D-phenylalanine and sucrose, inferring that dpa affects ability to detect sweetness of D-phenylalanine. The dpa locus also affects responses of sucrose-sensitive fibers of the CT nerve to D-phenylalanine. B6 mice carry a dominant allele of dpa that determines an ability to recognize the sweetness of D-phenylalanine, whereas BALB/c mice carry a recessive dpa allele conferring inability to detect D-phenylalanine sweetness. The dpa locus was mapped to proximal Chr 4, a region distinct from the subtelomeric Chr 4 harboring the Tas1r genes [99–102]. It was suggested that the dpa locus can also affect responses to sweeteners in two-bottle tests [51]. Consistent with this, a locus on proximal Chr 4, in the dpa region, was found to be suggestively linked to consumption of, and CT responses to, sucrose [75]. An epistatic interaction between effects on sucrose intake of this locus and the Tas1r3 locus suggests that these two loci may encode interacting components of sweet taste transduction [75].

In summary, these data show that sweetener preference has a complex genetic determination. In addition to the Tas1r3 gene, other genetic loci play the role in genetic variation of taste responses to sweeteners. These other genes may encode additional peripheral taste transduction elements, including novel taste receptors. Alternatively, they may be involved in central mechanisms of ingestive behavioral responses or feedback mechanisms of modulation of sweet taste responses.

Umami taste genetics

Umami is a taste quality exemplified by taste of MSG and evoked also by some amino acids and purine 5'-nucleotides. There is a strong evidence that a heterodimer of T1R1 and T1R3 proteins functions as an umami taste receptor in humans and is a more broadly tuned in rodents to respond to L-amino acids (Table 2) [87, 88, 90, 92]. A splice variant of a metabotropic glutamate receptor, mGluR4, was also proposed as a taste receptor for glutamate [103].

Little is known about genetic variation in umami taste responses. Humans appear to differ in perception of glutamate taste [104], but it is not known whether there is a genetic basis for this. A comparison of the CT nerve responses in three inbred mouse strains has shown differences in a synergistic effect between MSG and 5'-guanylate [105].

In the long-term two-bottle preference tests, mice from the B6 strain consumed more MSG and IMP than did mice from the 129 strain [106]. The strain difference in MSG consumption was in the opposite direction to the strain difference in NaCl consumption [66, 107]. Although the B6 mice have higher avidity for both MSG and sweeteners than do the 129 mice [65, 66, 106], there is no correlation between preferences for these solutions in the F2 hybrids derived from these two strains [106]. Thus, differences in MSG consumption between B6 and 129 mice are not related to the strain differences in salty or sweet taste responsiveness.

The role of the afferent gustatory input in these strain differences was examined by measuring integrated responses of the CT and IX nerves to umami taste stimuli. In the CT, responses to MSG and monoammonium L-glutamate were similar in B6 and 129 mice, but responses to IMP and guanosine-5'-monophosphate were lower in B6 than in 129 mice. Responses to umami stimuli in the IX nerve did not differ between the B6 and 129 strains [108]. Thus, the increased ingestive responses to the umami stimuli in B6 mice are accompanied by either unchanged or decreased neural responses to these stimuli. Lack of support for the role of the gustatory nerves in the enhanced consumption of MSG and IMP by B6 mice suggests that it is due to some other factors. A prior exposure to MSG affects subsequent MSG consumption [106], suggesting that it can be modulated by postingestive effects. The strain differences in gustatory neural responses to nucleotides but not glutamate [108] suggest that these compounds may activate distinct taste transduction mechanisms.

The T1R3 protein is involved in transduction of both sweet and umami tastes, and a disruption of the Tas1r3 gene diminishes behavioral or neural responses to umami taste stimuli [86–88, 90]. Hence, variation of the Tas1r3 gene might affect umami taste responses. However, an analysis of the F2 hybrids between the B6 and 129 inbred mouse strains has shown that the Tas1r3 allelic variants do not affect behavioral or neural taste responses to umami stimuli [69]. Thus, although the T1R3 receptor is involved in transduction of umami taste, the B6/129 sequence variants affecting its sensitivity to sweeteners do not affect its sensitivity to umami compounds.

Bitter taste

Bitter taste receptor genes

A large and diverse array of molecules evoke the sensation of bitterness, and the ability to detect and avoid these stimuli is assumed to have evolved as a mechanism to prevent ingestion of toxic foods [109]. Indeed, a strong correlation exists in the animal kingdom between bitter sensitivity and tolerance to toxic compounds [110]. A Tas2r family of G-protein coupled receptor genes linked to bitter taste sensitivity was identified in 2000 by laboratories led by Nicholas Ryba, Charles Zuker [111, 112] and Linda Buck [113]. The T2R receptors are related to class A GPCRs and are characterized by a short N terminus with a potential transmembrane ligand-binding domain. To date, about 28 human and 36 mouse intact bitter taste receptor genes have been identified, although only a few have been de-orphanized (Table 2) [114].

The PTC polymorphism in humans and mice

The first example of a genetic basis to taste ability involved the serendipitous discovery of human differences in sensitivity to the bitter-tasting compound PTC [115]. Population surveys indicated a dimorphism in taste sensitivity for this substance, with individuals falling into two categories: "tasters" and "nontasters" [116, 117]. Pedigree analysis suggested phenotypic control by a single, autosomal locus [118, 119] although more recent studies postulated that PTC inheritance was better described by a polygenic model [120, 121]. Linkage studies in humans supported associations with several chromosomal loci, including the KEL blood group antigen on Chr 7q (e.g. [122, 123]). Dennis Drayna and colleagues [124, 125] have shown that this locus corresponds to a polymorphic bitter taste receptor gene TAS2R38 responsible for up to 85% of the variation in PTC sensitivity among human subjects. A secondary QTL on Chr 16p also has an effect on PTC aversion, although the nature of this gene is not known [124].

From the early discovery of human variation in PTC taste sensitivity, investigators proceeded to animal models of bitter taste genetics. Richter and Clisby [126] reported individual variation in PTC avoidance thresholds of rats, but the genetic basis for this was not pursued. Klein and DeFries [127] found that two-bottle intake of PTC varied in mice, with BALB/c mice significantly more sensitive to the bitter taste of PTC than other inbred strains, including C57BL/Ibg. A Mendelian cross (F1, F2, F3, B1, and B2 generations) between these two strains yielded segregation ratios consistent with phenotypic control by a single autosomal locus, with a dominant taster allele. It was later recognized that PTC avoidance by BALB/c mice generally develops across 10–12 consecutive days of testing [128] (Figure 3). PTC is extremely toxic to mice (oral LD50 10 mg/kg), comparable to the rodent poison strychnine (oral LD50 2 mg/kg). Confining stimulus access to shorter durations helps to minimize or eliminate post-ingestive consequences (e.g. [29, 129–132]). A direct comparison of intake tests and brief-access procedures demonstrated that differential avoidance of PTC among inbred strains depended on the amount of stimulus consumed, and not necessarily on immediate orosensory or taste cues [133]. When injected intraperitoneally, PTC was nearly as effective as LiCl in serving as the unconditioned stimulus in a CTA experiment [134]. Strain differences in PTC avoidance that develop over time therefore likely reflect other variables in addition to sensory ability, such as sensitivity to its toxic effects, or differences in the speed of acquisition of a CTA.

Figure 3
figure 3

Aversion to PTC in taster mice develops over several days. Preference ratios (mean ± SE) for BALB/cBy (BALB), C3HeB/Fe (C3), and BALB × C3 F1 (n = 10/strain) to 0.03 mM PTC over six consecutive 48-h tests. The dotted line represents a preference score of 0.5, which indicates equivalent consumption from solution and water tubes. Preference ratios for BALB and F1 mice decreased following the initial test period; by the final test period these mice were strongly avoiding PTC. C3 mice remained indifferent to the taste of PTC across the entire test period. Strain differences in "developed aversion" to PTC have a genetic basis, with complete dominance of the avoider phenotype. However, the avoidance phenotype does not necessarily reflect increased bitter taste sensitivity. Figure from Boughter (unpublished).

Sucrose octaacetate (SOA) and quinine aversions and bitter taste receptor genes

The genetic basis of differences in intake of bitter compounds other than PTC has been studied throughout the last several decades, including substantial contributions from the laboratories of Ian Lush and Glayde Whitney (see [135, 136]). Inbred strains differ in sensitivity to the commonly-used bitter stimuli, quinine and SOA, assessed using two-bottle preference tests [66, 137–140]. For SOA, the strain difference is particularly robust as it occurs over a 2–3 log-step concentration range. SOA aversion appeared to be a fairly unambiguous model of monogenic control: Aversion ratios among segregating generations of mice were consistent with variation at a single autosomal locus (Soa, sucrose octaacetate aversion), with the taster allele dominant over either a "demitaster" (partial sensitivity) or nontaster allele [138, 141, 142]. Results of both brief-access behavioral tests and peripheral taste nerve electrophysiology yielded concentration-by-concentration similarity with two-bottle data, confirming that the strain difference was gustatory in nature [132, 140, 143]. Relevant to this finding is the fact that SOA is a non-toxic, apparently non-odorous compound. Soa was subsequently mapped to distal mouse Chr 6 near proline-rich salivary protein genes Prp2 and Prh1 [136, 144, 145]. At least one other study has found evidence for polygenic control of SOA aversion [146], and interestingly, the aversion phenotype is subject to environmental modulation: unlike control tasters, taster mice raised on drinking water adulterated with a normally avoided concentration of SOA display only minimal avoidance of SOA in a two-bottle test with water [147].

Unlike responses to SOA, responses to quinine in two-bottle tests were demonstrated to be under polygenic control in segregating crosses or recombinant inbred strains [148–150]. However, linkage studies positioned a locus with major effects on quinine intake, dubbed Qui (quinine sensitivity, taste), in a close proximity with the Prp2 and Prh1 genes on distal Chr 6 [139, 151, 152]. Lush also reported linkage to this region for aversion to other bitter compounds, including cycloheximide, raffinose undecaacetate, and copper glycinate (Figure 4; [136, 151, 153]) and sagely hypothesized the existence of a cluster of bitter taste genes at this location, which "may have evolved from one original bitterness gene by a process of local duplication and differentiation" [136]. A recently characterized bitter taste receptor gene family in mice (36 Tas2r genes) forms two clusters on distal Chr 6 and one on Chr 15, including 24 genes at the Soa and Qui loci that collectively comprise a large haplotype for quinine taste sensitivity (Figure 5) [30, 111, 113, 114]. Indeed, analysis suggests extensive gene duplication among the Tas2r genes [114]. Physical mapping of the Soa-Qui locus in strains of quinine taster (B6) and "non-taster" (DBA/2J) mice reveals considerable variance across the Tas2r genes [30]: Only two of 24 alleles were identical in both strains at the amino acid level. Furthermore, several of genes were either pseudogenes or deleted altogether in the nontaster strain, although a specific receptor-ligand relationship has not yet been uncovered.

Figure 4
figure 4

Genome-wide interval mapping of taste responses to bitter and sweet compounds: 0.1 – 0.4 mg/ml quinine, 0.3 mM beta-lactose acetate (BLA), 0.4 mM raffinose undecaacetate (RUA), 10 mM copper glycinate and 3.2 mM saccharin. LRS = Likelihood Ratio Score. The taste stimuli were tested in the two-bottle preference tests in the BXD recombinant inbred mouse strains. Phenotype data were previously published [46, 72, 151, 153] and are available at http://www.genenetwork.org. Mapping was conducted using WebQTL software http://www.genenetwork.org. The interval mapping illustrates previously published linkages for avoidance of all four bitter-tasting stimuli to Chr 6 and for saccharin preference to Chr 4 (Sac/Tas1r3 locus). A recent study of bitter taste in BXD mice using brief-access procedures demonstrates that the QTL on Chr 6 reflects orosensory processing, and not post-ingestive effects, of the bitter stimulus quinine [30].

Figure 5
figure 5

A map of the cluster of the Tas2r bitter taste genes on distal mouse Chr 6. Twenty-four intact Tas2r genes map to distal Chr 6 (black). The Tas2rs are found in two subclusters on either side of the polymorphic marker D6Mit13 (red) and two genes encoding proline-rich salivary proteins (Prp2 and Prh1; red). Map positions are in Mb (Build 33 assembly of the B6 genome). Figure copyright [30].

Among bitter receptor genes in the mouse, only two Tas2rs have been de-orphanized thus far with respect to ligand, one responding exclusively to cycloheximide, the other to PROP and denatonium [5, 154, 155]. Several human T2R receptors, including TAS2R38, have also been characterized with respect to ligand binding (Table 2).

Evolution of the bitter taste receptor gene family

Prominent features of the bitter receptor gene family in vertebrates include a high rate of non-synonymous (i.e. resulting in amino acid change) substitutions, and a relatively conserved family size (~15 – 33 functional genes) with multiple orthologues among mammalian species [156]. The most divergent portions of the Tas2r genes are those that code for extracellular domains, a possible site of ligand binding [114, 157]. The particular nature of the high rate of mutations in this gene family has led to speculation of relaxation of selective constraint and loss of function in primate species [158, 159]. On the other hand, analysis of TAS2R haplotypes among humans shows a greater diversity than expected, and suggests strong effects of natural selection [6, 160]. The number of functional receptors may have decreased in humans and other primates as the chemosensory function of these species has diminished in importance relative to other mammals, but positive selection has shaped the evolution of bitter taste receptors in response to variation in local plant species that are toxic.

Salt taste

A prototypical salty taste stimulus is NaCl, but several other compounds (e.g., chlorides and other salts of Na, Li and K) also evoke salty taste. Sodium enters epithelial cells through the sodium-selective epithelial channel ENaC. ENaC is a member of the degenerin/ENaC superfamily of ion channels and is inhibited by a diuretic, amiloride. Taste receptor cells express amiloride-blockable, Na+-selective channels that share a number of physiological properties with the ENaCs (for review, see [161]). Thus, a large portion of NaCl taste is likely dependent on the influx of sodium through ENaC. However, whether ENaC plays the role of a salt taste receptor requires further studies.

Sodium is an important nutrient, and its consumption is under a strong homeostatic control [162, 163]. Therefore, results of the long-term preference tests with sodium salts may be affected not only by perception of their taste, but also by postingestive effects. Thus, additional evidence is usually needed to link variation in salt consumption with differences in salty taste perception.

Salt consumption may be under genetic control in humans [164]. Inbred strain differences in voluntary NaCl consumption have been reported in both mice [28, 66, 107, 166–169] and rats [170–174]. Although these genetic differences in NaCl intake or preference are often profound, it is not clear whether they originate from differences in taste perception or are due to other factors. A few studies undertook a genetic analyses of NaCl consumption in mice [73] and rats [175–179]. In rats, salt intake was found to be linked to the Y Chr [177, 180]. No linkage data for NaCl consumption in mice have been published.

Despite large strain variation in NaCl preferences, most studies found either small or no differences in integrated responses of the gustatory nerves to NaCl among strains of mice [108, 166, 181–183] and rats [184, 185], with some exceptions [186].

In rodents, lingual application of amiloride suppresses the chorda tympani responses to NaCl [187]. There are mouse and rat strain differences in sensitivity of the chorda tympani response to amiloride. Initial studies suggested that amiloride sensitivity of the CT nerve response to NaCl is associated with behavioral responses to NaCl: strains with stronger amiloride-induced suppression of the neural response to NaCl had stronger NaCl avoidance [166, 182, 183, 186]. However, this relationship was not found in all studies. For example, NZB/BlNJ and CBA/J mice dramatically differ in NaCl consumption [107, 168] but have chorda tympani responses to NaCl of similar magnitude and sensitivity to amiloride [181]. Lack of correspondence between amiloride sensitivity of the CT response to NaCl and NaCl consumption was also found when three strains of rats, Fisher 344, Wistar and Sprague-Dawley, were compared [185, 186]. Furthermore, amiloride was found to affect NaCl perception in mice regardless of amiloride sensitivity of their gustatory neural responses to NaCl [32, 188]. More studies are needed to determine whether there is a mechanistic relationship between amiloride sensitivity of neural responses to NaCl and NaCl preference. It appears that the genetic variation in voluntary NaCl consumption can depend on a variety of factors, including, but not limited to, peripheral taste responsiveness.

Rats typically prefer NaCl solutions of intermediate (osmotically hypotonic or isotonic) concentrations. However, rats from the Fisher 344 strain do not display NaCl preferences [170]. A series of subsequent studies have presented a detailed analysis of mechanisms underlying this lack of NaCl preference in Fisher 344 rats. Relative to rats with typical responses to NaCl, Fisher 344 rats develop attenuated salt appetite in response to sodium deficiency or to interference with renin-angiotensin-aldosterone system, but they have similar sodium loss during sodium depletion. This suggests that differences in hedonic perception of NaCl rather than differences in sodium metabolism underlie aberrant NaCl consumption by the Fisher 344 rats [189–191]. Consistent with this, Fisher 344 rats show decreased appetitive and increased aversive oral motor responses to NaCl [192], and their NaCl aversion is abolished by CT, but not IX nerve transection [193, 194]. However, NaCl detection thresholds were not altered in Fisher 344 rats [195]. Thus, it appears that NaCl aversion in Fisher 344 rats is mediated by aversive gustatory information conveyed by the CT nerve.

Increased NaCl consumption by SHR (spontaneously hypertensive) rats does not depend on strain differences in peripheral gustatory input [184, 196], but appears to be mediated by the brain renin-angiotensin system [197, 198].

Taste and other complex phenotypes

Taste perception involves hedonic processes and is an important factor affecting ingestive behavior. Therefore, genetic variation in taste preferences is likely to affect complex phenotypes that depend on oral consumption of nutrients or drugs, or involve pleasure-seeking behavior. There are several examples of association of taste and complex traits in humans, including relationship between sweet taste and obesity [34, 41, 199, 200] or alcohol intake [201–205], and between bitter taste and alcohol intake [206–212], smoking [213, 214], food choice [215] and other health-related traits [216]. Studies of rodents elucidated some genetic factors and physiological mechanisms for association between sweet taste and alcohol, and have potential for unveiling mechanisms of the other associations between taste and complex phenotypes.

The oral consumption of alcohol is accompanied by chemosensory perception of its flavor, which plays an important role in its acceptance and rejection. Three independent sensory systems – taste, olfaction and chemosensory irritation – are involved in the perception of alcohol flavor. Humans perceive alcohol as a combination of sweet and bitter tastes, odors and oral irritation (e.g., burning sensation), all of which vary as a function of concentration [209, 217, 218]. Likewise, rodents detect the sweet (sucrose-like) and bitter (quinine-like) taste [219–221], odor volatiles [222, 223] of alcohol, and probably the other components detected by humans [224, 225].

Perception of the sweet taste component of ethanol by rodents was shown in behavioral and neurophysiological experiments. CTAs generalize between ethanol and sucrose [219, 221, 223, 226, 227]. Electrophysiological recordings indicate that lingual application of ethanol activates sweet-best neural fibers in the gustatory nerves [228, 229] and sweet-best units in the nucleus of the tractus solitarius [230, 231]; this activity is blocked by application of gurmarin, a peripheral antagonist of sweet taste [231]. Central mechanisms that determine hedonic responses to ethanol and sweeteners also overlap and involve opioidergic, serotonergic and dopaminergic brain neurotransmitter systems [232–236]. In addition, there may be common signals related to the caloric value of ethanol and sugars [237–242].

Positive correlations between preferences for ethanol and sweeteners in rats and mice were found among various strains and in segregating crosses [25, 45, 52, 66, 72, 73, 243–246], reviewed in [62, 63]. This genetically determined association can be underlaid by any of the mechanisms described above, including peripheral or central taste processing, or postingestive reward.

Genetic analysis of a cross between mice from a high ethanol- and sweetener-preferring B6 strain and a low ethanol- and sweetener-preferring 129 strain suggested that the strain differences in sweetener and ethanol consumption depend on relatively small and partially overlapping sets of genes [73]. One of these genetic loci, Ap3q (alcohol preference 3 QTL), maps to the subtelomeric region of Chr 4 [93] overlapping with the saccharin preference (Sac) locus that corresponds to the sweet taste receptor gene, Tas1r3 [77]. This suggests that the Tas1r3 gene is identical to the Ap3q locus and that its pleiotropic effect on ethanol consumption is mediated by genetic differences in perception of the sweet taste component of ethanol flavor: higher hedonic attractiveness of ethanol sweetness results in higher ethanol intake by B6 mice. The role of the T1R3 receptor in alcohol consumption was confirmed in a recent study showing that mutant mice lacking the Tas1r3 gene have diminished ethanol intakes and preferences [247]. In addition to the Tas1r3 gene, there are other genetic loci with pleiotropic effects on ethanol and sweetener intake [248, 249].

Conclusion

Behavioral genetic studies of taste responsiveness have been instrumental in the discovery of G-protein-coupled taste receptors. Genetic mapping of a human PTC/PROP taste sensitivity locus and mouse bitter and sweet taste loci have facilitated identification of the T1R and T2R families of taste receptors. The behavioral genetic approach has potential for more discoveries of taste mechanisms. There is strong evidence that multiple genes control behavioral taste responses. These yet unknown genes are likely to be involved in different stages of the taste processing pathway, including taste reception, transduction and transmission in the periphery and in the brain, and interaction of taste processing with homeostatic systems involved in the regulation of feeding, appetite, fluid balance, and reward. Genetics has experienced dramatic progress in recent years, with genome sequencing completed for several species, including mouse and human. These advances in genomic resources tremendously facilitate chromosomal mapping of genes affecting taste responsiveness and their identification. This turns behavioral genetics into a powerful approach for understanding mechanisms of taste.

References

  1. Bachmanov AA, Beauchamp GK: Taste Receptor Genes. Annu Rev Nutr. 2007, 27: 389-414. 10.1146/annurev.nutr.26.061505.111329.

    PubMed Central  CAS  PubMed  Google Scholar 

  2. Drayna D: Human taste genetics. Annu Rev Genomics Hum Genet. 2005, 6: 217-235. 10.1146/annurev.genom.6.080604.162340.

    CAS  PubMed  Google Scholar 

  3. Mombaerts P: Genes and ligands for odorant, vomeronasal and taste receptors. Nat Rev Neurosci. 2004, 5: 263-278. 10.1038/nrn1365.

    CAS  PubMed  Google Scholar 

  4. Scott K: Taste recognition: food for thought. Neuron. 2005, 48: 455-464. 10.1016/j.neuron.2005.10.015.

    CAS  PubMed  Google Scholar 

  5. Mueller KL, Hoon MA, Erlenbach I, Chandrashekar J, Zuker CS, Ryba NJ: The receptors and coding logic for bitter taste. Nature. 2005, 434: 225-229. 10.1038/nature03352.

    CAS  PubMed  Google Scholar 

  6. Kim UK, Drayna D: Genetics of individual differences in bitter taste perception: lessons from the PTC gene. Clin Genet. 2005, 67: 275-280. 10.1111/j.1399-0004.2004.00361.x.

    CAS  PubMed  Google Scholar 

  7. Maruyama Y, Pereira E, Margolskee RF, Chaudhari N, Roper SD: Umami responses in mouse taste cells indicate more than one receptor. J Neurosci. 2006, 26: 2227-2234. 10.1523/JNEUROSCI.4329-05.2006.

    PubMed Central  CAS  PubMed  Google Scholar 

  8. Li X, Li W, Wang H, Cao J, Maehashi K, Huang L, Bachmanov AA, Reed DR, Legrand-Defretin V, Beauchamp GK, Brand JG: Pseudogenization of a sweet-receptor gene accounts for cats' indifference toward sugar. PLoS Genet. 2005, 1: 27-35. 10.1371/journal.pgen.0010027.

    CAS  PubMed  Google Scholar 

  9. Scott TR: The role of taste in feeding. Appetite. 2001, 37: 111-113. 10.1006/appe.2001.0413.

    CAS  PubMed  Google Scholar 

  10. Cho YK, Li CS, Smith DV: Descending influences from the lateral hypothalamus and amygdala converge onto medullary taste neurons. Chem Senses. 2003, 28: 155-171. 10.1093/chemse/28.2.155.

    CAS  PubMed  Google Scholar 

  11. Hajnal A, Takenouchi K, Norgren R: Effect of intraduodenal lipid on parabrachial gustatory coding in awake rats. J Neurosci. 1999, 19: 7182-7190.

    CAS  PubMed  Google Scholar 

  12. Atchley DP, Weaver KL, Eckel LA: Taste responses to dilute sucrose solutions are modulated by stage of the estrous cycle and fenfluramine treatment in female rats. Physiol Behav. 2005, 86: 265-271. 10.1016/j.physbeh.2005.08.001.

    CAS  PubMed  Google Scholar 

  13. Curtis KS, Stratford JM, Contreras RJ: Estrogen increases the taste threshold for sucrose in rats. Physiol Behav. 2005, 86: 281-286. 10.1016/j.physbeh.2005.08.002.

    CAS  PubMed  Google Scholar 

  14. Simon SA, Liu L, Erickson RP: Neuropeptides modulate rat chorda tympani responses. Am J Physiol Regul Integr Comp Physiol. 2003, 284: R1494-1505.

    CAS  PubMed  Google Scholar 

  15. Shigemura N, Ohta R, Kusakabe Y, Miura H, Hino A, Koyano K, Nakashima K, Ninomiya Y: Leptin modulates behavioral responses to sweet substances by influencing peripheral taste structures. Endocrinology. 2004, 145: 839-847. 10.1210/en.2003-0602.

    CAS  PubMed  Google Scholar 

  16. Mook DG: Oral and postingestional determinants of the intake of various solutions in rats with esophageal fistulas. Journal of Comparative and Physiological Psychology. 1963, 56: 645-659. 10.1037/h0044824.

    Google Scholar 

  17. Marella S, Fischler W, Kong P, Asgarian S, Rueckert E, Scott K: Imaging taste responses in the fly brain reveals a functional map of taste category and behavior. Neuron. 2006, 49: 285-295. 10.1016/j.neuron.2005.11.037.

    CAS  PubMed  Google Scholar 

  18. Scott K, Brady R, Cravchik A, Morozov P, Rzhetsky A, Zuker C, Axel R: A chemosensory gene family encoding candidate gustatory and olfactory receptors in Drosophila. Cell. 2001, 104: 661-673. 10.1016/S0092-8674(01)00263-X.

    CAS  PubMed  Google Scholar 

  19. Huang AL, Chen X, Hoon MA, Chandrashekar J, Guo W, Trankner D, Ryba NJ, Zuker CS: The cells and logic for mammalian sour taste detection. Nature. 2006, 442: 934-938. 10.1038/nature05084.

    PubMed Central  CAS  PubMed  Google Scholar 

  20. Ishimaru Y, Inada H, Kubota M, Zhuang H, Tominaga M, Matsunami H: Transient receptor potential family members PKD1L3 and PKD2L1 form a candidate sour taste receptor. Proc Natl Acad Sci U S A. 2006, 103: 12569-12574. 10.1073/pnas.0602702103.

    PubMed Central  CAS  PubMed  Google Scholar 

  21. Harris H, Kalmus H: The measurement of taste sensitvity to phenylthiourea (P.T.C.). Annals of Eugenics. 1949, 15: 24-31.

    CAS  PubMed  Google Scholar 

  22. Bartoshuk LM, Duffy VB, Miller IJ: PTC/PROP tasting: anatomy, psychophysics, and sex effects. Physiol Behav. 1994, 56: 1165-1171. 10.1016/0031-9384(94)90361-1.

    CAS  PubMed  Google Scholar 

  23. Hansen JL, Reed DR, Wright MJ, Martin NG, Breslin PA: Heritability and Genetic Covariation of Sensitivity to PROP, SOA, Quinine HCl, and Caffeine. Chem Senses. 2006, 31: 403-413. 10.1093/chemse/bjj044.

    PubMed Central  CAS  PubMed  Google Scholar 

  24. Spector AC: Psychophysical evaluation of taste function in nonhuman mammals. Handbook of Olfaction and Gustation. Edited by: Doty RL. 2003, New York: Marcel Dekker, 869-879.

    Google Scholar 

  25. McClearn GE, Rodgers DA: Differences in alcohol preference among inbred strains of mice. Quart J Stud Alcohol. 1959, 20: 691-695.

    Google Scholar 

  26. Nachman M: The inheritance of saccharin preference. J Comp Physiol Psychol. 1959, 52: 451-457. 10.1037/h0048853.

    CAS  PubMed  Google Scholar 

  27. Hoshishima K, Yokoyama S, Seto K: Taste sensitivity in various strains of mice. Am J Physiol. 1962, 202: 1200-1204.

    CAS  PubMed  Google Scholar 

  28. Lush IE: The genetics of bitterness, sweetness, and saltiness in strains of mice. Genetics of Perception and Communication. Edited by: Wysocki CJ, Kare MR. 1991, New York: Marcel Dekker, 3: 227-241. Chemical Senses,

    Google Scholar 

  29. Glendinning JI, Gresack J, Spector AC: A high-throughput screening procedure for identifying mice with aberrant taste and oromotor function. Chem Senses. 2002, 27: 461-474. 10.1093/chemse/27.5.461.

    PubMed  Google Scholar 

  30. Nelson TM, Munger SD, Boughter JD: Haplotypes at the Tas2r locus on distal chromosome 6 vary with quinine taste sensitivity in inbred mice. BMC Genet. 2005, 6: 32-10.1186/1471-2156-6-32.

    PubMed Central  PubMed  Google Scholar 

  31. Eylam S, Spector AC: Stimulus processing of glycine is dissociable from that of sucrose and glucose based on behaviorally measured taste signal detection in Sac 'taster' and 'non-taster' mice. Chem Senses. 2004, 29: 639-649. 10.1093/chemse/bjh068.

    CAS  PubMed  Google Scholar 

  32. Eylam S, Spector AC: Taste discrimination between NaCl and KCl is disrupted by amiloride in inbred mice with amiloride-insensitive chorda tympani nerves. Am J Physiol Regul Integr Comp Physiol. 2005, 288: R1361-1368.

    CAS  PubMed  Google Scholar 

  33. Gent JF, Bartoshuk LM: Sweetness of sucrose, neohesperidin dihydrochalcone, and saccharin is related to genetic ability to taste the bitter substance 6-n-propylthiouracil. Chemical Senses. 1983, 7: 265-272. 10.1093/chemse/7.3-4.265.

    CAS  Google Scholar 

  34. Reed DR, Bachmanov AA, Beauchamp GK, Tordoff MG, Price RA: Heritable variation in food preferences and their contribution to obesity. Behavior Genetics. 1997, 27: 373-387. 10.1023/A:1025692031673.

    PubMed Central  CAS  PubMed  Google Scholar 

  35. Looy H, Weingarten HP: Facial expressions and genetic sensitivity to 6-n-propylthiouracil predict hedonic response to sweet. Physiol Behav. 1992, 52: 75-82. 10.1016/0031-9384(92)90435-5.

    CAS  PubMed  Google Scholar 

  36. Looy H, Callaghan S, Weingarten HP: Hedonic response of sucrose likers and dislikers to other gustatory stimuli. Physiol Behav. 1992, 52: 219-225. 10.1016/0031-9384(92)90261-Y.

    CAS  PubMed  Google Scholar 

  37. Beauchamp GK, Moran M: Dietary experience and sweet taste preference in human infants. Appetite. 1982, 3: 139-152.

    CAS  PubMed  Google Scholar 

  38. Desor JA, Greene LS, Maller O: Preferences for sweet and salty in 9- to 15-year-old and adult humans. Science. 1975, 190: 686-687. 10.1126/science.1188365.

    CAS  PubMed  Google Scholar 

  39. Greene LS, Desor JA, Maller O: Heredity and experience: their relative importance in the development of taste preference in man. J Comp Physiol Psychol. 1975, 89: 279-284. 10.1037/h0076802.

    CAS  PubMed  Google Scholar 

  40. Falciglia GA, Norton PA: Evidence for a genetic influence on preference for some foods. J Am Diet Assoc. 1994, 94: 154-158. 10.1016/0002-8223(94)90239-9.

    CAS  PubMed  Google Scholar 

  41. McDaniel AH, Reed DR: The human sweet tooth and its relationship to obesity. Genomics and Proteomics in Nutrition. Edited by: Berdanier CD, Moustaid-Moussa N. 2003, New York: Marcel Dekker, Inc, 51-70.

    Google Scholar 

  42. Reed DR, Li X, Bachmanov AA, Mascioli K, Beauchamp GK: The molecular basis of the mammalian sweet tooth. Progress in Obesity Research. 2003, Ltd. JLE. London, 9: 304-306. G Medeiros-Neto AH, and C. Bouchard (Series Editor)

    Google Scholar 

  43. Lewis SR, Ahmed S, Dym C, Khaimova E, Kest B, Bodnar RJ: Inbred mouse strain survey of sucrose intake. Physiol Behav. 2005, 85: 546-556. 10.1016/j.physbeh.2005.06.003.

    CAS  PubMed  Google Scholar 

  44. Fuller JL: Single-locus control of saccharin preference in mice. J Hered. 1974, 65: 33-36.

    CAS  PubMed  Google Scholar 

  45. Rodgers DA, McClearn GE: Sucrose versus ethanol appetite in inbred strains of mice. Q J Stud Alcohol. 1964, 25: 26-35.

    CAS  PubMed  Google Scholar 

  46. Lush IE: The genetics of tasting in mice. VI. Saccharin, acesulfame, dulcin and sucrose. Genetical Research. 1989, 53: 95-99.

    CAS  PubMed  Google Scholar 

  47. Ramirez I, Fuller JL: Genetic influence on water and sweetened water consumption in mice. Physiol Behav. 1976, 16: 163-168. 10.1016/0031-9384(76)90300-0.

    CAS  PubMed  Google Scholar 

  48. Pelz WE, Whitney G, Smith JC: Genetic influences on saccharin preference of mice. Physiol Behav. 1973, 10: 263-265. 10.1016/0031-9384(73)90308-9.

    CAS  PubMed  Google Scholar 

  49. Stockton MD, Whitney G: Effects of genotype, sugar, and concentration on sugar preference of laboratory mice (Mus musculus). J Comp Physiol Psychol. 1974, 86: 62-68. 10.1037/h0035929.

    CAS  PubMed  Google Scholar 

  50. Lush IE, Hornigold N, King P, Stoye JP: The genetics of tasting in mice. VII. Glycine revisited, and the chromosomal location of Sac and Soa. Genetical Research. 1995, 66: 167-174.

    CAS  PubMed  Google Scholar 

  51. Capeless CG, Whitney G: The genetic basis of preference for sweet substances among inbred strains of mice: Preference ratio phenotypes and the alleles of the Sac and dpa loci. Chemical Senses. 1995, 20: 291-298. 10.1093/chemse/20.3.291.

    CAS  PubMed  Google Scholar 

  52. Belknap JK, Crabbe JC, Young ER: Voluntary consumption of alcohol in 15 inbred mouse strains. Psychopharmacology. 1993, 112: 503-510. 10.1007/BF02244901.

    CAS  PubMed  Google Scholar 

  53. Reed DR, Li S, Li X, Huang L, Tordoff MG, Starling-Roney R, Taniguchi K, West DB, Ohmen JD, Beauchamp GK, Bachmanov AA: Polymorphisms in the taste receptor gene (Tas1r3) region are associated with saccharin preference in 30 mouse strains. J Neurosci. 2004, 24: 938-946. 10.1523/JNEUROSCI.1374-03.2004.

    PubMed Central  CAS  PubMed  Google Scholar 

  54. Kotlus BS, Blizard DA: Measuring gustatory variation in mice: A short-term fluid-intake test. Physiol Behav. 1998, 64: 37-47. 10.1016/S0031-9384(98)00016-X.

    CAS  PubMed  Google Scholar 

  55. Dotson CD, Spector AC: The relative affective potency of glycine, L-serine and sucrose as assessed by a brief-access taste test in inbred strains of mice. Chem Senses. 2004, 29: 489-498. 10.1093/chemse/bjh051.

    CAS  PubMed  Google Scholar 

  56. Glendinning JI, Chyou S, Lin I, Onishi M, Patel P, Zheng KH: Initial licking responses of mice to sweeteners: effects of tas1r3 polymorphisms. Chem Senses. 2005, 30: 601-614. 10.1093/chemse/bji054.

    CAS  PubMed  Google Scholar 

  57. Ninomiya Y, Higashi T, Katsukawa H, Mizukoshi T, Funakoshi M: Qualitative discrimination of gustatory stimuli in three different strains of mice. Brain Research. 1984, 322: 83-92. 10.1016/0006-8993(84)91183-1.

    CAS  PubMed  Google Scholar 

  58. Inoue M, McCaughey SA, Bachmanov AA, Beauchamp GK: Whole-nerve chorda tympani responses to sweeteners in C57BL/6ByJ and 129P3/J mice. Chemical Senses. 2001, 26: 915-923. 10.1093/chemse/26.7.915.

    PubMed Central  CAS  PubMed  Google Scholar 

  59. Frank ME, Blizard DA: Chorda tympani responses in two inbred strains of mice with different taste preferences. Physiol Behav. 1999, 67: 287-297. 10.1016/S0031-9384(99)00071-2.

    CAS  PubMed  Google Scholar 

  60. Ninomiya Y, Mizukoshi T, Higashi T, Katsukawa H, Funakoshi M: Gustatory neural responses in three different strains of mice. Brain Research. 1984, 302: 305-314. 10.1016/0006-8993(84)90244-0.

    CAS  PubMed  Google Scholar 

  61. Dess NK, Minor TR: Taste and emotionality in rats selectively bred for high versus low saccharin intake. Animal Learning and Behavior. 1996, 24: 105-115.

    Google Scholar 

  62. Overstreet DH, Rezvani AH, Parsian A: Behavioural features of alcohol-preferring rats: focus on inbred strains. Alcohol Alcohol. 1999, 34: 378-385.

    CAS  PubMed  Google Scholar 

  63. Kampov-Polevoy AB, Garbutt JC, Janowsky DS: Association between preference for sweets and excessive alcohol intake: a review of animal and human studies. Alcohol Alcohol. 1999, 34: 386-395.

    CAS  PubMed  Google Scholar 

  64. Frank ME, Wada Y, Makino J, Mizutani M, Umezawa H, Katsuie Y, Hettinger TP, Blizard DA: Variation in intake of sweet and bitter solutions by inbred strains of golden hamsters. Behav Genet. 2004, 34: 465-476. 10.1023/B:BEGE.0000023651.99481.d5.

    PubMed  Google Scholar 

  65. Bachmanov AA, Tordoff MG, Beauchamp GK: Sweetener preference of C57BL/6ByJ and 129P3/J mice. Chemical Senses. 2001, 26: 905-913. 10.1093/chemse/26.7.905.

    PubMed Central  CAS  PubMed  Google Scholar 

  66. Bachmanov AA, Tordoff MG, Beauchamp GK: Ethanol consumption and taste preferences in C57BL/6ByJ and 129/J mice. Alcohol Clin Exp Res. 1996, 20: 201-206. 10.1111/j.1530-0277.1996.tb01630.x.

    PubMed Central  CAS  PubMed  Google Scholar 

  67. Bachmanov AA, Reed DR, Tordoff MG, Price RA, Beauchamp GK: Nutrient preference and diet-induced adiposity in C57BL/6ByJ and 129P3/J mice. Physiol Behav. 2001, 72: 603-613. 10.1016/S0031-9384(01)00412-7.

    PubMed Central  CAS  PubMed  Google Scholar 

  68. Manita S, Bachmanov AA, Li X, Beauchamp GK, Inoue M: Is glycine "sweet" to mice? Mouse strain differences in perception of glycine taste. Chem Senses. 2006, 31: 785-793. 10.1093/chemse/bjl020.

    PubMed Central  CAS  PubMed  Google Scholar 

  69. Inoue M, Reed DR, Li X, Tordoff MG, Beauchamp GK, Bachmanov AA: Allelic variation of the Tas1r3 taste receptor gene selectively affects behavioral and neural taste responses to sweeteners in the F2 hybrids between C57BL/6ByJ and 129P3/J mice. J Neurosci. 2004, 24: 2296-2303. 10.1523/JNEUROSCI.4439-03.2004.

    PubMed Central  CAS  PubMed  Google Scholar 

  70. Fuller JL: Single-locus control of saccharin preference in mice. Journal of Heredity. 1974, 65: 33-36.

    CAS  PubMed  Google Scholar 

  71. Belknap JK, Crabbe JC, Plomin R, McClearn GE, Sampson KE, O'Toole LA, Gora-Maslak G: Single-locus control of saccharin intake in BXD/Ty recombinant inbred (RI) mice: Some methodological implications for RI strain analysis. Behavior Genetics. 1992, 22: 81-100. 10.1007/BF01066794.

    CAS  PubMed  Google Scholar 

  72. Phillips TJ, Crabbe JC, Metten P, Belknap JK: Localization of genes affecting alcohol drinking in mice. Alcohol Clin Exp Res. 1994, 18: 931-941. 10.1111/j.1530-0277.1994.tb00062.x.

    CAS  PubMed  Google Scholar 

  73. Bachmanov AA, Reed DR, Tordoff MG, Price RA, Beauchamp GK: Intake of ethanol, sodium chloride, sucrose, citric acid, and quinine hydrochloride solutions by mice: a genetic analysis. Behavior Genetics. 1996, 26: 563-573. 10.1007/BF02361229.

    PubMed Central  CAS  PubMed  Google Scholar 

  74. Blizard DA, Kotlus B, Frank ME: Quantitative trait loci associated with short-term intake of sucrose, saccharin and quinine solutions in laboratory mice. Chemical Senses. 1999, 24: 373-385. 10.1093/chemse/24.4.373.

    CAS  PubMed  Google Scholar 

  75. Bachmanov AA, Reed DR, Ninomiya Y, Inoue M, Tordoff MG, Price RA, Beauchamp GK: Sucrose consumption in mice: major influence of two genetic loci affecting peripheral sensory responses. Mammalian Genome. 1997, 8: 545-548. 10.1007/s003359900500.

    PubMed Central  CAS  PubMed  Google Scholar 

  76. Li X, Inoue M, Reed DR, Huque T, Puchalski RB, Tordoff MG, Ninomiya Y, Beauchamp GK, Bachmanov AA: High-resolution genetic mapping of the saccharin preference locus (Sac) and the putative sweet taste receptor (T1R1) gene (Gpr70) to mouse distal Chromosome 4. Mammalian Genome. 2001, 12: 13-16. 10.1007/s003350010236.

    PubMed Central  CAS  PubMed  Google Scholar 

  77. Bachmanov AA, Li X, Reed DR, Ohmen JD, Li S, Chen Z, Tordoff MG, de Jong PJ, Wu C, West DB, et al: Positional cloning of the mouse saccharin preference (Sac) locus. Chemical Senses. 2001, 26: 925-933. 10.1093/chemse/26.7.925.

    PubMed Central  CAS  PubMed  Google Scholar 

  78. Li X, Bachmanov AA, Li S, Chen Z, Tordoff MG, Beauchamp GK, de Jong PJ, Wu C, Chen L, West DB, et al: Genetic, physical and comparative map of the subtelomeric region of mouse chromosome 4. Mammalian Genome. 2002, 13: 5-19.

    PubMed Central  CAS  PubMed  Google Scholar 

  79. Wong GT, Gannon KS, Margolskee RF: Transduction of bitter and sweet taste by gustducin. Nature. 1996, 381: 796-800. 10.1038/381796a0.

    CAS  PubMed  Google Scholar 

  80. Montmayeur JP, Liberles SD, Matsunami H, Buck LB: A candidate taste receptor gene near a sweet taste locus. Nat Neurosci. 2001, 4: 492-498.

    CAS  PubMed  Google Scholar 

  81. Kitagawa M, Kusakabe Y, Miura H, Ninomiya Y, Hino A: Molecular genetic identification of a candidate receptor gene for sweet taste. Biochem Biophys Res Commun. 2001, 283: 236-242. 10.1006/bbrc.2001.4760.

    CAS  PubMed  Google Scholar 

  82. Hoon MA, Adler E, Lindemeier J, Battey JF, Ryba NJ, Zuker CS: Putative mammalian taste receptors: a class of taste-specific GPCRs with distinct topographic selectivity. Cell. 1999, 96: 541-551. 10.1016/S0092-8674(00)80658-3.

    CAS  PubMed  Google Scholar 

  83. Max M, Shanker YG, Huang L, Rong M, Liu Z, Campagne F, Weinstein H, Damak S, Margolskee RF: Tas1r3, encoding a new candidate taste receptor, is allelic to the sweet responsiveness locus Sac. Nat Genet. 2001, 28: 58-63. 10.1038/88270.

    CAS  PubMed  Google Scholar 

  84. Sainz E, Korley JN, Battey JF, Sullivan SL: Identification of a novel member of the T1R family of putative taste receptors. J Neurochem. 2001, 77: 896-903. 10.1046/j.1471-4159.2001.00292.x.

    CAS  PubMed  Google Scholar 

  85. Nelson G, Hoon MA, Chandrashekar J, Zhang Y, Ryba NJ, Zuker CS: Mammalian sweet taste receptors. Cell. 2001, 106: 381-390. 10.1016/S0092-8674(01)00451-2.

    CAS  PubMed  Google Scholar 

  86. Li X, Staszewski L, Xu H, Durick K, Zoller M, Adler E: Human receptors for sweet and umami taste. Proc Natl Acad Sci U S A. 2002, 99: 4692-4696. 10.1073/pnas.072090199.

    PubMed Central  CAS  PubMed  Google Scholar 

  87. Nelson G, Chandrashekar J, Hoon MA, Feng L, Zhao G, Ryba NJ, Zuker CS: An amino-acid taste receptor. Nature. 2002, 416: 199-202. 10.1038/nature726.

    CAS  PubMed  Google Scholar 

  88. Zhao GQ, Zhang Y, Hoon MA, Chandrashekar J, Erlenbach I, Ryba NJ, Zuker CS: The receptors for mammalian sweet and umami taste. Cell. 2003, 115: 255-266. 10.1016/S0092-8674(03)00844-4.

    CAS  PubMed  Google Scholar 

  89. Liao J, Schultz PG: Three sweet receptor genes are clustered in human chromosome 1. Mamm Genome. 2003, 14: 291-301. 10.1007/s00335-002-2233-0.

    CAS  PubMed  Google Scholar 

  90. Damak S, Rong M, Yasumatsu K, Kokrashvili Z, Varadarajan V, Zou S, Jiang P, Ninomiya Y, Margolskee RF: Detection of sweet and umami taste in the absence of taste receptor T1r3. Science. 2003, 301: 850-853. 10.1126/science.1087155.

    CAS  PubMed  Google Scholar 

  91. Nie Y, Vigues S, Hobbs JR, Conn GL, Munger SD: Distinct contributions of T1R2 and T1R3 taste receptor subunits to the detection of sweet stimuli. Curr Biol. 2005, 15: 1948-1952. 10.1016/j.cub.2005.09.037.

    CAS  PubMed  Google Scholar 

  92. Li X, Staszewski L, Xu H, Durick K, Zoller M, Adler E: Human receptors for sweet and umami taste. Proc Natl Acad Sci U S A. 2002, 99: 4692-4696. 10.1073/pnas.072090199.

    PubMed Central  CAS  PubMed  Google Scholar 

  93. Bachmanov AA, Reed DR, Li X, Li S, Beauchamp GK, Tordoff MG: Voluntary ethanol consumption by mice: genome-wide analysis of quantitative trait loci and their interactions in a C57BL/6ByJ × 129P3/J F2 intercross. Genome Res. 2002, 12: 1257-1268. 10.1101/gr.129702.

    PubMed Central  CAS  PubMed  Google Scholar 

  94. Lu K, McDaniel AH, Tordoff MG, Li X, Beauchamp GK, Bachmanov AA, VanderWeele DA, Chapman CD, Dess NK, Huang L, et al: No relationship between sequence variation in protein coding regions of the Tas1r3 gene and saccharin preference in rats. Chem Senses. 2005, 30: 231-240. 10.1093/chemse/bji019.

    PubMed Central  CAS  PubMed  Google Scholar 

  95. Foroud T, Bice P, Castelluccio P, Bo R, Ritchotte A, Stewart R, Lumeng L, Li TK, Carr L: Mapping of QTL influencing saccharin consumption in the selectively bred alcohol-preferring and -nonpreferring rat lines. Behav Genet. 2002, 32: 57-67. 10.1023/A:1014459912935.

    CAS  PubMed  Google Scholar 

  96. Xu H, Staszewski L, Tang H, Adler E, Zoller M, Li X: Different functional roles of T1R subunits in the heteromeric taste receptors. Proc Natl Acad Sci U S A. 2004, 101: 14258-14263. 10.1073/pnas.0404384101.

    PubMed Central  CAS  PubMed  Google Scholar 

  97. Beauchamp GK, Maller O, Rogers JG: Flavor preferences in cats (Felis catus and Panthera sp.). Journal of Comparative and Physiological Psychology. 1977, 91: 1118-1127. 10.1037/h0077380.

    CAS  Google Scholar 

  98. Bartoshuk LM, Jacobs HL, Nichols TL, Hoff LA, Ryckman JJ: Taste rejection of nonnutritive sweeteners in cats. J Comp Physiol Psychol. 1975, 89: 971-975. 10.1037/h0077172.

    CAS  PubMed  Google Scholar 

  99. Ninomiya Y, Sako N, Katsukawa H, Funakoshi M: Taste receptor mechanisms influenced by a gene on chromosome 4 in mice. Genetics of Perception and Communication. Edited by: Wysocki CJ, Kare MR. 1991, New York: Marcel Dekker, 3: 267-278. Chemical Senses,

    Google Scholar 

  100. Ninomiya Y, Higashi T, Mizukoshi T, Funakoshi M: Genetics of the ability to perceive sweetness of d-phenylalanine in mice. Annals of the New York Academy of Sciences. 1987, 510: 527-529. 10.1111/j.1749-6632.1987.tb43613.x.

    Google Scholar 

  101. Ninomiya Y, Nomura T, Katsukawa H: Genetically variable taste sensitivity to D-amino acids in mice. Brain Res. 1992, 596: 349-352. 10.1016/0006-8993(92)91571-U.

    CAS  PubMed  Google Scholar 

  102. Shigemura N, Yasumatsu K, Yoshida R, Sako N, Katsukawa H, Nakashima K, Imoto T, Ninomiya Y: The Role of the dpa Locus in Mice. Chem Senses. 2005, 30 (Suppl 1): i84-i85. 10.1093/chemse/bjh125.

    CAS  PubMed  Google Scholar 

  103. Chaudhari N, Landin AM, Roper SD: A metabotropic glutamate receptor variant functions as a taste receptor. Nat Neurosci. 2000, 3: 113-119. 10.1038/72053.

    CAS  PubMed  Google Scholar 

  104. Lugaz O, Pillias AM, Faurion A: A new specific ageusia: some humans cannot taste L-glutamate. Chem Senses. 2002, 27: 105-115. 10.1093/chemse/27.2.105.

    CAS  PubMed  Google Scholar 

  105. Ninomiya Y, Kurenuma S, Nomura T, Uebayashi H, Kawamura H: Taste synergism between monosodium glutamate and 5'-ribonucleotide in mice. Comp Biochem Physiol A. 1992, 101: 97-102. 10.1016/0300-9629(92)90634-3.

    CAS  PubMed  Google Scholar 

  106. Bachmanov AA, Tordoff MG, Beauchamp GK: Intake of umami-tasting solutions by mice: a genetic analysis. Journal of Nutrition. 2000, 130: 935S-941S.

    PubMed Central  CAS  PubMed  Google Scholar 

  107. Bachmanov AA, Tordoff MG, Beauchamp GK: Voluntary sodium chloride consumption by mice: Differences among five inbred strains. Behavior Genetics. 1998, 28: 117-124. 10.1023/A:1021471924143.

    PubMed Central  CAS  PubMed  Google Scholar 

  108. Inoue M, Beauchamp GK, Bachmanov AA: Gustatory Neural Responses to Umami Taste Stimuli in C57BL/6ByJ and 129P3/J Mice. Chem Senses. 2004, 29: 789-795. 10.1093/chemse/bjh083.

    PubMed Central  CAS  PubMed  Google Scholar 

  109. Spielman AI, Huque T, Whitney G, Brand JG: The diversity of bitter taste signal transduction mechanisms. Sensory Transduction. Edited by: Corey DP, Roper SD. 1992, New York: Rockefeller Univ. Press, 307-324.

    Google Scholar 

  110. Glendinning JI: Is the bitter rejection response always adaptive?. Physiol Behav. 1994, 56: 1217-1227. 10.1016/0031-9384(94)90369-7.

    CAS  PubMed  Google Scholar 

  111. Adler E, Hoon MA, Mueller KL, Chandrashekar J, Ryba NJ, Zuker CS: A novel family of mammalian taste receptors. Cell. 2000, 100: 693-702. 10.1016/S0092-8674(00)80705-9.

    CAS  PubMed  Google Scholar 

  112. Chandrashekar J, Mueller KL, Hoon MA, Adler E, Feng L, Guo W, Zuker CS, Ryba NJ: T2Rs function as bitter taste receptors. Cell. 2000, 100: 703-711. 10.1016/S0092-8674(00)80706-0.

    CAS  PubMed  Google Scholar 

  113. Matsunami H, Montmayeur JP, Buck LB: A family of candidate taste receptors in human and mouse. Nature. 2000, 404: 601-604. 10.1038/35007072.

    CAS  PubMed  Google Scholar 

  114. Shi P, Zhang J, Yang H, Zhang YP: Adaptive diversification of bitter taste receptor genes in Mammalian evolution. Mol Biol Evol. 2003, 20: 805-814. 10.1093/molbev/msg083.

    CAS  PubMed  Google Scholar 

  115. Fox AL: Six in ten "tasteblind" to bitter chemicals. Sci News Lett. 1931, 9: 249-

    Google Scholar 

  116. Fox AL: The relationship between chemical constituion and taste. Proc Natl Acad Sci U S A. 1932, 18: 115-120. 10.1073/pnas.18.1.115.

    PubMed Central  CAS  PubMed  Google Scholar 

  117. Harris H, Kalmus H: The measurement of taste sensitivity to phenylthiourea (P.T.C.). Annals of Eugenics. 1949, 15: 24-31.

    CAS  PubMed  Google Scholar 

  118. Snyder LH: Inherited taste deficiency. Science. 1931, 74: 151-152. 10.1126/science.74.1910.151.

    CAS  PubMed  Google Scholar 

  119. Blakeslee AF: Genetics of sensory thresholds: Taste for phenyl thio carbamide. Proc Natl Acad Sci U S A. 1932, 18: 120-130. 10.1073/pnas.18.1.120.

    PubMed Central  CAS  PubMed  Google Scholar 

  120. Olson JM, Boehnke M, Neiswanger K, Roche AF, Siervogel RM: Alternative genetic models for the inheritance of the phenylthiocarbamide taste deficiency. Genet Epidemiol. 1989, 6: 423-434. 10.1002/gepi.1370060305.

    CAS  PubMed  Google Scholar 

  121. Reddy BM, Rao DC: Phenylthiocarbamide taste sensitivity revisited: complete sorting test supports residual family resemblance. Genet Epidemiol. 1989, 6: 413-421. 10.1002/gepi.1370060304.

    CAS  PubMed  Google Scholar 

  122. Conneally PM, Dumont-Driscoll M, Huntzinger RS, Nance WE, Jackson CE: Linkage relations of the loci for Kell and phenylthiocarbamide taste sensitivity. Hum Hered. 1976, 26: 267-271.

    CAS  PubMed  Google Scholar 

  123. Reed DR, Nanthakumar E, North M, Bell C, Bartoshuk LM, Price RA: Localization of a gene for bitter-taste perception to human chromosome 5p15. Am J Hum Genet. 1999, 64: 1478-1480. 10.1086/302367.

    PubMed Central  CAS  PubMed  Google Scholar 

  124. Drayna D, Coon H, Kim UK, Elsner T, Cromer K, Otterud B, Baird L, Peiffer AP, Leppert M: Genetic analysis of a complex trait in the Utah Genetic Reference Project: a major locus for PTC taste ability on chromosome 7q and a secondary locus on chromosome 16p. Hum Genet. 2003, 112: 567-572.

    CAS  PubMed  Google Scholar 

  125. Kim UK, Jorgenson E, Coon H, Leppert M, Risch N, Drayna D: Positional cloning of the human quantitative trait locus underlying taste sensitivity to phenylthiocarbamide. Science. 2003, 299: 1221-1225. 10.1126/science.1080190.

    CAS  PubMed  Google Scholar 

  126. Richter TA, Clisby KH: Phenylthiocarbamide taste thresholds of rats and human beings. Am J Physiol. 1941, 134: 157-164.

    CAS  Google Scholar 

  127. Klein TW, DeFries JC: Similar polymorphism of taste sensitivity to PTC in mice and men. Nature. 1970, 225: 555-557. 10.1038/225555a0.

    CAS  PubMed  Google Scholar 

  128. Whitney G, Harder DB: PTC preference among laboratory mice: understanding of a previously "unreplicated" report. Behav Genet. 1986, 16: 605-610. 10.1007/BF01066287.

    CAS  PubMed  Google Scholar 

  129. Smith JC, Davis JD, O-Keefe GB: Lack of an order effect in brief contact taste tests with closely spaced test trials. Physiol Behav. 1992, 52: 1107-1111. 10.1016/0031-9384(92)90467-G.

    CAS  PubMed  Google Scholar 

  130. Spector AC: Linking gustatory neurobiology to behavior in vertebrates. Neurosci Biobehav Rev. 2000, 24: 391-416. 10.1016/S0149-7634(00)00013-0.

    CAS  PubMed  Google Scholar 

  131. Smith BK, Volaufova J, West DB: Increased flavor preference and lick activity for sucrose and corn oil in SWR/J vs. AKR/J mice. Am J Physiol Regul Integr Comp Physiol. 2001, 281: R596-606.

    CAS  PubMed  Google Scholar 

  132. Boughter JD, St John SJ, Noel DT, Ndubuizu O, Smith DV: A brief-access test for bitter taste in mice. Chem Senses. 2002, 27: 133-142. 10.1093/chemse/27.2.133.

    PubMed  Google Scholar 

  133. Nelson TM, Munger SD, Boughter JD: Taste sensitivities to PROP and PTC vary independently in mice. Chem Senses. 2003, 28: 695-704. 10.1093/chemse/bjg062.

    CAS  PubMed  Google Scholar 

  134. St John SJ, Pour L, Boughter JD: Phenylthiocarbamide produces conditioned taste aversions in mice. Chem Senses. 2005, 30: 377-382. 10.1093/chemse/bji032.

    PubMed  Google Scholar 

  135. Whitney G, Harder DB: Genetics of bitter perception in mice. Physiol Behav. 1994, 56: 1141-1147. 10.1016/0031-9384(94)90358-1.

    CAS  PubMed  Google Scholar 

  136. Lush IE, Hornigold N, King P, Stoye JP: The genetics of tasting in mice. VII. Glycine revisited, and the chromosomal location of Sac and Soa. Genet Res. 1995, 66: 167-174.

    CAS  PubMed  Google Scholar 

  137. Warren RP, Lewis RC: Taste polymorphism in mice involving a bitter sugar derivate. Nature. 1970, 227: 77-78. 10.1038/227077a0.

    CAS  PubMed  Google Scholar 

  138. Lush IE: The genetics of tasting in mice. I. Sucrose octaacetate. Genetical Research. 1981, 38: 93-95.

    CAS  PubMed  Google Scholar 

  139. Lush IE: The genetics of tasting in mice. III. Quinine. Genet Res. 1984, 44: 151-160.

    CAS  PubMed  Google Scholar 

  140. Harder DB, Whitney G, Frye P, Smith JC, Rashotte ME: Strain differences among mice in taste psychophysics of sucrose octaacetate. Chemical Senses. 1984, 9: 311-323. 10.1093/chemse/9.4.311.

    CAS  Google Scholar 

  141. Whitney G, Harder DB: Single locus control of sucrose octaacetate tasting among mice. Behavior Genetics. 1986, 16: 559-574. 10.1007/BF01066342.

    CAS  PubMed  Google Scholar 

  142. Harder DB, Capeless CG, Maggio JC, Boughter JD, Gannon KS, Whitney G, Azen EA: Intermediate sucrose octa-acetate sensitivity suggests a third allele at mouse bitter taste locus Soa and Soa-Rua identity. Chemical Senses. 1992, 17: 391-401. 10.1093/chemse/17.4.391.

    CAS  Google Scholar 

  143. Inoue M, Li X, McCaughey SA, Beauchamp GK, Bachmanov AA: Soa genotype selectively affects mouse gustatory neural responses to sucrose octaacetate. Physiological Genomics. 2001, 5: 181-186.

    PubMed Central  CAS  PubMed  Google Scholar 

  144. Capeless CG, Whitney G, Azen EA: Chromosome mapping of Soa, a gene influencing gustatory sensitivity to sucrose octaacetate in mice. Behavior Genetics. 1992, 22: 655-663. 10.1007/BF01066636.

    CAS  PubMed  Google Scholar 

  145. Bachmanov AA, Li X, Li S, Neira M, Beauchamp GK, Azen EA: High-resolution genetic mapping of the sucrose octaacetate taste aversion (Soa) locus on mouse Chromosome 6. Mamm Genome. 2001, 12: 695-699. 10.1007/s00335-001-2061-7.

    PubMed Central  CAS  PubMed  Google Scholar 

  146. Le Roy I, Pager J, Roubertoux PL: Genetic dissection of gustatory sensitivity to bitterness (sucrose octaacetate) in mice. C R Acad Sci III. 1999, 322: 831-836.

    CAS  PubMed  Google Scholar 

  147. Harder DB, Maggio JC, Whitney G: Assessing gustatory detection capabilities using preference procedures. Chemical Senses. 1989, 14: 547-564. 10.1093/chemse/14.4.547.

    CAS  Google Scholar 

  148. Boughter JD, Harder DB, Capeless CG, Whitney G: Polygenic determination of quinine sensitivity among mice. Chem Senses. 1992, 17: 427-434. 10.1093/chemse/17.4.427.

    CAS  Google Scholar 

  149. Bachmanov AA, Reed DR, Tordoff MG, Price RA, Beauchamp GK: Intake of ethanol, sodium chloride, sucrose, citric acid, and quinine hydrochloride solutions by mice: a genetic analysis. Behav Genet. 1996, 26: 563-573. 10.1007/BF02361229.

    PubMed Central  CAS  PubMed  Google Scholar 

  150. Harder DB, Whitney G: A common polygenic basis for quinine and PROP avoidance in mice. Chem Senses. 1998, 23: 327-332.

    CAS  PubMed  Google Scholar 

  151. Lush IE, Holland G: The genetics of tasting in mice. V. Glycine and cycloheximide. Genet Res. 1988, 52: 207-212.

    CAS  PubMed  Google Scholar 

  152. Blizard DA, Kotlus B, Frank ME: Quantitative trait loci associated with short-term intake of sucrose, saccharin and quinine solutions in laboratory mice. Chem Senses. 1999, 24: 373-385. 10.1093/chemse/24.4.373.

    CAS  PubMed  Google Scholar 

  153. Lush IE: The genetics of tasting in mice. IV. The acetates of raffinose, galactose and β-lactose. Genetical Research. 1986, 47: 117-123.

    CAS  PubMed  Google Scholar 

  154. Chandrashekar J, Mueller KL, Hoon MA, Adler E, Feng L, Guo W, Zuker CS, Ryba JP: T2Rs function as bitter taste receptors. Cell. 2000, 100: 703-711. 10.1016/S0092-8674(00)80706-0.

    CAS  PubMed  Google Scholar 

  155. Conte C, Guarin E, Marcuz A, Andres-Barquin PJ: Functional expression of mammalian bitter taste receptors in Caenorhabditis elegans. Biochimie. 2006

    Google Scholar 

  156. Go Y: Lineage-specific expansions and contractions of the bitter taste receptor gene repertoire in vertebrates. Mol Biol Evol. 2006, 23: 964-972. 10.1093/molbev/msj106.

    CAS  PubMed  Google Scholar 

  157. Conte C, Ebeling M, Marcuz A, Nef P, Andres-Barquin PJ: Evolutionary relationships of the Tas2r receptor gene families in mouse and human. Physiol Genomics. 2003, 14: 73-82.

    CAS  PubMed  Google Scholar 

  158. Wang X, Thomas SD, Zhang J: Relaxation of selective constraint and loss of function in the evolution of human bitter taste receptor genes. Hum Mol Genet. 2004, 13: 2671-2678. 10.1093/hmg/ddh289.

    CAS  PubMed  Google Scholar 

  159. Go Y, Satta Y, Takenaka O, Takahata N: Lineage-specific loss of function of bitter taste receptor genes in humans and nonhuman primates. Genetics. 2005, 170: 313-326. 10.1534/genetics.104.037523.

    PubMed Central  CAS  PubMed  Google Scholar 

  160. Soranzo N, Bufe B, Sabeti PC, Wilson JF, Weale ME, Marguerie R, Meyerhof W, Goldstein DB: Positive selection on a high-sensitivity allele of the human bitter-taste receptor TAS2R16. Curr Biol. 2005, 15: 1257-1265. 10.1016/j.cub.2005.06.042.

    CAS  PubMed  Google Scholar 

  161. Gilbertson TA, Boughter JD: Taste transduction: appetizing times in gustation. NeuroReport. 2003, 14: 905-911. 10.1097/00001756-200305230-00001.

    CAS  PubMed  Google Scholar 

  162. Fitzsimons JT: The Physiology of Thirst and Sodium Appetite. 1979, Cambridge: Cambridge University Press

    Google Scholar 

  163. Denton D: The hunger for salt: An anthropological, physiological and medical analysis. 1984, Berlin: Springer-Verlag

    Google Scholar 

  164. Austin MA, King MC, Bawol RD, Hulley SB, Friedman GD: Risk factors for coronary heart disease in adult female twins. Genetic heritability and shared environmental influences. Am J Epidemiol. 1987, 125: 308-318.

    CAS  PubMed  Google Scholar 

  165. Hoshishima K, Yokoyama S, Seto K: Taste sensitivity in various strains of mice. American Journal of Physiology. 1962, 202: 1200-1204.

    CAS  PubMed  Google Scholar 

  166. Ninomiya Y, Sako N, Funakoshi M: Strain differences in amiloride inhibition of NaCl responses in mice, Mus musculus. J Comp Physiol [A]. 1989, 166: 1-5.

    CAS  Google Scholar 

  167. Bachmanov AA, Schlager G, Tordoff MG, Beauchamp GK: Consumption of electrolytes and quinine by mouse strains with different blood pressures. Physiol Behav. 1998, 64: 323-330. 10.1016/S0031-9384(98)00069-9.

    PubMed Central  CAS  PubMed  Google Scholar 

  168. Bachmanov AA, Beauchamp GK, Tordoff MG: Voluntary consumption of NaCl, KCl, CaCl2 and NH4Cl solutions by 28 mouse strains. Behavior Genetics. 2002, 32: 445-457. 10.1023/A:1020832327983.

    PubMed Central  PubMed  Google Scholar 

  169. Beauchamp GK, Fisher AS: Strain differences in consumption of saline solutions by mice. Physiol Behav. 1993, 54: 179-184. 10.1016/0031-9384(93)90063-L.

    CAS  PubMed  Google Scholar 

  170. Midkiff EE, Fitts DA, Simpson JB, Bernstein IL: Absence of sodium chloride preference in Fischer-344 rats. American Journal of Physiology. 1985, 249: R438-R442.

    CAS  PubMed  Google Scholar 

  171. Midkiff EE, Bernstein IL, Fitts DA, Simpson JB: Salt preference and salt appetite of Fischer-344 rats. Chem Senses. 1985, 10: 446-

    Google Scholar 

  172. Bachmanov AA: The salt appetite of rats with spontaneous arterial hypertension. Fiziol Zh SSSR (in Russian). 1989, 75: 942-947.

    CAS  Google Scholar 

  173. Tordoff MG: Influence of dietary calcium on sodium and calcium intake of spontaneously hypertensive rats. American Journal of Physiology. 1992, 262: R370-R381.

    CAS  PubMed  Google Scholar 

  174. DiNicolantonio R, Mendelsohn FAO, Hutchinson JS: Sodium chloride preference of genetically hypertensive and normotensive rats. American Journal of Physiology. 1983, 245: R38-R44.

    CAS  Google Scholar 

  175. Sollars SI, Midkiff EE, Bernstein IL: Genetic transmission of NaCl aversion in the Fisher-344 rat. Chemical Senses. 1990, 15: 521-527. 10.1093/chemse/15.5.521.

    CAS  Google Scholar 

  176. Bachmanov AA, Dmitriev YS: Genetic analysis of some behavioral and physiological traits in hybrids between hypertensive and normotensive rats: Analysis of the inheritance nature (In Russian; English translation: Soviet Genetics, 28: 796–804). Genetika. 1992, 28: 150-159.

    CAS  PubMed  Google Scholar 

  177. Bachmanov AA, Dmitriev YS: Genetic analysis of some behavioral and physiological traits in hybrids between hypertensive and normotensive rats: Analysis of reciprocal differences (In Russian; English translation: Soviet Genetics, 28: 921–926). Genetika. 1992, 28: 120-127.

    CAS  PubMed  Google Scholar 

  178. Bachmanov AA, Dmitriev YS: Genetic analysis of some behavioral and physiological traits in hybrids between hypertensive and normotensive rats: Analysis of correlational associations (In Russian; English translation: Soviet Genetics, 28: 927–936). Genetika. 1992, 28: 128-138.

    CAS  PubMed  Google Scholar 

  179. Harrap SB: Genetic analysis of blood pressure and sodium balance in spontaneously hypertensive rats. Hypertension. 1986, 8: 572-582.

    CAS  PubMed  Google Scholar 

  180. Di Nicolantonio R, Kren V, Zidek V, Pravenec M: Salt preference of congenic strains derived from the spontaneously hypertensive rat. Physiol Behav. 2004, 80: 617-622. 10.1016/j.physbeh.2003.11.001.

    CAS  PubMed  Google Scholar 

  181. Ninomiya Y, Bachmanov AA, Yatabe A, Beauchamp GK: NaCl-preferring NZB/B1NJ mice and NaCl-avoiding CBA/J mice have similar amiloride inhibition of chorda tympani responses to NaCl. Chemical Senses. 1998, 23: 411-415.

    PubMed Central  CAS  PubMed  Google Scholar 

  182. Ninomiya Y, Fukami Y, Yamazaki K, Beauchamp GK: Amiloride inhibition of chorda tympani responses to NaCl and its temperature dependency in mice. Brain Research. 1996, 708: 153-158. 10.1016/0006-8993(95)01218-4.

    CAS  PubMed  Google Scholar 

  183. Gannon KS, Contreras RJ: Sodium intake linked to amiloride-sensitive gustatory transduction in C57BL/6J and 129/J mice. Physiol Behav. 1995, 57: 231-239. 10.1016/0031-9384(94)00279-E.

    CAS  PubMed  Google Scholar 

  184. Formaker BK, Hill DL: Lack of amiloride sensitivity in SHR and WKY glossopharyngeal taste responses to NaCl. Physiol Behav. 1991, 50: 765-769. 10.1016/0031-9384(91)90015-G.

    CAS  PubMed  Google Scholar 

  185. Minear MM, Hammack SE, Lundy RF, Contreras RJ: Amiloride inhibits taste nerve responses to NaCl and KCl in Sprague-Dawley and Fischer 344 rats. Physiol Behav. 1996, 60: 507-516. 10.1016/S0031-9384(96)80026-6.

    CAS  PubMed  Google Scholar 

  186. Bernstein IL, Longley A, Taylor EM: Amiloride sensitivity of chorda tympani response to NaCl in Fischer 344 and Wistar rats. Am J Physiol. 1991, 261: R329-R333.

    CAS  PubMed  Google Scholar 

  187. Brand J, Teeter J, Silver W: Inhibition by amiloride of chorda tympani responses evoked by monovalent salts. Brain Research. 1985, 334: 207-214. 10.1016/0006-8993(85)90212-4.

    CAS  PubMed  Google Scholar 

  188. Eylam S, Spector AC: Oral amiloride treatment decreases taste sensitivity to sodium salts in C57BL/6J and DBA/2J mice. Chem Senses. 2003, 28: 447-458. 10.1093/chemse/28.5.447.

    CAS  PubMed  Google Scholar 

  189. Midkiff EE, Fitts DA, Simpson JB, Bernstein IL: Attenuated sodium appetite in response to sodium deficiency in Fischer-344 rats. American Journal of Physiology. 1987, 252: R562-R566.

    CAS  PubMed  Google Scholar 

  190. Rowland NE, Fregly MJ: Induction of an appetite for sodium in rats that show no spontaneous preference for sodium chloride solution – the Fischer 344 strain. Behavioral Neuroscience. 1988, 102: 961-968. 10.1037/0735-7044.102.6.961.

    CAS  PubMed  Google Scholar 

  191. Caputo FA, Rowland NE, Fregly MJ: Angiotensin-related intakes of water and NaCl in Fischer 344 and Sprague-Dawley rats. Am J Physiol. 1992, 262: R382-388.

    CAS  PubMed  Google Scholar 

  192. Grill HJ, Bernstein IL: Strain differences in taste reactivity to NaCl. American Journal of Physiology. 1988, 255: R424-R430.

    CAS  PubMed  Google Scholar 

  193. Sollars SI, Sollars PJ, Bernstein IL: Reversal of the sodium chloride aversion of Fischer 344 rats by chorda tympani nerve transection. Behav Neurosci. 1991, 105: 603-605. 10.1037/0735-7044.105.4.603.

    CAS  PubMed  Google Scholar 

  194. Sollars SI, Bernstein IL: Gustatory deafferentation and desalivation: effects on NaCl preference of Fischer 344 rats. Am J Physiol. 1994, 266: R510-517.

    CAS  PubMed  Google Scholar 

  195. Clarke SN, Koh MT, Bernstein IL: NaCl detection thresholds: comparison of Fischer 344 and Wistar rats. Chem Senses. 2001, 26: 253-257. 10.1093/chemse/26.3.253.

    CAS  PubMed  Google Scholar 

  196. Formaker BK, Hill DL: Peripheral taste responses in genetically hypertensive rats. Physiol Behav. 1990, 47: 1229-1237. 10.1016/0031-9384(90)90376-F.

    CAS  PubMed  Google Scholar 

  197. DiNicolantonio R, Hutchinson JS, Mendelsohn FA: Exaggerated salt appetite of spontaneously hypertensive rats is decreased by central angiotensin-converting enzyme blockade. Nature. 1982, 298: 846-848. 10.1038/298846a0.

    CAS  PubMed  Google Scholar 

  198. Bachmanov AA, Whittlestone TH, Fitzsimons JT: The enhanced sodium appetite of spontaneously hypertensive (SH) rats depends on angiotensin (Abstract). Journal of Physiology (London). 1994, 480: 85P-

    Google Scholar 

  199. Drewnowski AJ, Brunzell JD, Sande K, Iverius PH, Greenwood MR: Sweet tooth reconsidered: taste responsiveness in human obesity. Physiol Behav. 1985, 35: 617-622. 10.1016/0031-9384(85)90150-7.

    CAS  PubMed  Google Scholar 

  200. Salbe AD, DelParigi A, Pratley RE, Drewnowski A, Tataranni PA: Taste preferences and body weight changes in an obesity-prone population. Am J Clin Nutr. 2004, 79: 372-378.

    CAS  PubMed  Google Scholar 

  201. Yamamoto ME, Block GD, Ishii E: Food pattens among adolescents: influence of alcohol consumption. Alcohol Clin Exp Res. 1991, 15: 359-

    Google Scholar 

  202. Kampov-Polevoy AB, Garbutt JC, Khalitov E: Family history of alcoholism and response to sweets. Alcohol Clin Exp Res. 2003, 27: 1743-1749. 10.1097/01.ALC.0000093739.05809.DD.

    PubMed  Google Scholar 

  203. Kampov-Polevoy AB, Eick C, Boland G, Khalitov E, Crews FT: Sweet liking, novelty seeking, and gender predict alcoholic status. Alcohol Clin Exp Res. 2004, 28: 1291-1298. 10.1097/01.ALC.0000137808.69482.75.

    PubMed  Google Scholar 

  204. Kampov-Polevoy AB, Garbutt JC, Davis CE, Janowsky DS: Preference for higher sugar concentrations and Tridimensional Personality Questionnaire scores in alcoholic and nonalcoholic men. Alcohol Clin Exp Res. 1998, 22: 610-614. 10.1111/j.1530-0277.1998.tb04300.x.

    CAS  PubMed  Google Scholar 

  205. Kampov-Polevoy AB, Garbutt JC, Janowsky D: Evidence of preference for a higher concentration sucrose solution in alcoholic men. American Journal of Psychiatry. 1997, 154: 269-270.

    CAS  PubMed  Google Scholar 

  206. Guinard J-X, Zoumas-Morse C, Dietz J, Goldberg S, Holz M, Heck E, Amoros A: Does consumption of beer, alcohol, and bitter substances affect bitterness perception. Physiol Behav. 1996, 59: 625-631. 10.1016/0031-9384(95)02124-8.

    CAS  PubMed  Google Scholar 

  207. Duffy VB, Davidson AC, Kidd JR, Kidd KK, Speed WC, Pakstis AJ, Reed DR, Snyder DJ, Bartoshuk LM: Bitter receptor gene (TAS2R38), 6-n-propylthiouracil (PROP) bitterness and alcohol intake. Alcohol Clin Exp Res. 2004, 28: 1629-1637. 10.1097/01.ALC.0000145789.55183.D4.

    PubMed Central  CAS  PubMed  Google Scholar 

  208. Intranuovo LR, Powers AS: The perceived bitterness of beer and 6-n-propylthiouracil (PROP) taste sensitivity. Ann N Y Acad Sci. 1998, 855: 813-815. 10.1111/j.1749-6632.1998.tb10665.x.

    CAS  PubMed  Google Scholar 

  209. Bartoshuk LM, Conner E, Grubin D, Karrer T, Kochenbach K, Palsco M, Snow D, Pelchat M, Danovski S: PROP supertasters and the perception of ethyl alcohol. Chemical Senses. 1993, 18: 526-527.

    Google Scholar 

  210. Lanier SA, Hayes JE, Duffy VB: Sweet and bitter tastes of alcoholic beverages mediate alcohol intake in of-age undergraduates. Physiol Behav. 2005, 83: 821-831. 10.1016/j.physbeh.2004.10.004.

    CAS  PubMed  Google Scholar 

  211. Pelchat ML, Danowski S: A possible genetic association between PROP-tasting and alcoholism. Physiol Behav. 1992, 51: 1261-1266. 10.1016/0031-9384(92)90318-V.

    CAS  PubMed  Google Scholar 

  212. DiCarlo ST, Powers AS: Propylthiouracil tasting as a possible genetic association marker for two types of alcoholism. Physiol Behav. 1998, 64: 147-152. 10.1016/S0031-9384(98)00043-2.

    CAS  PubMed  Google Scholar 

  213. Enoch MA, Harris CR, Goldman D: Does a reduced sensitivity to bitter taste increase the risk of becoming nicotine addicted?. Addict Behav. 2001, 26: 399-404. 10.1016/S0306-4603(00)00117-9.

    CAS  PubMed  Google Scholar 

  214. Cannon DS, Baker TB, Piper ME, Scholand MB, Lawrence DL, Drayna DT, McMahon WM, Villegas GM, Caton TC, Coon H, Leppert MF: Associations between phenylthiocarbamide gene polymorphisms and cigarette smoking. Nicotine Tob Res. 2005, 7: 853-858. 10.1080/14622200500330209.

    CAS  PubMed  Google Scholar 

  215. Dinehart ME, Hayes JE, Bartoshuk LM, Lanier SL, Duffy VB: Bitter taste markers explain variability in vegetable sweetness, bitterness, and intake. Physiol Behav. 2006, 87: 304-313. 10.1016/j.physbeh.2005.10.018.

    CAS  PubMed  Google Scholar 

  216. Basson MD, Bartoshuk LM, Dichello SZ, Panzini L, Weiffenbach JM, Duffy VB: Association between 6-n-propylthiouracil (PROP) bitterness and colonic neoplasms. Dig Dis Sci. 2005, 50: 483-489. 10.1007/s10620-005-2462-7.

    CAS  PubMed  Google Scholar 

  217. Green BG: The sensitivity of the tongue to ethanol. Annals of the New York Academy of Sciences. 1987, 510: 315-317. 10.1111/j.1749-6632.1987.tb43541.x.

    Google Scholar 

  218. Mattes RD, DiMeglio D: Ethanol perception and ingestion. Physiol Behav. 2001, 72: 217-229. 10.1016/S0031-9384(00)00397-8.

    CAS  PubMed  Google Scholar 

  219. Blizard DA: Sweet and bitter taste of ethanol in C57BL/6J and DBA2/J mouse strains. Behav Genet. 2007, 37: 146-159. 10.1007/s10519-006-9121-4.

    PubMed  Google Scholar 

  220. Kiefer SW, Lawrence GJ: The sweet-bitter taste of alcohol: aversion generalization to various sweet-quinine mixtures in the rat. Chemical Senses. 1988, 13: 633-641. 10.1093/chemse/13.4.633.

    CAS  Google Scholar 

  221. Kiefer SW, Mahadevan RS: The taste of alcohol for rats as revealed by aversion generalization tests. Chemical Senses. 1993, 18: 509-522. 10.1093/chemse/18.5.509.

    CAS  Google Scholar 

  222. Molina JC, Domínguez HD, López MF, Pepino MY, Faas AE: The role of fetal and infantile experience with alcohol in later recognition and acceptance patterns of the drug. Alcohol and Alcoholism: Brain and Development. Edited by: Spear NE, Spear LP, Hanningan JH, Goodlett CR. 1999, Hillsdale, N.J.: Lawrence Erlbaum Associates, Inc, 199-228.

    Google Scholar 

  223. Kiefer SW, Morrow NS: Odor cue mediation of alcohol aversion learning in rats lacking gustatory neocortex. Behav Neurosci. 1991, 105: 25-32. 10.1037/0735-7044.105.1.25.

    CAS  PubMed  Google Scholar 

  224. Nachman M, Larue C, Le Magnen J: The role of olfactory and orosensory factors in the alcohol preference of inbred strains of mice. Physiol Behav. 1971, 6: 53-95. 10.1016/0031-9384(71)90014-X.

    CAS  PubMed  Google Scholar 

  225. Belknap JK, Belknap ND, Berg JH, Coleman R: Preabsorbtive vs. postabsorbtive control of ethanol intake in C57BL/6J and DBA/2J mice. Behavior Genetics. 1977, 7: 413-425. 10.1007/BF01066776.

    CAS  PubMed  Google Scholar 

  226. Blizard DA, McClearn GE: Association between ethanol and sucrose intake in the laboratory mouse: exploration via congenic strains and conditioned taste aversion. Alcohol Clin Exp Res. 2000, 24: 253-258. 10.1111/j.1530-0277.2000.tb04605.x.

    CAS  PubMed  Google Scholar 

  227. Lawrence GJ, Kiefer SW: Generalization of specific taste aversions to alcohol in the rat. Chemical Senses. 1987, 12: 591-599. 10.1093/chemse/12.4.591.

    CAS  Google Scholar 

  228. Hellekant G, Danilova V, Roberts T, Ninomiya Y: The taste of ethanol in a primate model: I. Chorda tympani nerve response in Macaca mulatta. Alcohol. 1997, 14: 473-484. 10.1016/S0741-8329(96)00215-7.

    CAS  PubMed  Google Scholar 

  229. Sako N, Yamamoto T: Electrophysiological and behavioral studies on taste effectiveness of alcohols in rats. American Journal of Physiology. 1999, 276: R388-396.

    CAS  PubMed  Google Scholar 

  230. Di Lorenzo PM, Kiefer SW, Rice AG, Garcia J: Neural and behavioral responsivity to ethyl alcohol as a tastant. Alcohol. 1986, 3: 55-61. 10.1016/0741-8329(86)90071-6.

    CAS  PubMed  Google Scholar 

  231. Lemon CH, Brasser SM, Smith DV: Alcohol activates a sucrose-responsive gustatory neural pathway. J Neurophysiol. 2004, 92: 536-544. 10.1152/jn.00097.2004.

    CAS  PubMed  Google Scholar 

  232. Levine AS, Kotz CM, Gosnell BA: Sugars: hedonic aspects, neuroregulation, and energy balance. Am J Clin Nutr. 2003, 78: 834S-842S.

    CAS  PubMed  Google Scholar 

  233. Gosnell BA, Majchrzak MJ: Centrally administered opioid peptides stimulate saccharin intake in nondeprived rats. Pharmacol Biochem Behav. 1989, 33: 805-810. 10.1016/0091-3057(89)90474-7.

    CAS  PubMed  Google Scholar 

  234. George SR, Roldan L, Lui A, Naranjo CA: Endogenous opioids are involved in the genetically determined high preference for ethanol consumption. Alcohol Clin Exp Res. 1991, 15: 668-672. 10.1111/j.1530-0277.1991.tb00576.x.

    CAS  PubMed  Google Scholar 

  235. Hubell CL, Marglin SH, Spitalnic SJ, Abelson ML, Wild KD, Reid LD: Opioidergic, serotoninergic, and dopaminergic manipulations and rats' intake of a sweetened alcoholic beverage. Alcohol. 1991, 8: 355-367. 10.1016/0741-8329(91)90573-F.

    Google Scholar 

  236. Pucilowski O, Rezvani AH, Janowsky DS: Suppression of alcohol and saccharin preference in rats by a novel Ca2+ channel inhibitor, Goe 5438. Psychopharmacology. 1992, 107: 447-452. 10.1007/BF02245174.

    CAS  PubMed  Google Scholar 

  237. Gentry RT, Dole VP: Why does a sucrose choice reduce the consumption of alcohol in C57BL/6J mice?. Life Sciences. 1987, 40: 2191-2194. 10.1016/0024-3205(87)90010-5.

    CAS  PubMed  Google Scholar 

  238. McMillen BA, Williams HL: Role of taste and calories in the selection of ethanol by C57BL/6NHsd and Hsd:ICR mice. Alcohol. 1998, 15: 193-198. 10.1016/S0741-8329(97)00111-0.

    CAS  PubMed  Google Scholar 

  239. Rodgers DA, McClearn GE, Bennett EL, Hebert M: Alcohol preference as a function of its caloric utility in mice. J Comp Physiol Psychol. 1963, 56: 666-672. 10.1037/h0040350.

    CAS  PubMed  Google Scholar 

  240. Rodgers DA: Factors underlying differences in alcohol preference among inbred strains of mice. Psychosomatic Medicine. 1966, 28: 498-513.

    Google Scholar 

  241. Freed EX: Alcohol polydipsia in the rat as a function of caloric need. Quart J Stud Alc. 1972, 33: 504-507.

    CAS  Google Scholar 

  242. Richter CP: Alcohol as a food. Quarterly Journal of Studies on Alcohol. 1941, 1: 650-662.

    CAS  Google Scholar 

  243. Overstreet DH, Kampov-Polevoy AB, Rezvani AH, Murelle L, Halikas JA, Janowsky DS: Saccharin intake predicts ethanol intake in genetically heterogeneous rats as well as different rat strains. Alcohol Clin Exp Res. 1993, 17: 366-369. 10.1111/j.1530-0277.1993.tb00777.x.

    CAS  PubMed  Google Scholar 

  244. Stewart RB, Russell RN, Lumeng L, Li TK, Murphy JM: Consumption of sweet, salty, sour, and bitter solutions by selectively bred alcohol-preferring and alcohol-nonpreferring lines of rats. Alcohol Clin Exp Res. 1994, 18: 375-381. 10.1111/j.1530-0277.1994.tb00028.x.

    CAS  PubMed  Google Scholar 

  245. Dess NK, Badia-Elder NE, Thiele TE, Kiefer SW, Blizard DA: Ethanol consumption in rats selectively bred for differential saccharin intake. Alcohol. 1998, 16: 275-278. 10.1016/S0741-8329(98)00010-X.

    CAS  PubMed  Google Scholar 

  246. Stewart RB, Bice P, Foroud T, Lumeng L, Li TK, Carr LG: Correlation of saccharin and ethanol intake in the F2 progeny of HAD2 and LAD2 crosses. Alcohol Clin Exp Res. 2003, 27: 49A-10.1097/01.ALC.0000052704.14282.B2.

    Google Scholar 

  247. Blednov YA, Walker D, Martinez M, Levine M, Damak S, Margolskee RF: Perception of sweet taste is important for voluntary alcohol consumption in mice. Genes Brain Behav. 2007

    Google Scholar 

  248. Terenina-Rigaldie E, Moisan MP, Colas A, Beauge F, Shah KV, Jones BC, Mormede P: Genetics of behaviour: phenotypic and molecular study of rats derived from high- and low-alcohol consuming lines. Pharmacogenetics. 2003, 13: 543-554. 10.1097/00008571-200309000-00003.

    PubMed  Google Scholar 

  249. Terenina-Rigaldie E, Jones BC, Mormede P: Pleiotropic effect of a locus on chromosome 4 influencing alcohol drinking and emotional reactivity in rats. Genes Brain Behav. 2003, 2: 125-131. 10.1034/j.1601-183X.2003.00018.x.

    CAS  PubMed  Google Scholar 

  250. Bufe B, Hofmann T, Krautwurst D, Raguse JD, Meyerhof W: The human TAS2R16 receptor mediates bitter taste in response to beta-glucopyranosides. Nat Genet. 2002, 32: 397-401. 10.1038/ng1014.

    CAS  PubMed  Google Scholar 

  251. Kuhn C, Bufe B, Winnig M, Hofmann T, Frank O, Behrens M, Lewtschenko T, Slack JP, Ward CD, Meyerhof W: Bitter taste receptors for saccharin and acesulfame K. J Neurosci. 2004, 24: 10260-10265. 10.1523/JNEUROSCI.1225-04.2004.

    CAS  PubMed  Google Scholar 

  252. Behrens M, Brockhoff A, Kuhn C, Bufe B, Winnig M, Meyerhof W: The human taste receptor hTAS2R14 responds to a variety of different bitter compounds. Biochem Biophys Res Commun. 2004, 319: 479-485. 10.1016/j.bbrc.2004.05.019.

    CAS  PubMed  Google Scholar 

  253. Bachmanov AA, Reed DR, Li X, Beauchamp GK: Genetics of sweet taste preference. Pure and Applied Chemistry. 2002, 74: 1135-1140.

    CAS  Google Scholar 

Download references

Acknowledgements

Supported by NIH grants DC00882 (G.K. Beauchamp), AA11028 (AAB), DC000353 and NS052366 (JDB), and an Ajinomoto Amino Acid Research Program Focused Research grant (AAB).

This article has been published as part of BMC Neuroscience Volume 8 Supplement 3, 2007: The chemical senses: recent advances and new promises. The full contents of the supplement are available online at http://0-www-biomedcentral-com.brum.beds.ac.uk/1471-2202/8?issue=S3.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to John D Boughter Jr.

Additional information

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

JDB and AAB contributed equally to this review.

Rights and permissions

This article is published under license to BioMed Central Ltd. This is an open access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Reprints and permissions

About this article

Cite this article

Boughter, J.D., Bachmanov, A.A. Behavioral genetics and taste. BMC Neurosci 8 (Suppl 3), S3 (2007). https://0-doi-org.brum.beds.ac.uk/10.1186/1471-2202-8-S3-S3

Download citation

  • Published:

  • DOI: https://0-doi-org.brum.beds.ac.uk/10.1186/1471-2202-8-S3-S3

Keywords