Hostname: page-component-7c8c6479df-p566r Total loading time: 0 Render date: 2024-03-28T03:00:13.142Z Has data issue: false hasContentIssue false

Review: Chemosensing of nutrients and non-nutrients in the human and porcine gastrointestinal tract

Published online by Cambridge University Press:  07 August 2019

E. Roura*
Affiliation:
Centre for Nutrition and Food Sciences, Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, St Lucia, Queensland, Australia
I. Depoortere
Affiliation:
Translational Research Center for Gastrointestinal Disorders, Gut Peptide Research Lab, University of Leuven, Belgium
M. Navarro
Affiliation:
Centre for Nutrition and Food Sciences, Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, St Lucia, Queensland, Australia
*

Abstract

The gastrointestinal tract (GIT) is an interface between the external and internal milieus that requires continuous monitoring for nutrients or pathogens and toxic chemicals. The study of the physiological/molecular mechanisms, mediating the responses to the monitoring of the GIT contents, has been referred to as chemosensory science. While most of the progress in this area of research has been obtained in laboratory rodents and humans, significant steps forward have also been reported in pigs. The objective of this review was to update the current knowledge on nutrient chemosensing in pigs in light of recent advances in humans and laboratory rodents. A second objective relates to informing the existence of nutrient sensors with their functionality, particularly linked to the gut peptides relevant to the onset/offset of appetite. Several cell types of the intestinal epithelium such as Paneth, goblet, tuft and enteroendocrine cells (EECs) contain subsets of chemosensory receptors also found on the tongue as part of the taste system. In particular, EECs show specific co-expression patterns between nutrient sensors and/or transceptors (transport proteins with sensing functions) and anorexigenic hormones such as cholecystokinin (CCK), peptide tyrosine tyrosine (PYY) or glucagon-like peptide-1 (GLP-1), amongst others. In addition, the administration of bitter compounds has an inhibitory effect on GIT motility and on appetite through GLP-1-, CCK-, ghrelin- and PYY-labelled EECs in the human small intestine and colon. Furthermore, the mammalian chemosensory system is the target of some bacterial metabolites. Recent studies on the human microbiome have discovered that commensal bacteria have developed strategies to stimulate chemosensory receptors and trigger host cellular functions. Finally, the study of gene polymorphisms related to nutrient sensors explains differences in food choices, food intake and appetite between individuals.

Type
Review Article
Copyright
© The Animal Consortium 2019 

Implications

How the gastrointestinal tract senses the arrival of dietary nutrients and non-nutrients (e.g. toxins) has a tremendous impact on the hunger–satiety cycle. Nutrient sensing is mediated by the activation of taste receptors or other sensors/transporters present in the intestinal epithelium associated with the enteroendocrine system. For example, the excess of specific non-limiting dietary amino acids in pigs has the capacity to strongly trigger satiating signals through chemosensory mechanisms (Muller and Roura, unpublished). In the future, standard feed formulation guidelines in farm animals (including pigs) will have to include not only essential-limiting amino acids but the wider array of dietary amino acids as well. A full understanding of these mechanisms is essential to develop dietary strategies to optimize feed intake in farm animals such as the pig.

Introduction

Nutritional chemosensing is the scientific discipline studying how nutrients are perceived in biological systems including genomic, metabolic, physiological and behavioural mechanisms (Roura et al., Reference Roura, Koopmans, Lallès, Le Huerou-Luron, de Jager, Schuurman and Val-Laillet2016). The molecular mechanisms of oral nutrient and non-nutrient sensing involve a large repertoire of receptors including taste receptors (TRs). The activation of TRs trigger the depolarization of the sensory cell in the tongue and the stimulation of the gustatory cortex of the brain mediated by the signalling of the cranial nerves VII, IX and X (Barretto et al., Reference Barretto, Gillis-Smith, Chandrashekar, Yarmolinsky, Schnitzer, Ryba and Zuker2015). In addition, the mechanisms of nutrient perception discovered in the oral cavity have also been described outside the oral cavity as part of the enteroendocrine system (EES) mediating the hunger–satiety cycle (reviewed by Steensels and Depoortere, Reference Steensels and Depoortere2018). In the intestinal epithelium, there are several cell types, such as enterocytes, enteroendocrine cells (EECs), tuft, Paneth, goblet, microfold and cup cells, which play a key role reporting the luminal content to the brain (Depoortere, Reference Depoortere2014). These mechanisms were originally studied in humans and laboratory rodents; however, in recent reports homologous mechanisms have been uncovered in pigs (reviewed by Roura and Fu, Reference Roura and Fu2017; Roura and Foster, Reference Roura and Foster2018).

In addition, some nutrient transporters seem to play a dual role meaning that the main role of transporting might be coupled to nutrient-sensing signalling. These transporters have been referred to as ‘transceptors’ (Reimann et al., Reference Reimann, Habib, Tolhurst, Parker, Rogers and Gribble2008; Poncet and Taylor, Reference Poncet and Taylor2013). Sensory functions of nutrient transporters in and outside the oral cavity are increasingly being recognized in mammalian species (Diallinas, Reference Diallinas2017; Roura and Foster, Reference Roura and Foster2018; Steensels and Depoortere, Reference Steensels and Depoortere2018). The molecular mechanisms of oral nutrient sensing and transporters are complex and imply a high degree of specificity to each nutrient type. This review aims at summarizing the current knowledge on nutrient (carbohydrates, proteins and lipids) and non-nutrient (bitter or bacterial compounds) chemosensing and the mediation of appetite-regulating gut peptides in pigs, presented using the progress obtained in humans and laboratory rodents as a reference. Novel research avenues on ‘microbial and parasite sensing’ have been highlighted in the ‘Microbial and parasite sensing…’ section of the review.

Carbohydrate sensing

Carbohydrate sensing has been related to two taste-like types referred as sweet and starchy tastes in humans (Aji et al., Reference Aji, Warren and Roura2019). On the one hand, sweet taste has evolved around mono-, di- and tri-saccharides (simple sugars), not only in humans but also in other mammalian species including pigs (Sclafani, Reference Sclafani1987; Glaser et al., Reference Glaser, Wanner, Tinti and Nofre2000; Lapis et al., Reference Lapis, Penner and Lim2014; Low et al., Reference Low, Lacy, McBride and Keast2017; Roura and Fu, Reference Roura and Fu2017). On the other hand, starch is the primary carbohydrate source in pigs. In recent years, starch-related sweet taste has gained relevance as part of the dietary nutrient-sensing mechanisms in the oral cavity. Despite a short contact time with starch in the mouth, salivary alpha-amylase has the potential to elicit sweet taste in humans by releasing maltose and maltotriose (Aji et al., Reference Aji, Warren and Roura2019).

However, the sensing of complex carbohydrates is independent of sweet taste as described in laboratory rodents (Sclafani, Reference Sclafani1987). Lapis et al. (Reference Lapis, Penner and Lim2014) demonstrated the taste of glucose was correlated with sucrose but not with the sensing of complex carbohydrates. Pigs were also reported to sense complex carbohydrates from hydrolysed corn starch (Roura et al., Reference Roura, Shrestha and Diffey2013).

Carbohydrate sensors and transceptors in the gastrointestinal tract

Most mammalian species (except strict carnivores) have a very conserved mechanism of simple sugar perception (related to sweet taste in humans). Table 1 summarizes the main receptors and transporters known to be involved in sensing sugars in the gastrointestinal tract (GIT) in humans and pigs. Among other potential receptors, simple carbohydrates sensing involves a heterodimer of two G-protein-coupled receptors (GPCRs) known as taste receptor type 1 member 2 and member 3 (TAS1R2 and TAS1R3, respectively) (Bachmanov and Beauchamp, Reference Bachmanov and Beauchamp2007). In addition, a TAS1R2/TAS1R3-independent sensing of monosaccharides (e.g. glucose and fructose) has been recently described in the oral cavity. The system was related to the glucose transporters/co-transporters (GLUTs) and sodium-dependent glucose transporter type 1 and 2 (SGLT1/2), and the brush border enzymes present in the apical membrane of some taste sensory cells (Glendinning et al., Reference Glendinning, Stano, Holter, Azenkot, Goldman, Margolskee, Vasselli and Sclafani2015; Sukumaran et al. Reference Sukumaran, Yee, Iwata, Kotha, Quezada-Calvillo, Nichols, Mohan, Pinto, Shigemura and Ninomiya2016). An analogous system has been previously described in the small intestine (Cheng et al., Reference Cheng, Chegeni, Kim, Zhang, Benmoussa, Quezada-Calvillo, Nichols and Hamaker2014; Zhang et al., Reference Zhang, Hasek, Lee and Hamaker2015). In addition, the stimulation of the TAS1R sweet receptor dimer seemed to upregulate SGLT1 to facilitate glucose uptake in the intestine (Mace et al., Reference Mace, Affleck, Patel and Kellett2007; Margolskee et al., Reference Margolskee, Dyer, Kokrashvili, Salmon, Ilegems, Daly, Maillet, Ninomiya, Mosinger and Shirazi-Beechey2007). GLUT5 has also been reported to influence glucagon-like peptide-1 (GLP-1) release from enteroendocrine K-cells (Douard and Ferraris, Reference Douard and Ferraris2008). However, potential dual roles for other sugar transporters/sensors known to be expressed in the GIT (i.e. KATP channel, SGLT2, GLUT2 or GLUT5) have not been reported to date (Table 1). Some of these molecular mechanisms have also been described in pigs (Roura and Fu, Reference Roura and Fu2017). The identification of putative receptors responsible for the sensing of starch and glucose polymers remains elusive to date in mammalian species.

Table 1. Main simple carbohydrates receptors and transporters known to be involved in GIT sensing in humans and pigs1

GIT = gastrointestinal tract; GI = gastrointestinal; GIP = glucose insulinotropic peptide; GLUT = glucose transporter; PYY = peptide YY; SGLT = sodium–glucose cotransporter 1; T1R = taste receptor family 1; KATP channel = ATP-sensitive K+ channel; GLP-1 = glucagon-like peptide 1; Fru = fructose; Gluc = glucose; Gal = galactose; Man = mannose; Mal = maltose; Suc = sucrose; Glucos = glucosamine.

1 All the receptors and transporters presented in the table are relevant to humans and pigs except if noted with the superscript 3.

2 Table references are provided in Supplementary Material S1. Note: some references to laboratory rodent research have been used to illustrate the discovery or proof of the GIT-related function of some genes.

3 No literature evidence of the functionality of this gene has been found in pigs.

Carbohydrates sensors and the enteroendocrine system

The presence of simple sugars in the GIT activates the expression and stimulation of TAS1Rs in EECs which, in turn, release gut peptides relevant to the orchestration of the hunger–satiety cycle (Rozengurt et al., Reference Rozengurt2006). The main hormones involved in this response include cholecystokinin (CCK), peptide tyrosine tyrosine (PYY) and GLP-1 (Badman and Flier, Reference Badman and Flier2005). These hormones are known to regulate energy and glucose metabolism by modulating the homoeostatic and food reward systems in the brain implicated in hunger and satiety (Berridge and Robinson, Reference Berridge and Robinson1998). In particular, carbohydrate sensing mediated by TAS1R2/TAS1R3, SGLT1 and/or the KATP has been described on L-cells and K-cells known to secrete GLP-1 and glucose insulinotropic peptide (GIP), respectively (Steensels and Depoortere, Reference Steensels and Depoortere2018). The expression and co-localization of TAS1R2, TAS1R3 and transceptor SGLT1 in L-cells has been related to GLP-1 secretion in humans and rodents (Jang et al., Reference Jang, Kokrashvili, Theodorakis, Carlson, Kim, Zhou, Kim, Xu, Chan, Juhaszova, Bernier, Mosinger, Margolskee and Egan2007; Steinert et al., Reference Steinert, Gerspach, Gutmann, Asarian, Drewe and Beglinger2011a; Gerspach et al., Reference Gerspach, Steinert, Schonenberger, Graber-Maier and Beglinger2011). In addition, sugar sensors are found in human stomach, expressed in endocrine P/D1 cells (also referred to X/A cells in lab rodents) and inhibit the release of the hunger hormone ghrelin (Wang et al., Reference Wang, Liszt, Deloose, Canovai, Thijs, Farre, Ceulemans, Lannoo, Tack and Depoortere2019). However, the effect of glucose on GLP-1 and PYY release could be overruled or potentiated by other nutrients such as proteins or fats (Gerspach et al., Reference Gerspach, Steinert, Schonenberger, Graber-Maier and Beglinger2011). Interestingly, artificial sweeteners showed no effect on GLP-1 in vivo in rodents and humans, suggesting that they may not induce physiological effects in the GIT (Steinert et al., 2011b; Steensels et al., Reference Steensels, Cools, Avau, Vancleef, Farre, Verbeke and Depoortere2016).

In pigs, Moran et al. (Reference Moran, Al-Rammahi, Arora, Batchelor, Coulter, Daly, Ionescu, Bravo and Shirazi-Beechey2010a and Reference Moran, Al-Rammahi, Arora, Batchelor, Coulter, Ionescu, Bravo and Shirazi-Beechey2010b) found that dietary carbohydrates or saccharin enhanced SGLT1 expression in small intestine epithelial cells including L and K cells resulting in an increased glucose absorption. In addition, L and K cells co-expressed pTas1r2/pTas1r3, SGLT1 and GIP and GLP-1. Thus, SGLT1 was shown to be the main route of absorption of dietary sugars and that the increased expression of SGLT1 in epithelial cells was mediated by the stimulation of pTas1rs in pigs (Moran et al., Reference Moran, Al-Rammahi, Arora, Batchelor, Coulter, Ionescu, Bravo and Shirazi-Beechey2010b).

Gene polymorphisms in carbohydrate sensing

Based on population genomic analyses, 18 single-nucleotide polymorphisms (SNPs) (of which 10 were non-synonymous – ns – that is, causing a change in the amino acid (AA) sequence of the receptor) have been identified in TAS1R2 (Kim et al., Reference Kim, Wooding, Riaz, Jorde and Drayna2006). TAS1R2 variants have been associated with higher sucrose taste thresholds and dietary sugar intake (Eny et al., Reference Eny, Wolever, Corey and El-Sohemy2010) or to lower carbohydrate intake (Ramos-Lopez et al., Reference Ramos-Lopez, Panduro, Martinez-Lopez and Roman2016). In addition, Dias et al. (Reference Dias, Eny, Cockburn, Chiu, Nielsen, Duizer and El-Sohemy2015) found that the functional impact of another TAS1R2 polymorphism was body mass index (BMI) dependent – that is, high sucrose thresholds and sugar intake found in overweight individuals (BMI>25) but not in normal-weight individuals (BMI<25). Furthermore, low compared to high sweet taste sensitivity volunteers consumed a higher amount of energy from a buffet meal, implying a strong involvement of TAS1R2 allelic variants on food choices (Han et al., Reference Han, Keast and Roura2017). In the same study, low sweet sensitivity was related to high salivary leptin. Similarly, a high oral sensitivity to the taste of complex carbohydrates (maltodextrin and oligofructose) was associated with higher consumption of energy and starch and waist circumference (Low et al., Reference Low, Lacy, McBride and Keast2017). Regarding genetic polymorphisms in pigs, the studies conducted to date have not reported potential pTas1r2 variants because the gene was not annotated in the pig genome at the time the studies were conducted (Da Silva et al., Reference Da Silva, De Jager, Burgos, Reverter, Perez-Enciso and Roura2014; Clop et al., Reference Clop, Sharaf, Castello, Ramos-Onsins, Cirera, Mercade, Derdak, Huisman, Fredholm, van As and Sanchez2016).

Protein/amino acid sensing

Dietary protein, as a source of AA, plays a fundamental role in growth and development. Of the 20 proteinogenic AAs needed for protein synthesis in eukaryotic cells, a few cannot be metabolically synthesized ‘de novo’ from other carbon and nitrogen sources within the cells, and need to be consumed as part of the diet. Thus, optimal growth and development in pigs requires a balanced supply of these so-called dietary essential AAs; one of the key aspects in current pig feed formulation practices. Failure to supply a balanced diet in terms of essential AA results in deficient growth and development and ultimately death. Thus, it is not surprising that a wide array of AAs and peptide sensors exist in mammalian species. In humans, the oral sensing of dietary AA was originally related to glutamate (and aspartate) and defined as the umami taste (Ikeda, Reference Ikeda1909). Other AAs sensed include aromatic AA (e.g. L-Phe), basic AA (L-Arg) and dietary peptides (Zhang et al., Reference Zhang, Huang, Jiang, Mulpuri, Wei, Hamelberg, Brown and Yang2014). However, in other mammals such as laboratory rodents and pigs, the oral/umami sensing of AA involves several L-AAs (Tinti et al., Reference Tinti, Glaser, Wanner and Nofre2000; Roura et al., Reference Roura, Humphrey, Klasing and Swart2011).

Amino acid sensors and transceptors in the gastrointestinal tract

Table 2 shows the main receptors and transporters known to be involved in AA sensing in the GIT in humans and pigs. The umami taste receptor is a GPCR heterodimer: TAS1R1/TAS1R3 (Nelson et al., Reference Nelson, Chandrashekar, Hoon, Feng, Zhao, Ryba and Zuker2002). In addition, the metabotropic glutamate receptors (particularly mGluR1 and mGluR4) have also been related to glutamate sensing in humans, in and outside the oral cavity (San Gabriel and Uneyama, Reference San Gabriel and Uneyama2013). Other AA sensors have been identified including the calcium sensing receptor (CaSR, sensing basic AA and Ca2+ as a heterotrophic cooperative enhancer) and GPRC6A (sensing aromatic AA) (Zhang et al., Reference Zhang, Huang, Jiang, Mulpuri, Wei, Hamelberg, Brown and Yang2014; Steensels and Depoortere, Reference Steensels and Depoortere2018). CaSR acts in concert with GPRC6A and are found expressed in D-, G- and L-cells (Haid et al., Reference Haid, Jordan-Biegger, Widmayer and Breer2012). Finally, di- and tripeptides are sensed by GPR92/93. Similar to previous receptors, AA sensors are also widely expressed throughout the GIT in humans, lab rodents and pigs (Wellendorph et al., Reference Wellendorph, Johansen and Brauner-Osborne2010; Roura and Foster, Reference Roura and Foster2018) (Table 2).

Table 2. Main AA receptors and transceptors known to be involved in GIT sensing in humans and pigs1

AA = amino acid; GIT = gastrointestinal tract; GI = gastrointestinal; LPA = lysophoshatidic acid; CaSR = calcium-sensor receptor; CCK = cholecystokinin; L-AA = L isomer of amino acids; GPCRs = G-protein-coupled receptors; SCFA = short-chain fatty acid; GLP-1 = glucagon-like peptide 1; GIP = glucose-dependent insulinotropic peptide; mGluRs = metabotropic glutamate receptors; PepT1,2: peptide transporter 1 and 2; T1R = taste receptor family 1.

1 All the receptors and transporters presented in the table are relevant to humans and pigs.

2 Table references are provided in Supplementary Material S1. Note: some references to laboratory rodent research have been added to illustrate the discovery or proof of the GIT-related function of some genes.

3 Refers to L-AA stimulating the umami taste receptor dimer in humans (glutamic and aspartic acids) or pigs (Ala, Asn, Asp, Glu, Gln, Pro, Ser and Thr) (source: Roura et al., Reference Roura, Humphrey, Klasing and Swart2011).

There is a complex and highly specific network of AA and peptide intestinal transporters belonging to the solute carrier (SLC) family. A detailed description of these transporters can be found elsewhere (Broer, Reference Broer2008). However, the evidence of any of these transporters to function as AA sensors remains to be fully studied.

Amino acids and the enteroendocrine system

In the GIT, the stimulation of the umami heterodimer and the CaSR have been associated with the secretion of CCK, ghrelin and GLP-1 (Liou et al., Reference Liou, Sei, Zhao, Feng, Lu, Thomas, Raybould and Wank2011a; Diakogiannaki et al., Reference Diakogiannaki, Pais, Tolhurst, Parker, Horscroft, Horscroft, Rauscher, Zietek, Daniel, Gribble and Reinmann2013; Vancleef et al., Reference Vancleef, Van Den Broeck, Thijs, Steensels, Briand, Tack and Depoortere2015). In addition, GPR92/93 has been reported in stomach G-cells and STC-1 cells responding to a protein hydrolysate by releasing CCK (Choi et al., Reference Choi, Lee, Shiu, Yo, Halldén and Aponte2007; Rettenberger et al., Reference Rettenberger, Schulze, Breer and Haid2015).

Similar to the TAS1R-independent mechanisms of sweet taste perception, the AA sensing also seems to partially rely on AA transceptors as an alternative pathway to signal responses through EEC. Di/tripeptide uptake in L cells occurs via peptide transporter 1 (PEPT1) and results in subsequent basolateral activation of the CaSR and GLP-1 release (Diakogiannaki et al., Reference Diakogiannaki, Pais, Tolhurst, Parker, Horscroft, Horscroft, Rauscher, Zietek, Daniel, Gribble and Reinmann2013; Daniel and Zietek, Reference Daniel and Zietek2015; Modvig et al., Reference Modvig, Kuhre and Holst2019). Another potential example of AA transceptor is the sodium-dependent neutral AAs transporter 2 (SNAT2) involved in GLP-1 secretion (Reimann et al., Reference Reimann, Ward and Gribble2006; Young et al., Reference Young, Rey, Sternini and Rozengurt2010). A large number of additional AA transporters (e.g. the SLC family) are known to be expressed in the GIT but, as indicated previously, their potential role as transceptors has not been fully described (Broer, Reference Broer2008). In pigs, the first fully functional taste receptor gene to be sequenced, cloned and expressed in a cell reporter system was the umami heterodimer pTas1r1/pTas1r3 (Humphrey et al., Reference Humphrey, Tedó, Klasing and Roura2009; Tedo Perez, Reference Tedo Perez2009; Roura et al., Reference Roura, Humphrey, Klasing and Swart2011). The results indicated that the umami taste in pigs was tuned to 8 L-AA (Ala, Asn, Asp, Glu, Gln, Pro, Ser and Thr) (Roura et al., Reference Roura, Humphrey, Klasing and Swart2011). The expressions of the porcine metabotropic glutamate receptors (mGluR1 and mGluR4) and other AA and peptone receptors (i.e. CaSR, GPRC6A and GPR92) have also been reported more recently in pig tongue and stomach epithelia (Haid et al., Reference Haid, Jordan-Biegger, Widmayer and Breer2012; Da Silva et al., Reference Da Silva, De Jager, Burgos, Reverter, Perez-Enciso and Roura2014). In addition, the AA receptors involved in sensing protein breakdown products were identified in G-cells and D-cells in pigs (Haid et al., Reference Haid, Jordan-Biegger, Widmayer and Breer2012). Finally, several AA transporters of the SLC family have been identified in the pig GIT; however their potential role as sensors has not been addressed (Vigors et al., Reference Vigors, Sweeney, O’Shea, Browne and O’Doherty2014).

Gene polymorphisms in amino acid sensing

In humans, 17 SNPs (14 ns) and 12 SNPs (6 ns) were reported for TAS1R1 and TAS1R3, respectively (Kim et al., Reference Kim, Wooding, Riaz, Jorde and Drayna2006). These polymorphisms have been associated with a lower ability to taste glutamate (Chen et al., Reference Chen, Alarcon, Tharp, Ahmed, Estrella, Greene, Rucker and Breslin2009) and with specific food choices (Han et al., Reference Han, Keast and Roura2018). In particular, the research published from Han et al. (Reference Han, Keast and Roura2018) reported that human carriers of one of the TAS1R1 SNPs consumed more fat and calories from a buffet meal. In addition, Raliou et al. (Reference Raliou, Wiencis, Pillias, Planchais, Eloit, Boucher, Trotier, Montmayeur and Faurion2009) showed that mGluR1 polymorphisms contributed to a lack of sensitivity to glutamate. Genetic variants in other AA sensors (i.e. CaSR and GPRC6A) have also been reported; however, the physiological impact of this variation is currently unknown.

In pigs, an SNP analysis of 79 pig genomes (belonging to 14 different breeds) revealed 13 (5 ns and 1 stop-lost) and 9 (1 ns) polymorphisms in pTasS1r1 and pTas1r3, respectively (Da Silva et al., Reference Da Silva, De Jager, Burgos, Reverter, Perez-Enciso and Roura2014). The research also showed several SNPs for the other AA sensors: 22 (2 ns), 6 (3 ns), 16 (1 ns) and 28 (2 ns) for CaSR, GPRC6A, mGluR1 and mGluR4, respectively. Clop et al. (Reference Clop, Sharaf, Castello, Ramos-Onsins, Cirera, Mercade, Derdak, Huisman, Fredholm, van As and Sanchez2016) identified 31 (including 1 splice, 1 stop-gained and 1 stop lost, 3 frame shifts and 4 moderate impact) pTas1r1 variants and 14 (including 1 stop gained and 1 moderate impact) pTas1r3 variants. In addition, they identified an mGluR1 SNP linked to umami taste, feed intake and growth. However, the incidence of SNP in AA sensors compared to the bitter sensing system was very low (Da Silva et al., Reference Da Silva, De Jager, Burgos, Reverter, Perez-Enciso and Roura2014). This limited number of ns SNPs may indicate that AA receptor/transceptor functions are highly conserved across individuals and across pig breeds.

Lipid/fatty acid sensing

Fats are an essential dietary energy source that play a key role in gut hormone release (Hara et al., Reference Hara, Kashihara, Ichimura, Kimura, Tsujimoto and Hirasawa2014). Triglycerides, the main dietary fat source, are digested by lipases releasing free fatty acids (FFAs) and monoacylglycerides.

Fatty acid sensors and transceptors in the gastrointestinal tract

The chemosensory system for fats has evolved mainly around the sensing of FFAs and consists of an array of nine receptors (FFARs) and transceptors featuring a degree of specificity based on chain length (Table 3). In particular, the main ligands for FFAR2 and FFAR3 and olfactory receptor OLFR78 are short-chain fatty acids (SCFAs). The receptors FFAR1 and GPR84 showed the highest affinity for medium-chain fatty acids (MCFAs)(Wang et al. Reference Wang, Wu, Simonavicius, Tian and Ling2006; Liu et al., Reference Liu, Costanzo, Evans, Archer, Nowson, Duesing and Keast2018), whereas FFAR4 (also kwon as GPR120) and fatty acid (FA)-binding protein 2 (FABP2), FA transport protein 4 (FATP4) and cluster of differentiation 36 (CD36) have been characterized as receptors for long-chain fatty acids (LCFAs) (Bachmanov and Beauchamp, Reference Bachmanov and Beauchamp2007; Mattes, Reference Mattes2011). In addition, GPR119 has been proposed as a putative receptor for endogenous lipids containing oleic acid (e.g. oeloylethanolamide) and 2-monoacylglycerol (Hansen et al., Reference Hansen, Rosenkilde, Holst and Schwartz2012).

Table 3. Main FFA receptors and transceptors known to be involved in GIT sensing in humans and pigs1

FFA = free fatty acids; GIT = gastrointestinal tract; GI = gastrointestinal; FFARs = free fatty acid receptors; CCK = cholecystokinin; CD36 = cluster of differentiation 36; FABP2 = fatty acid binding protein 2; FATP4 = fatty acid transporter 4; SLC16A1 = solute carrier family 16 member; GLP-1 = glucagon-like peptide 1; GIP = glucose insulinotropic peptide; GLP-1 = glucagon-like peptide 1; GPBAR1 = G-protein-coupled bile receptor; TGR5 = Takeda G-protein-coupled receptor 5; GPR = G-protein-coupled receptor; MCT1 = monocarboxylate transporter 1; OLFR78 = olfactory receptor 78; PYY = peptide YY

1 All the receptors and transporters presented in the table are relevant to humans and pigs except if noted with the superscript 4.

2 Table references are provided in Supplementary Material S1.

3 Refers to the oral sensation elicited by free fatty acids (Note: to date, the FFA sensing has not achieved full recognition as a primary taste type by the sensory science community).

4 No literature evidence of the existence or functionality of these genes has been found in pigs.

Fatty acids and the enteroendocrine system

The receptors FFAR1 and FFAR4 are present throughout the GIT found in EECs. The uptake of dietary FFAs is slow (compared to sugars and AA) and requires bile acids secreted in the duodenum. In contrast, FFAR2, FFAR3 and OLFR78 are preferentially expressed in the colon, where abundant SCFAs are produced resulting from bacterial fermentation (Canfora et al. Reference Canfora, Jocken and Blaak2015; Fleischer et al., Reference Fleischer, Bumbalo, Bautze, Strotmann and Breer2015). GPR84 has been reported in mouse gastric mucosa (Widmayer et al., Reference Widmayer, Kusumakshi, Hägele, Boehm and Breer2017). GPR119 expression has been associated with EECs (L-cells) and pancreatic cells (Overton et al. Reference Overton, Fyfe and Reynet2008; Lan et al., Reference Lan, Vassileva, Corona, Liu, Baker, Golovko, Abbondanzo, Hu, Yang, Ning, Del Vecchio, Poulet, Laverty, Gustafson, Hedrick and Kowalski2009; Hansen et al., Reference Hansen, Rosenkilde, Holst and Schwartz2012).

On the one hand, intragastric administration of dietary oral gavage of LCFA has been reported to increase the orexigenic (appetite) hormone ghrelin secretion presumably through the stimulation of FFAR4 (Janssen et al. Reference Janssen, Laermans, Iwakura, Tack and Depoortere2012). In addition, the activation of FFAR2-expressing gastric X/A-cells by SCFA inhibited ghrelin (Engelstoft et al. Reference Engelstoft, Park, Sakata, Kristensen, Husted, Osborne-Lawrence, Piper, Walker, Pedersen, Nøhr, Pan, Sinz, Carrington, Akiyama, Jones, Tang, Ahmed, Offermanns, Egerod, Zigman and Schwartz2013). Short-chain fatty acid can reach the stomach through the portal vein (Morrison and Preston, Reference Morrison and Preston2016). This may be indicative of an excessive fermentation occurring in the lower GIT which is consistent with an anorexegenic (satiating) response. On the other hand, some FFARs have also been related to anorexigenic events associated with CCK and/or GLP-1 and GIP. An acute oral dose of butyrate increased GLP-1 and PYY levels in mice, presumably through FFAR3 (Lin et al. Reference Lin, Frassetto, Kowalik, Nawrocki, Lu, Kosinski, Hubert, Szeto, Yao, Forrest and Marsh2012). The expression of GPR84 in X/A-like ghrelin cells and surface cells suggests an important role of MCFA in the developing gastric mucosa of suckling mice (Widmayer et al., Reference Widmayer, Kusumakshi, Hägele, Boehm and Breer2017). In addition, SCFA olfactory receptor OLFR78 and GLP-1 and PYY co-express in murine colonic L-cells (Pluznick, Reference Pluznick2014; Fleischer et al., Reference Fleischer, Bumbalo, Bautze, Strotmann and Breer2015). Furthermore, GPR119 ligands (i.e. monoglycerides) triggered GLP-1 secretion from intestinal primary cultures, particularly from colon (Moss et al., Reference Moss, Glass, Diakogiannaki, Pais, Lenaghan, Smith, Wedin, Bohlooly-Y, Gribble and Reimann2016). Fatty acid transceptors CD36 and FATP4 have been also reported to mediate lipid-induced gut hormone secretion (Sundaresan et al., Reference Sundaresan, Shahid, Riehl, Chandra, Nassir, Stenson, Liddle and Abumrad2013; Poreba et al., Reference Poreba, Dong, Li, Stahl, Miner and Brubaker2012).

In pigs, De Jager et al. (Reference De Jager, Zhan, Rzepus and Roura2013) reported the expression of FFAR1, FFAR2, FFAR3, FFAR4 and GPR84 in circumvallate papillae. In addition, Da Silva et al. (Reference Da Silva, De Jager, Burgos, Reverter, Perez-Enciso and Roura2014) revealed a very low incidence of allelic variants across FFARs and GPR84 compared to other TR genes such as the TAS2R family (bitter taste) indicating that FFARs were highly conserved in pigs.

The FFARs expression pattern described in pigs evidenced some differences compared to humans. In particular, FFAR2 and FFAR3 were predominantly found in the distal small intestine (Haenen et al., Reference Haenen, Zhang, Souza da Silva, Bosch, van der Meer, van Arkel, van den Borne, Perez Gutierrez, Smidt, Kemp, Muller and Hooiveld2013) while FFAR4 in colon (Colombo et al., Reference Colombo, Trevisi, Gandolfi and Bosi2012; van der Wielen et al., Reference van der Wielen, van Avesaat, de Wit, Vogels, Troost, Masclee, Koopmans, van der Meulen, Boekschoten, Muller, Hendriks, Witkamp and Meijerink2014). It is tempting to speculate that these findings may be related to the higher fermentative capacity of the hindgut of the adult pigs compared to humans (Stevens, Reference Stevens1988). In contrast, FFAR2 and FFAR3 were found expressed in colonic enteroendocrine L-cells responding to increased levels of SCFA (i.e. butyrate) released after high inclusion of resistant starch (Haenen et al., Reference Haenen, Zhang, Souza da Silva, Bosch, van der Meer, van Arkel, van den Borne, Perez Gutierrez, Smidt, Kemp, Muller and Hooiveld2013). In addition, a co-expression pattern was uncovered between FFAR2 and FFAR3 with PYY, GLP-1 and serotonin in pig colon (Weatherburn, Reference Weatherburn2015).

Gene polymorphisms in fatty acid sensing

The ability to sense fats has been associated with an increased consumption of fatty foods, higher BMI and obesity (Stewart et al., Reference Stewart, Newman and Keast2011; Ichimura et al., Reference Ichimura, Hirasawa, Poulain-Godefroy, Bonnefond, Hara, Yengo, Kimura, Leloire, Liu, Iida, Choquet, Besnard, Lecoeur, Vivequin, Ayukawa, Takeuchi, Ozawa, Tauber, Maffeis, Morandi, Buzzetti, Elliott, Pouta, Jarvelin, Körner, Kiess, Pigeyre, Caiazzo, Van Hul, Van Gaal, Horber, Balkau, Lévy-Marchal, Rouskas, Kouvatsi, Hebebrand, Hinney, Scherag, Pattou, Meyre, Koshimizu, Wolowczuk, Tsujimoto and Froguel2012). FFA4 gene variants have been found to have a significant impact on receptor responses (Hudson et al., Reference Hudson, Murdoch and Milligan2013). In addition, the FFAR4 mutation was found to increase the risk of obesity, demonstrating the key role in fat sensing and the control of energy balance in humans and rodents (Ichimura et al., Reference Ichimura, Hirasawa, Poulain-Godefroy, Bonnefond, Hara, Yengo, Kimura, Leloire, Liu, Iida, Choquet, Besnard, Lecoeur, Vivequin, Ayukawa, Takeuchi, Ozawa, Tauber, Maffeis, Morandi, Buzzetti, Elliott, Pouta, Jarvelin, Körner, Kiess, Pigeyre, Caiazzo, Van Hul, Van Gaal, Horber, Balkau, Lévy-Marchal, Rouskas, Kouvatsi, Hebebrand, Hinney, Scherag, Pattou, Meyre, Koshimizu, Wolowczuk, Tsujimoto and Froguel2012). In addition, FA transporter CD36 was shown to play a crucial role in oral fat sensing as well (Pepino et al., Reference Pepino, Love-Gregory, Klein and Abumrad2012). Genetic CD36 variants were associated with the taste intensity of oleic acid and triolein, total dietary fat and energy intake, and the development of obesity in teenagers (Toguri, Reference Toguri2008; Pepino et al., Reference Pepino, Love-Gregory, Klein and Abumrad2012; Keller et al., Reference Keller, Liang, Sakimura, May, van Belle, Breen, Driggin, Tepper, Lanzano, Deng and Chung2012; Daoudi et al., Reference Daoudi, Plesník, Sayed, Šerý, Rouabah, Rouabah and Khan2015; Mrizak et al., Reference Mrizak, Sery, Plesnik, Arfa, Fekih, Bouslema, Zaouali, Tabka and Khan2015). In addition, CD36 gene variants have also been implicated in obesity, type 2 diabetes, the metabolic syndrome, hypertension and coronary heart disease (Precone et al., Reference Precone, Beccari, Stuppia, Baglivo, Paolacci, Manara, Miggiano, Falsini, Trifirò, Zanlari, Herbst, Unfer and Bertelli2019).

In pigs Da Silva et al. (Reference Da Silva, De Jager, Burgos, Reverter, Perez-Enciso and Roura2014) revealed a low incidence of polymorphisms in FFARs genes when comparing to bitter taste sensors. In particular, the total number of SNP for FFAR1, FFAR2, FFAR3, and FFAR4 were 8 (4 ns), 11 (1 ns), 11 (2 ns) and 1 (0 ns), respectively (Da Silva et al., Reference Da Silva, De Jager, Burgos, Reverter, Perez-Enciso and Roura2014). In addition, the results published from the genomic analysis in pigs by Clop et al. (Reference Clop, Sharaf, Castello, Ramos-Onsins, Cirera, Mercade, Derdak, Huisman, Fredholm, van As and Sanchez2016) identified three CD36 variants associated with growth and fat deposition. Finally, significant differences in allele frequencies of FFAR4 were observed between two extreme pig groups based on growth rates (Fontanesi et al., Reference Fontanesi, Bertolini, Scotti, Schiavo, Colombo, Trevisi, Riban, Buttazzoni, Russo and Dall’Olio2015).

Bitter sensing

Bitter sensing has been associated with harmful contaminants, toxic compounds and general synthetic chemicals such as pharmaceuticals present in foods/feeds (Nelson and Sanregret, Reference Nelson and Sanregret1997; Meyerhof et al., Reference Meyerhof, Batram, Kuhn, Brockhoff, Chudoba, Bufe, Appendino and Behrens2009). These compounds cause defensive and protective responses in the host including food aversion, vomiting, and inhibition of gastric motility and activation of efflux from enterocytes accompanied by an increase in satiation and satiety (Sarkadi et al., Reference Sarkadi, Homolya, Szakacs and Varadi2006; Jeon et al., Reference Jeon, Seo and Osborne2011; Avau et al., Reference Avau, Rotondo, Thijs, Andrews, Janssen, Tack and Depoortere2015; Deloose et al., Reference Deloose, Corsetti, Van Oudenhove, Depoortere and Tack2017a and Reference Deloose, Janssen, Corsetti, Biesiejiersji, Masuy, Rotondo, Van Oudenhove, Depoortere and Tack2017b). In contrast, some non-toxic plant-derived compounds (such as polyphenols) may also elicit bitter taste (Soares et al., Reference Soares, Silva, Garcia-Estevez, GroBmann, Bras, Brandao, Mateus, de Freitas, Behrens and Meyerfof2018). Overall, close to 1000 compounds are known to be bitter to humans while 81 to laboratory rodents and 27 to pigs (Wang et al., Reference Wang, Fu, Navarro and Roura2017; Roura and Fu, Reference Roura and Fu2017; Dagan-Wiener et al., Reference Dagan-Wiener, Di Pizio, Nissim, Bahia, Dubovski, Margulis and Masha2019).

Bitter sensors in the gastrointestinal tract

Bitterants activate the TAS2R family, which consists of 25 functional genes in humans (Meyerhof et al., Reference Meyerhof, Batram, Kuhn, Brockhoff, Chudoba, Bufe, Appendino and Behrens2009). The size of the bitter taste receptor (TAS2R) repertoire is species specific, ranging from the 36 genes in the rat to none in carnivorous marine mammals (Roura and Foster, Reference Roura and Foster2018). The sensitivity of pigs to bitterness has been widely reported in the literature (Nelson and Sanregret, Reference Nelson and Sanregret1997; Danilova et al., Reference Danilova, Roberts and Hellekant1999; Roura et al., Reference Roura, Humphrey, Tedó and Ipharragerre2008; Roura and Navarro, Reference Roura and Navarro2018). The porcine pTas2r repertoire was recently characterized consisting of 16 functional genes and 3 pseudogenes (Colombo et al., Reference Colombo, Trevisi, Gandolfi and Bosi2012; Roura, et al., Reference Roura, Koopmans, Lallès, Le Huerou-Luron, de Jager, Schuurman and Val-Laillet2016; Roura and Fu, Reference Roura and Fu2017).

Bitter sensing and the enteroendocrine system

TAS2R transcripts have been observed in the oral and GIT mucosa of several mammalian species including humans and pigs (Rozengurt, Reference Rozengurt2006; Da Silva et al., Reference Da Silva, De Jager, Burgos, Reverter, Perez-Enciso and Roura2014). In humans, TAS2R5 and TAS2R38 have been co-localized with GLP-1-, CCK- and PYY-labelled EECs in the human small intestine and colon and TAS2R10 with ghrelin cells in the human stomach (Park et al., Reference Park, Kim, Kim, Lee, Jeong, Kim and Jang2015; Latorre et al., Reference Latorre, Huynh, Mazzoni, Gupta, Bonora, Clavenzani, Chang, Mayer, De Giorgio and Sternini2016; Wang et al., Reference Wang, Liszt, Deloose, Canovai, Thijs, Farre, Ceulemans, Lannoo, Tack and Depoortere2019). Bitter herbal medicines were shown to affect GLP-1 and CCK release in EEC lines (Avau and Depoortere, Reference Avau and Depoortere2016). However, the active compounds of the medicinal extracts studied remain to be identified. Finally, in tuft cells, bitter agonist denatonium benzoate elicited a paracrine activation of enterocytes presumably following the release of acetylcholine (Schutz et al., Reference Schutz, Jurastow, Bader, Ringer, von Engelhardt, Chubanov, Gudermann, Diener, Kunner, Krasteva-Christ and Weihe2015).

The presence of pTas2r in the porcine GIT has been reported by several groups (Colombo et al., Reference Colombo, Trevisi, Gandolfi and Bosi2012; Da Silva et al., Reference Da Silva, De Jager, Burgos, Reverter, Perez-Enciso and Roura2014; Ribani et al., Reference Ribani, Bertolini, Schiavo, Scotti, Utzeri, Dall’Olio, Trevisi, Bosi and Fontanesi2017; Clop et al., Reference Clop, Sharaf, Castello, Ramos-Onsins, Cirera, Mercade, Derdak, Huisman, Fredholm, van As and Sanchez2016). However, little is known to date about the function, except that dietary quinine and caffeine increased plasma insulin and GLP-1 (Fu et al., Reference Fu, Val-Laillet, Guerin and Roura2018).

Gene polymorphisms in bitter sensing

TAS2R38 variants determine the sensitivity to bitter substance phenylthiocarbamide in humans (Sandell and Breslin, Reference Sandell and Breslin2006; Risso et al., Reference Risso, Mezzavilla, Pagani, Robino, Morini, Tofanelli, Carrai, Campa, Barale, Caradonna, Gasparini, Luiselli, Wooding and Drayna2016) and have been associated with food preferences (Sandell and Breslin, Reference Sandell and Breslin2006), alcohol intake (Duffy et al., Reference Duffy, Davidson, Kidd, Kidd, Speed, Pakstis, Reed, Snyder and Bartoshuk2004), obesity (Tepper et al., Reference Tepper, Koelliker, Zhao, Ullrich, Lanzara, D’Adam, Ferrara, Ulivi, Esposito and Gasparini2008) and susceptibility to respiratory pathogens (Lee et al., Reference Lee, Xiong, Kofonow, Chen, Lysenko, Jiang, Abraham, Doghramji, Adappa, Palmer, Kennedy, Beauchamp, Doulias, Ischiropoulos, Kreindler, Reed and Cohen2012). Similarly, other gene variants of TAS2R14 and TAS2R50 have been associated with human diseases such as cancer and cardiovascular disease, respectively (Campa et al., Reference Campa, Vodicka, Pardini, Naccarati, Carrai, Vodickova, Novotny, Hemminki, Försti, Barale and Canzian2010; Akao et al., Reference Akao, Polisecki, Kajinami, Trompet, Robertson, Ford, Jukema, de Craen, Westendorp, Shepherd, Packard, Buckley and Schaefer2012). In addition, TAS2R16 variants appear to have had an evolutionary role to prevent consumption of dangerous raw foods (Valente et al., Reference Valente, Alvarez, Marques, Gusmão, Amorim, Seixas and Prata2018). Other genetic TAS2Rs have been related to the perception of bitterness in coffee (TAS2R2, TAS2R4 and TAS2R5), alcohol consumption (TAS2R13) and grapefruit liking (TAS2R19) (Hayes et al., Reference Hayes, Feeney and Alle2013).

The porcine bitter taste system presented a high incidence of allelic variants compared with the non-bitter taste genes, suggesting a potential role for these genes in ecological adaptation in pigs (Da Silva et al., Reference Da Silva, De Jager, Burgos, Reverter, Perez-Enciso and Roura2014). This high variability within and between species of the TAS2R gene repertoire seems to reflect an adaptive nature to survive in specific/novel ecological niches particularly to avoid plant-derived toxins. In addition, three phenotype–genotype studies reported SNPs with functional significance on the porcine bitter receptors pTas2r38, pTas2r39 (Clop et al., Reference Clop, Sharaf, Castello, Ramos-Onsins, Cirera, Mercade, Derdak, Huisman, Fredholm, van As and Sanchez2016; Ribani et al., Reference Ribani, Bertolini, Schiavo, Scotti, Utzeri, Dall’Olio, Trevisi, Bosi and Fontanesi2017) and pTas2r40 (Herrero-Medrano et al., Reference Herrero-Medrano, Megens, Groenen, Bose, Pérez-Enciso and Crooijmans2014). The associations reflected the impact of the fixed alleles on pig growth, fat deposition and environmental adaptation.

Microbial and parasite sensing in the gastrointestinal tract

While the role of nutrient receptors and transceptors has been mostly linked to exogenous or dietary nutrients and potential harmful compounds, recent findings indicate that this sensors may also respond to compounds produced within the intestinal tract. For example, products of the microbial population in the GIT, such as SCFA and MCFA, have the capacity to affect the chemosensory system. Similarly, metabolites produced in the GIT by parasitic or protozoan infections may also be able to activate some of the receptors and transceptors.

Microbial metabolites

SCFA and MCFA resulting from bacterial fermentation in the GIT affect the expression of nutrient sensors and gut peptides in EECs (Steensels and Depoortere, Reference Steensels and Depoortere2018). A decrease in FA sensors (FFAR1, FFAR4 and CD36), together with an increase in glucose and AA sensors (TAS1R3 and SGLT1), were reported in germ-free mice (Duca et al., Reference Duca, Swartz, Sakar and Covasa2012; Swartz et al., Reference Swartz, Duca, de Wouters, Sakar and Covasa2012). These changes were associated with reduced CCK, GLP-1 and PYY. In addition, bacterial endotoxins activate the toll-like receptors which are co-localized in CCK, PYY and serotonin secreting EECs (Bogunovic et al., Reference Bogunovic, Dave, Tilstra, Chang, Harpaz, Xiong, Mayer and Plevy2007; Larraufie et al., Reference Larraufie, Dore, Lapaque and Blottiere2017).

Commensal bacteria have evolved to produce metabolites that chemically mimic mammalian agonists and trigger eukaryotic cellular responses (Cohen et al., Reference Cohen, Kang, Chu, Huang, Gordon, Reddy, Ternei, Craig and Brady2015). Bacterial N-acyl amides showed high affinity to host GPR119 functioning to regulate GIT physiology, gut hormones and glucose homeostasis (Cohen et al., Reference Cohen, Esterhazy, Kim, Lemetre, Aguilar, Gordon, Pickard, Cross, Emiliano, Han, Chu, Vila-Farres, Kaplitt, Rogoz, Calle, Hunter, Bitok and Brady2017). Sung et al. (Reference Sung, Kim, Denou, Soltys, Hamza, Byme, Masson, Park, Wishart, Madsen, Shertzer and Dyck2017) replicated the positive effect of oral resveratrol by fecal microbiome transplants to obese (but naive to dietary resveratrol) mice. In addition, Clostridium coli and Escherichia coli were shown to affect intestinal motility by modulating serotonin synthesis from enterochromaffin cells (Cao et al., Reference Cao, Liu, An, Zhou, Liu, Xu, Dong, Wang, Yan, Jiang and Wang2017). Taken together, robust evidences are accumulating, showing that gut microbes have evolved to interact and modulate animal host GIT physiology.

Parasites

Parasitic worms and protozoan infections initiate a signalling cascade in tuft cells mediated by TAS1Rs and/or TAS2Rs (Gerbe et al., Reference Gerbe, Sidot, Smyth, Ohmoto, Matsumoto, Dardalhon, Cesses, Garnier, Pouzolles, Brulin, Bruschi, Harcus, Zimmermann, Taylor, Maizels and Jay2016; Howitt et al., Reference Howitt, Lavoie, Michaud, Blum, Tran, Weinstock, Gallini, Redding, Margolskee, Osborne, Artis and Garrett2016; von Moltke et al., Reference von Moltke, Ji, Liang and Locksley2016). Tuft cells orchestrate type 2 cell-mediated immunity in a process where TR sensing signals mediate the differentiation of epithelial crypt progenitors to tuft cells and goblet cells. Tuft and goblet cells hyperplasia is instrumental to achieve worm clearance (Zhao et al., Reference Zhao, Urban, Anthony, Sun, Stiltz, van Rooijen, Wynn, Gause and Shea-Donohue2008). Furthermore, the succinate receptor and TAS2Rs are expressed on tuft cells to detect the metabolites secreted by the parasites (Nadjsombati et al., Reference Nadjsombati, McGinty, Lyons-Cohen, Jaffe, DiPeso, Schneider, Miller, Pollack, Nagana Gowda, Fontana, Erle, Anderson, Locksley, Raftery and von Moltke2018; Luo et al., Reference Luo, Chen, Xue, Zhao, Lu, Li, Li, Du, Liu, Wang, Liu and Huang2019).

Conclusions

The nutrient and non-nutrient sensing in the GIT tract has evolved as a continuum function necessary to orchestrate ingestion, digestion, absorption, metabolism and neutralization of harmful substances. The mechanisms related to the sensing of carbohydrates, AAs, FAs, bitter compounds and microbial and parasite metabolites involve specialized cells in the enteric mucosa (i.e. EEC) that elicit hormonal responses (i.e. CCK, GLP-1, PYY, ghrelin, etc.) which, in turn, mediate changes in passage rate and appetite. Gene variations have been related to food choices in humans while in pigs to ecological adaptations particularly regarding the bitter taste receptor repertoire. In addition, genetic mutations have the potential to lead to the development of novel nutritional strategies in pigs, for example, regarding FFA sensing. In addition, our understanding on the impact of gut microbiome on the host’s gut-brain communications has started to unfold.

Acknowledgements

None.

Eugeni Roura 0000-0002-9073-9946

Declaration of interest

The authors declare no conflicts of interest.

Ethics statement

Not applicable.

Software and data repository resources

Not applicable.

Supplementary material

To view supplementary material for this article, please visit https://doi.org/10.1017/S1751731119001794

References

Aji, GK, Warren, FJ and Roura, E 2019. Starch, salivary α-amylase activity and maltose taste thresholds in humans. Chemical Senses 44, 249256.CrossRefGoogle ScholarPubMed
Akao, H, Polisecki, E, Kajinami, K, Trompet, S, Robertson, M, Ford, I, Jukema, JW, de Craen, AJ, Westendorp, RG, Shepherd, J, Packard, C, Buckley, BM and Schaefer, EJ 2012. KIF6, LPA, TAS2R50, and VAMP8 genetic variation, low density lipoprotein cholesterol lowering response to pravastatin, and heart disease risk reduction in the elderly. Atherosclerosis 220, 456462.CrossRefGoogle ScholarPubMed
Avau, B and Depoortere, I 2016. The bitter truth about bitter taste receptors: beyond sensing bitter in the oral cavity. Acta Physiologica 216, 407420.CrossRefGoogle ScholarPubMed
Avau, B, Rotondo, A, Thijs, T, Andrews, CN, Janssen, P, Tack, J and Depoortere, I 2015. Targetin extra-oral bitter taste receptors modulates gastrointestinal motility with effects on satiation. Scientific Reports 5, 15985. doi: 10.1038/srep15985.CrossRefGoogle Scholar
Bachmanov, AA and Beauchamp, GK 2007. Taste receptor genes. Annual Review of Nutrition 27, 389414.CrossRefGoogle ScholarPubMed
Badman, MK and Flier, JS 2005. The gut and energy balance: visceral allies in the obesity wars. Science 307, 19091914.CrossRefGoogle ScholarPubMed
Barretto, RPJ, Gillis-Smith, S, Chandrashekar, J, Yarmolinsky, DA, Schnitzer, MJ, Ryba, NJP and Zuker, CS 2015. The neural representation of taste quality at the periphery. Nature 517, 373376.CrossRefGoogle ScholarPubMed
Berridge, KC and Robinson, TE 1998. What is the role of dopamine in reward: hedonic impact, reward learning, or incentive salience? Brain Research Reviews 28, 309369.CrossRefGoogle ScholarPubMed
Bogunovic, M, Dave, SH, Tilstra, JS, Chang, DT, Harpaz, N, Xiong, H, Mayer, LF and Plevy, SE 2007. Enteroendocrine cells express functional toll-like receptors. American Journal of Physiology. Gastrointestinal and Liver Physiology 292, 17701783.CrossRefGoogle ScholarPubMed
Broer, S 2008. Amino acid transport across mammalian renal and intestinal epithelia. Physiological Reviews 88, 249286.CrossRefGoogle Scholar
Campa, D, Vodicka, P, Pardini, B, Naccarati, A, Carrai, M, Vodickova, L, Novotny, J, Hemminki, K, Försti, A, Barale, R and Canzian, F 2010. A gene-wide investigation on polymorphisms in the taste receptor 2R14 (TAS2R14) and susceptibility to colorectal cancer. BioMed Central Medical Genetics 11, 88. doi: 10.1186/1471-2350-11-88.CrossRefGoogle ScholarPubMed
Canfora, EE, Jocken, JW and Blaak, EE 2015. Short-chain fatty acids in control of body weight and insulin sensitivity. Natural Reviews Endocrinology 11, 577591.CrossRefGoogle ScholarPubMed
Cao, H, Liu, X, An, Y, Zhou, G, Liu, Y, Xu, M, Dong, W, Wang, S, Yan, F, Jiang, K and Wang, B 2017. Dysbiosis contributes to chronic constipation development via regulation of serotonin transporter in the intestine. Scientific Reports 7, 10322. doi: 10.1038/s41598-017-10835-8.CrossRefGoogle ScholarPubMed
Chen, QY, Alarcon, S, Tharp, A, Ahmed, OM, Estrella, NL, Greene, TA, Rucker, J and Breslin, PA 2009. Perceptual variation in umami taste and polymorphisms in TAS1R taste receptor genes. American Journal of Clinical Nutrition 90, 770779.CrossRefGoogle ScholarPubMed
Cheng, MW, Chegeni, M, Kim, KH, Zhang, G, Benmoussa, M, Quezada-Calvillo, R, Nichols, BL and Hamaker, BR 2014. Different sucrose-isomaltase response of Caco-2 cells to glucose and maltose suggests dietary maltose sensing. Journal of Clinical Biochemistry and Nutrition 54, 5560.CrossRefGoogle ScholarPubMed
Choi, S, Lee, M, Shiu, AL, Yo, SJ, Halldén, G and Aponte, GW 2007. GPR93 activation by protein hydrolysate induces CCK transcription and secretion in STC-1 cells. American Journal of Physiology. Gastrointestinal and Liver Physiology 292, 13661375.CrossRefGoogle ScholarPubMed
Clop, A, Sharaf, A, Castello, A, Ramos-Onsins, S, Cirera, S, Mercade, A, Derdak, S, Huisman, A, Fredholm, M, van As, P and Sanchez, A 2016. Identification of protein-damaging mutations in 10 swine taste receptors and 191 appetite-reward genes. BioMed Central Genomics 17, 685. doi: 10.1186/s12864-016-2972-z.Google Scholar
Cohen, LJ, Esterhazy, D, Kim, S-H, Lemetre, C, Aguilar, RR, Gordon, EA, Pickard, AJ, Cross, JR, Emiliano, AB, Han, SM, Chu, J, Vila-Farres, X, Kaplitt, J, Rogoz, A, Calle, PY, Hunter, C, Bitok, JK and Brady, SF 2017. Commensal bacteria make GPCR ligands that mimic human signalling molecules. Nature 549, 4853.CrossRefGoogle ScholarPubMed
Cohen, LJ, Kang, H-S, Chu, J, Huang, Y-H, Gordon, EA, Reddy, BVB, Ternei, MA, Craig, JW and Brady, SF 2015. Functional metagenomic discovery of bacterial effectors in the human microbiome and isolation of commendamide, a GPCR G2A/132 agonist. Proceedings of the National Academy of Sciences of the United States of America 112, 48254834.CrossRefGoogle ScholarPubMed
Colombo, M, Trevisi, P, Gandolfi, G and Bosi, P 2012. Assessment of the presence of chemosensing receptors based on bitter and fat taste in the gastrointestinal tract of young pig. Journal of Animal Science 90, 128130.CrossRefGoogle ScholarPubMed
Dagan-Wiener, A, Di Pizio, A, Nissim, I, Bahia, MS, Dubovski, N, Margulis, E and Masha, Y 2019. BitterDB: taste ligands and receptors database in 2019. Nucleic Acids Research 47, 11791185.CrossRefGoogle ScholarPubMed
Daniel, H and Zietek, T 2015. Taste and move: glucose and peptide transporters in the gastrointestinal tract. Experimental Physiology 100, 14411450.CrossRefGoogle ScholarPubMed
Danilova, V, Roberts, T and Hellekant, G 1999. Responses of single taste fibers and whole chorda tympani and glossopharyngeal nerve in the domestic pig, Sus scrofa. Chemical Senses 24, 301316.CrossRefGoogle ScholarPubMed
Daoudi, H, Plesník, J, Sayed, A, Šerý, O, Rouabah, A, Rouabah, L and Khan, NA 2015. Oral fat sensing and CD36 gene polymorphism in Algerian lean and obese teenagers. Nutrients 7, 90969104.CrossRefGoogle ScholarPubMed
Da Silva, EC, De Jager, N, Burgos, W, Reverter, A, Perez-Enciso, M and Roura, E 2014. Characterization of the porcine GPCR nutrient sensor and taste receptor gene repertoire across international and local domestic breeds and wild populations. BioMed Central Genomics 15, 1057.Google Scholar
Dehmlow, H, Alvarez Sanchez, R, Bachmann, S, Bissantz, C, Bliss, F, Conde-Knape, K, Graf, M, Martin, RE, Obst Sander, U, Raab, S, Richter, HGF, Sewing, S, Sprecher, U, Ullmer, C and Mattei, P 2013. Discovery and optimisation of 1-hydroxyimino-3,3-diphenylpropane, a new class of orally active GPBAR1 (TGR5) agonists. Bioorganic & Medical Chemistry Letters 23, 46274632.CrossRefGoogle Scholar
De Jager, N, Zhan, M, Rzepus, M and Roura, E 2013. Towards defining the taste receptor repertoire in the pig. In Proceedings of Manipulating Pig Production XIV, APSA Conference, 24–27 November 2013, Melbourne, Australia, pp. 47.Google Scholar
Deloose, E, Corsetti, M, Van Oudenhove, L, Depoortere, I and Tack, J 2017a. Intragastric infusion of the bitter tastan quinine suppresses hormone release and antral motility during the fastin state in health female volunteers. Neurogastroenterology and Motility 30, 313171. doi: 10.1111/nmo.13171.Google Scholar
Deloose, E, Janssen, P, Corsetti, M, Biesiejiersji, J, Masuy, I, Rotondo, A, Van Oudenhove, L, Depoortere, I and Tack, J 2017b. Intragastric infusion of denatonium benzoate attenuates interdigestive gastric motility and hunger scores in healthy female volunteers. The American Journal of Clinical Nutrition 105, 580588.CrossRefGoogle ScholarPubMed
Depoortere, I 2014. Taste receptors of the gut: emerging roles in health and disease. Gut 63, 179190.CrossRefGoogle ScholarPubMed
Diakogiannaki, E, Pais, R, Tolhurst, G, Parker, HE, Horscroft, J, Horscroft, J, Rauscher, B, Zietek, T, Daniel, H, Gribble, FM and Reinmann, F 2013. Oligopeptides stimulate glucagon-like peptide-1 secretion in mice through proton-coupled uptake and the calcium-sensing re-ceptor. Diabetologia 56, 26882696.CrossRefGoogle Scholar
Diallinas, G 2017. Transceptors as a functional link of transporters and receptors. Microbial cell 4, 6973.CrossRefGoogle ScholarPubMed
Dias, AG, Eny, KM, Cockburn, M, Chiu, W, Nielsen, DE, Duizer, L and El-Sohemy, A 2015. Variation in the TAS1R2 gene, sweet taste perception and intake of sugars. Journal of Nutrigenetics and Nutrigenomics 8, 8190.CrossRefGoogle ScholarPubMed
Douard, V and Ferraris, RP 2008. Regulation of the fructose transporter GLUT5 in health and disease. American Journal of Physiology, Endocrinology and Metabolism 295, 227237.CrossRefGoogle ScholarPubMed
Duca, FA, Swartz, TD, Sakar, Y and Covasa, M 2012. Increased oral detection, but decreased intestinal signaling for fats in mice lacking gut microbiota. PLoS ONE 7, e39748. doi: 10.1371/journal.pone.0039748.CrossRefGoogle ScholarPubMed
Duffy, VB, Davidson, AC, Kidd, JR, Kidd, KK, Speed, WC, Pakstis, AJ, Reed, DR, Snyder, DJ and Bartoshuk, LM 2004. Bitter receptor gene (TAS2R38), 6-n-propylthiouracil (PROP) bitterness and alcohol intake. Alcoholism Clinical and Experimental Research 28, 16291637.CrossRefGoogle ScholarPubMed
Engelstoft, MS, Park, WM, Sakata, I, Kristensen, LV, Husted, AS, Osborne-Lawrence, S, Piper, PK, Walker, AK, Pedersen, MH, Nøhr, MK, Pan, J, Sinz, CJ, Carrington, PE, Akiyama, TE, Jones, RM, Tang, C, Ahmed, K, Offermanns, S, Egerod, KL, Zigman, JM and Schwartz, TW 2013. Seven transmembrane G protein-coupled receptor repertoire of gastric ghrelin cells. Molecular Metabolism 2, 376392.CrossRefGoogle ScholarPubMed
Eny, KM, Wolever, TM, Corey, PN and El-Sohemy, A 2010. Genetic variation in TAS1R2 (Ile191Val) is associated with consumption of sugars in overweight and obese individuals in 2 distinct populations. American Journal of Clinical Nutrition 92, 15011510.CrossRefGoogle ScholarPubMed
Fleischer, J, Bumbalo, R, Bautze, V, Strotmann, J and Breer, H 2015. Expression of odorant receptor Olfr78 in enteroendocrine cells of the colon. Cell Tissue Research 361, 697710.CrossRefGoogle ScholarPubMed
Fontanesi, L, Bertolini, F, Scotti, E, Schiavo, G, Colombo, M, Trevisi, P, Riban, A, Buttazzoni, A,Russo, V and Dall’Olio, S 2015. Next Generation Semiconductor based-sequencing of a nutrigenetics target gene (GPR120) and association with growth rate in Italian large white pigs. Animal Biotechnology 26, 9297.CrossRefGoogle ScholarPubMed
Fu, M, Val-Laillet, D, Guerin, S and Roura, E 2018. Dietary bitter compounds delayed gastric emptying and glucose uptake while increased plasma insulinotropic hormone GLP-1 in pigs. In Proceedings of the 14th International Symposium of Digestive Physiology of Pigs, 21–24 August 2018, Brisbane, Australia, pp. 207.Google Scholar
Gerbe, F, Sidot, E, Smyth, DJ, Ohmoto, M, Matsumoto, I, Dardalhon, V, Cesses, P, Garnier, L, Pouzolles, M, Brulin, B, Bruschi, M, Harcus, Y, Zimmermann, V, Taylor, N, Maizels, RM and Jay, P 2016. Intestinal epithelial tuft cells initiate type 2 mucosal immunity to helminth parasites. Nature 529, 226–30.CrossRefGoogle ScholarPubMed
Gerspach, AC, Steinert, RE, Schonenberger, L, Graber-Maier, A and Beglinger, C 2011. The role of the gut sweet taste receptor in regulating GLP-1, PYY, and CCK release in humans. American Journal of Physiology Endocrinology and Metabolism 301, 317325.CrossRefGoogle ScholarPubMed
Glaser, D, Wanner, M, Tinti, JM and Nofre, C 2000. Gustatory responses of pigs to various natural and artificial compounds known to be sweet in man. Food Chemistry 68, 375385.CrossRefGoogle Scholar
Glendinning, JI, Stano, S, Holter, M, Azenkot, T, Goldman, O, Margolskee, RF, Vasselli, JR and Sclafani, A 2015. Sugar-induced cephalic-phase insulin release is mediated by a T1r2+T1r3-independent taste transduction pathway in mice. American Journal of Physiology-Regulatory, Integrative and Comparative Physiology 309, 552560.CrossRefGoogle ScholarPubMed
Haenen, D, Zhang, J, Souza da Silva, C, Bosch, G, van der Meer, IM, van Arkel, J, van den Borne, JJ, Perez Gutierrez, O, Smidt, H, Kemp, B, Muller, M and Hooiveld, GJ 2013. A diet high in resistant starch modulates microbiota composition, SCFA concentrations, and gene expression in pig intestine. Journal of Nutrition 143, 274283.CrossRefGoogle ScholarPubMed
Haid, DC, Jordan-Biegger, C, Widmayer, P and Breer, H 2012. Receptors responsive to protein breakdown products in G-cells and D-cells of mouse, swine and human. Frontiers in Physiology 3, 65. doi: 10.3389/fphys.2012.00065.CrossRefGoogle ScholarPubMed
Han, P, Keast, R and Roura, E 2017. Salivary leptin and TAS1R2/TAS1R3 polymorphisms are related to sweet taste sensitivity and carbohydrate intake in a buffet meal. British Journal of Nutrition 118, 763770.CrossRefGoogle Scholar
Han, P, Keast, R and Roura, E 2018. TAS1R1 and TAS1R3 polymorphic genotypes relate to savoury and protein-rich food choices from a buffet meal in young adults. Nutrients 10, 1906. doi: 10.3390/nu10121906.CrossRefGoogle Scholar
Hansen, HS, Rosenkilde, MM, Holst, JJ and Schwartz, TW 2012. GPR119 as a fat sensor. Trends in Pharmacological Sciences 33, 374381.CrossRefGoogle ScholarPubMed
Hara, T, Kashihara, D, Ichimura, A, Kimura, I, Tsujimoto, G and Hirasawa, A 2014. Role of free fatty acid receptors in the regulation of energy metabolism. Biochimica et Biophysica Acta 1841, 12921300.CrossRefGoogle ScholarPubMed
Hayes, JE, Feeney, EL and Alle, AL 2013. Do polymorphisms in chemosensory genes matter for human ingestive behavior? Food Quality and Preference 30, 202216.CrossRefGoogle ScholarPubMed
Herrero-Medrano, JM, Megens, HJ, Groenen, MAM, Bose, M, Pérez-Enciso, M and Crooijmans, RPMA 2014. Whole-genome sequence analysis reveals differences in population management and selection of European low-input pig breeds. BioMed Central Genomics 15, 601. doi: 10.1186/1471-2164-15-601.Google ScholarPubMed
Howitt, MR, Lavoie, S, Michaud, M, Blum, AM, Tran, SV, Weinstock, JV, Gallini, CA, Redding, K, Margolskee, RF, Osborne, LC, Artis, D and Garrett, WS 2016. Tuft cells, taste-chemosensory cells, orchestrate parasite type 2 immunity in the gut. Science 351, 13291333.CrossRefGoogle Scholar
Hudson, BD, Murdoch, H and Milligan, G 2013. Minireview: the effects of species ortholog and SNP variation on receptors for free fatty acids. Molecular Endocrinology 27, 11771187.CrossRefGoogle ScholarPubMed
Humphrey, B, Tedó, G, Klasing, KC and Roura, E 2009. Caractérisation des polypeptides récepteurs umami porcins (pT1r1 et pT1r3). Journées de la Recherche Porcine 41, 165166.Google Scholar
Ichimura, A, Hirasawa, A, Poulain-Godefroy, O, Bonnefond, A, Hara, T, Yengo, L, Kimura, I, Leloire, A, Liu, N, Iida, K, Choquet, H, Besnard, P, Lecoeur, C, Vivequin, S, Ayukawa, K, Takeuchi, M, Ozawa, K, Tauber, M, Maffeis, C, Morandi, A, Buzzetti, R, Elliott, P, Pouta, A, Jarvelin, MR, Körner, A, Kiess, W, Pigeyre, M, Caiazzo, R, Van Hul, W, Van Gaal, L, Horber, F, Balkau, B, Lévy-Marchal, C, Rouskas, K, Kouvatsi, A, Hebebrand, J, Hinney, A, Scherag, A, Pattou, F, Meyre, D, Koshimizu, TA, Wolowczuk, I, Tsujimoto, G and Froguel, P 2012. Dysfunction of lipid sensor GPR120 leads to obesity in both mouse and human. Nature 483, 350354.CrossRefGoogle ScholarPubMed
Ikeda, K 1909. New seasonings. Journal of Tokyo Chemical Society 30, 820836.Google Scholar
Jang, HJ, Kokrashvili, Z, Theodorakis, MJ, Carlson, OD, Kim, BJ, Zhou, J, Kim, HH, Xu, X, Chan, SL, Juhaszova, M, Bernier, M, Mosinger, B, Margolskee, RF and Egan, JM 2007. Gut-expressed gustducin and taste receptors regulate secretion of glucagon-like peptide-1. Proceedings of the National Academy of Sciences of the United States of America 104, 1506915074.CrossRefGoogle Scholar
Janssen, S, Laermans, J, Iwakura, H, Tack, J and Depoortere, I 2012. Sensing of fatty acids for octanoylation of ghrelin involves a gustatory G-protein. PLoS ONE 7, e40168. doi: 10.1371/journal.pone.0040168.CrossRefGoogle ScholarPubMed
Jeon, T-I, Seo, YK and Osborne, TF 2011. Gut bitter taste receptor signalling induces ABCB1 through a mechanism involving CCK. Biochemical Journal 438, 3337.CrossRefGoogle ScholarPubMed
Keller, KL, Liang, LCH, Sakimura, J, May, D, van Belle, C, Breen, C, Driggin, E, Tepper, BJ, Lanzano, PC, Deng, L and Chung, WK 2012. Common variants in the CD36 gene are associated with oral fat perception, fat preferences, and obesity in African Americans. Obesity (Silver Spring) 20, 10661073.CrossRefGoogle ScholarPubMed
Kim, UK, Wooding, S, Riaz, N, Jorde, LB and Drayna, D 2006. Variation in the human TAS1R taste receptor genes. Chemical Senses 31, 599611.CrossRefGoogle ScholarPubMed
Lan, H, Vassileva, G, Corona, A, Liu, L, Baker, H, Golovko, A, Abbondanzo, SJ, Hu, W, Yang, S, Ning, Y, Del Vecchio, RA, Poulet, F, Laverty, M, Gustafson, EL, Hedrick, JA and Kowalski, TJ 2009. GPR119 is required for physiological regulation of glucagon-like peptide-1 secretion but not for metabolic homeostasis. Journal of Endocrinology 201, 219230.CrossRefGoogle Scholar
Lapis, TJ, Penner, MH and Lim, J 2014. Evidence that humans can taste glucose polymers. Chemical Senses 39, 737747.CrossRefGoogle ScholarPubMed
Larraufie, P, Dore, J, Lapaque, N and Blottiere, HM 2017. TLR ligands and butyrate increase Pyy expression through two distinct but inter-regulated pathways. Cellular Microbiology 19, e12648. doi: 10.1111/cmi.12648.CrossRefGoogle Scholar
Latorre, R, Huynh, J, Mazzoni, M, Gupta, A, Bonora, E, Clavenzani, P, Chang, L, Mayer, EA, De Giorgio, R and Sternini, C 2016. Expression of the bitter taste receptor, T2R38, in enteroendocrine cells of the colonic mucosa of overweight/obese vs. lean subjects. PLoS ONE 11, e0147468. doi: 10.1371/journal.pone.0147468.CrossRefGoogle ScholarPubMed
Lee, RJ, Xiong, G, Kofonow, JM, Chen, B, Lysenko, A, Jiang, P, Abraham, V, Doghramji, L, Adappa, ND, Palmer, JN, Kennedy, DW, Beauchamp, GK, Doulias, PT, Ischiropoulos, H, Kreindler, JL, Reed, DR and Cohen, NA 2012. T2R38 taste receptor polymorphisms underlie susceptibility to upper respiratory infection. The Journal of Clinical Investigation 122, 41454159.CrossRefGoogle ScholarPubMed
Lin, HV, Frassetto, A, Kowalik, EJ Jr., Nawrocki, AR, Lu, MM, Kosinski, JR, Hubert, JA, Szeto, D, Yao, X, Forrest, G and Marsh, DJ 2012. Butyrate and propionate protect against diet-induced obesity and regulate gut hormones via free fatty acid receptor 3-independent mechanisms. PLoS ONE 7, e35240. doi: 10.1371/journal.pone.0035240.CrossRefGoogle ScholarPubMed
Liou, AP, Sei, Y, Zhao, X, Feng, J, Lu, X, Thomas, C, Raybould, HE and Wank, SA 2011a. The extracellular calcium-sensing receptor is required for cholecystokinin secretion in response to L-phenylalanine in acutely isolated intestinal I cells. American Journal of Physiology Gastrointestinal and Liver Physiology 300, 538546.CrossRefGoogle ScholarPubMed
Liu, D, Costanzo, A, Evans, MDM, Archer, NS, Nowson, C, Duesing, K and Keast, R 2018. Expression of the candidate fat taste receptors in human fungiform papillae and the association with fat taste function. British Journal of Nutrition 120, 6473.CrossRefGoogle ScholarPubMed
Low, JY, Lacy, KE, McBride, RL and Keast, RSJ 2017. Carbohydrate taste sensitivity is associated with starch intake and waist circumference in adults. Journal of Nutrition 147, 22352242.CrossRefGoogle ScholarPubMed
Luo, XC, Chen, ZH, Xue, JB, Zhao, DX, Lu, C, Li, YH, Li, SM, Du, YW, Liu, Q, Wang, P, Liu, M and Huang, L 2019. Infection by the parasitic helminth Trichinella spiralis activates a Tas2r-mediated signaling pathway in intestinal tuft cells. Proceedings of the National Academy of Sciences of the United States of America 116, 55645569.CrossRefGoogle ScholarPubMed
Mace, OJ, Affleck, J, Patel, N and Kellett, GL 2007. Sweet taste receptors in rat small intestine stimulate glucose absorption through apical GLUT2. The Journal of Physiology 582, 379392.CrossRefGoogle ScholarPubMed
Margolskee, RF, Dyer, J, Kokrashvili, Z, Salmon, KSH, Ilegems, E, Daly, K, Maillet, EL, Ninomiya, Y, Mosinger, B and Shirazi-Beechey, SP 2007. T1R3 and gustducin in gut sense sugars to regulate exression of Na+-glucose cotransporter 1. Proceedings of the National Academy of Sciences of the United States of America 104, 1507515080.CrossRefGoogle Scholar
Mattes, RD 2011. Accumulating evidence supports a taste component for free fatty acids in humans. Physiology and Behavior 104, 624631.CrossRefGoogle ScholarPubMed
Meyerhof, W, Batram, C, Kuhn, C, Brockhoff, A, Chudoba, E, Bufe, B, Appendino, G and Behrens, M 2009. The molecular receptive ranges of human TAS2R bitter taste receptors. Chemical senses 35, 157170.CrossRefGoogle ScholarPubMed
Modvig, IM, Kuhre, RE and Holst, JJ 2019. Peptone‐mediated glucagon‐like peptide‐1 secretion depends on intestinal absorption and activation of basolaterally located calcium‐sensing receptors. Physiological Reports 7, e14056. doi: 10.14814/phy2.14056.CrossRefGoogle ScholarPubMed
Moran, AW, Al-Rammahi, MA, Arora, DK, Batchelor, DJ, Coulter, EA, Daly, K, Ionescu, C, Bravo, D, and Shirazi-Beechey, SP 2010a. Expression of Na+/glucose co-transporter 1 (SGLT1) is enhanced by supplementation of the diet of weaning piglets with artificial sweeteners. British Journal of Nutrition 104, 637646.CrossRefGoogle ScholarPubMed
Moran, AW, Al-Rammahi, MA, Arora, DK, Batchelor, DJ, Coulter, EA, Ionescu, C, Bravo, D and Shirazi-Beechey, SP, 2010b. Expression of Na+/glucose co-transporter 1 (SGLT1) in the intestine of piglets weaned to different concentrations of dietary carbohydrate. British Journal of Nutrition 104, 647655.CrossRefGoogle ScholarPubMed
Morrison, DJ and Preston, T 2016. Formation of short chain fatty acids by the gut microbiota and their impact on human metabolism. Gut Microbes 7, 189200.CrossRefGoogle ScholarPubMed
Moss, CE, Glass, LL, Diakogiannaki, E, Pais, R, Lenaghan, C, Smith, DM, Wedin, M, Bohlooly-Y, M, Gribble, FM and Reimann, F 2016. Lipid derivatives activate GPR119 and trigger GLP-1 secretion in primary murine L-cells. Peptides 77, 1620.CrossRefGoogle ScholarPubMed
Mrizak, I, Sery, O, Plesnik, J, Arfa, A, Fekih, M, Bouslema, A, Zaouali, M, Tabka, Z and Khan, NA 2015. The A allele of cluster of differentiation 36 (CD36) SNP 1761667 associates with decreased lipid taste perception in obese Tunisian women. British Journal of Nutrition 113, 13301337.CrossRefGoogle ScholarPubMed
Nadjsombati, MS, McGinty, JW, Lyons-Cohen, MR, Jaffe, JB, DiPeso, L, Schneider, C, Miller, CN, Pollack, JL, Nagana Gowda, GA, Fontana, MF, Erle, DJ, Anderson, MS, Locksley, RM, Raftery, D and von Moltke, J 2018. Detection of succinate by intestinal tuft cells triggers a type 2 innate immune circuit. Immunity 49, 3341.CrossRefGoogle ScholarPubMed
Nelson, G, Chandrashekar, J, Hoon, MA, Feng, LX, Zhao, G, Ryba, NJ and Zuker, CS 2002. An amino-acid taste receptor. Nature 416, 199202.CrossRefGoogle Scholar
Nelson, SL and Sanregret, JD 1997. Response of pigs to bitter-tasting compounds. Chemical Senses 22, 129132.CrossRefGoogle ScholarPubMed
Nøhr, MK, Pedersen, MH, Gille, A, Egerod, KL, Engelstoft, MS, Husted, AS, Sichlau, RM, Grundda, KVl, Poulsen, SS, Han, S, Jones, RM, Offermanns, S and Schwartz, TW 2013. GPR41/FFAR3 and GPR43/FFAR2 as cosensors for short-chain fatty acids in enteroendocrine cells vs FFAR3 in enteric neurons and FFAR2 in enteric leukocytes. Endocrinology 154, 35523564.CrossRefGoogle ScholarPubMed
Overton, HA, Fyfe, MCT and Reynet, C 2008. GPR119, a novel G protein-coupled receptor target for the treatment of type 2 diabetes and obesity. British Journal of Pharmacology 153, 7681.CrossRefGoogle Scholar
Park, J, Kim, KS, Kim, KH, Lee, IS, Jeong, HS, Kim, Y and Jang, HJ 2015. GLP-1 secretion is stimulated by 1,10-phenanthroline via colocalized T2R5 signal transduction in human enteroendocrine L cell. Biochemical and Biophysical Research Communications 468, 306311.CrossRefGoogle ScholarPubMed
Pepino, MY, Love-Gregory, L, Klein, S and Abumrad, NA 2012. The fatty acid translocase gene CD36 and lingual lipase influence oral sensitivity to fat in obese subjects. The Journal of Lipid Research 53, 561566.CrossRefGoogle Scholar
Pluznick, J 2014. A novel SCFA receptor, the microbiota, and blood pressure regulation. Gut Microbes 5, 202207.CrossRefGoogle ScholarPubMed
Poncet, N and Taylor, PM 2013. The role of amino acid transporters in nutrition. Current Opinion in Clinical Nutrition and Metabolic Care 16, 5765.CrossRefGoogle ScholarPubMed
Poreba, MA, Dong, CX, Li, SK, Stahl, A, Miner, JH and Brubaker, PL 2012. Role of fatty acid transport protein 4 in oleic acid-induced glucagon-like peptide-1 secretion from murine intestinal L cells. American Journal of Physiology Endocrinology and Metabolism 303, 899907.CrossRefGoogle ScholarPubMed
Precone, V, Beccari, T, Stuppia, I, Baglivo, M, Paolacci, S, Manara, E, Miggiano, GAD, Falsini, B, Trifirò, A, Zanlari, A, Herbst, KL, Unfer, V and Bertelli, M 2019. Taste, olfactory and texture related genes and food choices: implications on health status. European Review for Medical and Pharmacological Sciences 23, 13051321.Google ScholarPubMed
Raliou, M, Wiencis, A, Pillias, AM, Planchais, A, Eloit, C, Boucher, Y, Trotier, D, Montmayeur, JP and Faurion, A 2009. Nonsynonymous single nucleotide polymorphisms in human tas1r1, tas1r3, and mGluR1 and individual taste sensitivity to glutamate. American Journal of Clinical Nutrition 90, 789799.CrossRefGoogle Scholar
Ramos-Lopez, O, Panduro, A, Martinez-Lopez, E and Roman, S 2016. Sweet taste receptor TAS1R2 poly-morphism (Val191Val) is associated with a higher carbohydrate intake and hypertriglyceridemia among the population of West Mexico. Nutrients 8, 101.CrossRefGoogle Scholar
Reimann, F, Habib, AM, Tolhurst, G, Parker, HE, Rogers, GJ and Gribble, FM 2008. Glucose sensing in L cells: a primary cell study. Cell Metabolism 8:532539.CrossRefGoogle Scholar
Reimann, F, Ward, PS and Gribble, FM 2006. Signaling mechanisms underlying the release of glucagon-like peptide 1. Diabetes 55, 7885.CrossRefGoogle Scholar
Rettenberger, AT, Schulze, W, Breer, H and Haid, D 2015. Analysis of th eprotein related receptor GPR92 in G-cells. Frontiers in Physiology 6, 261. doi: 10.3389/fphys.2015.00261.CrossRefGoogle Scholar
Ribani, A, Bertolini, F, Schiavo, G, Scotti, E, Utzeri, VJ, Dall’Olio, S, Trevisi, P, Bosi, P and Fontanesi, L 2017. Next generation semiconductor based sequencing of bitter taste receptor genes in different pig populations and association analysis using a selective DNA pool-seq approach. Animal Genetics 48, 97102.CrossRefGoogle ScholarPubMed
Risso, DS, Mezzavilla, M, Pagani, L, Robino, A, Morini, G, Tofanelli, S, Carrai, M, Campa, D, Barale, R, Caradonna, F, Gasparini, P, Luiselli, D, Wooding, S and Drayna, D 2016. Global diversity in the TAS2R38 bitter taste receptor: revisiting a classic evolutionary proposal. Scientific Reports 6, 25506. doi: 10.1038/srep25506.CrossRefGoogle ScholarPubMed
Roura, E and Foster, S 2018. Nutrient-sensing biology in mammals and birds. Annual Reviews Animal Biosciences 6, 197225.CrossRefGoogle ScholarPubMed
Roura, E and Fu, M 2017. Taste receptors and feed intake in pigs (130 years of research: then, now and future). Animal Feed Science and Technology 233, 312.CrossRefGoogle Scholar
Roura, E, Humphrey, B, Klasing, KC and Swart, M 2011. Is the pig a good umami sensing model for humans? A comparative taste receptor study. Flavour and Fragrance Journal 26, 282285.CrossRefGoogle Scholar
Roura, E, Humphrey, B, Tedó, G and Ipharragerre, I 2008. Unfolding the codes of short-term feed appetence in farm and companion animals. A comparative oronasal nutrient sensing biology review. Canadian Journal of Animal Science 88, 535558.CrossRefGoogle Scholar
Roura, E, Koopmans, SJ, Lallès, JP, Le Huerou-Luron, I, de Jager, N, Schuurman, T and Val-Laillet, D 2016. Critical review evaluating the pig as a model for human nutritional physiology. Nutrition Research Reviews 29, 6090.CrossRefGoogle ScholarPubMed
Roura, E and Navarro, M 2018. Physiological and metabolic control of diet selection. Festschrift for Prof John L Black: celebrating 45 years of excellence in animal production science and application. Animal Production Science 58, 613626.CrossRefGoogle Scholar
Roura, E, Shrestha, B and Diffey, S 2013. Pigs show no sensory-motivated intake for several cereal and tuber starches except hydrolysed corn starch. In Proceedings of Manipulating Pig Production XIV, APSA Conference, 24–27 November 2013, Melbourne, Australia, pp. 45.Google Scholar
Rozengurt, E 2006. Taste receptors in the gastrointestinal tract. I. Bitter taste receptors and α-gustducin in the mammalian gut. American Journal of Physiology Gastrointestinal and Liver Physiology 291, 171177.CrossRefGoogle ScholarPubMed
Rozengurt, N, Wu, SV, Chen, MC, Huang, C, Sternini, C and Rozengurt, E 2006. Colocalization of the alpha-subunit of gustducin with PYY and GLP-1 in L cells of human colon. American Journal of Physiology, Gastrointestinal and Liver Physiology 291, 792802.CrossRefGoogle Scholar
Sandell, MA and Breslin, PAS 2006. Variability in a taste-receptor gene determines whether we taste toxins in food. Current Biology 16, 792794.CrossRefGoogle Scholar
San Gabriel, A and Uneyama, H 2013. AA sensing in the gastrointestinal tract. Amino Acids 45, 451461.CrossRefGoogle ScholarPubMed
Sarkadi, B, Homolya, L, Szakacs, G and Varadi, A 2006. Human multidrug resistance ABCB and ABCG transporters: participation in a chemoimmunity defense system. Physiological Reviews 86, 11791236.CrossRefGoogle Scholar
Schutz, B, Jurastow, I, Bader, S, Ringer, C, von Engelhardt, J, Chubanov, V, Gudermann, T, Diener, M, Kunner, W, Krasteva-Christ, G and Weihe, E 2015. Chemical coding and chemosensory properties of cholinergic brush cells in the mouse gastrointestinal and biliary tract. Frontiers in Physiology 6, 87. doi: 10.3389/fphys.2015.00087.Google ScholarPubMed
Sclafani, A 1987. Carbohydrate taste, appetite, and obesity: an overview. Neuroscience and Biobehavioral Reviews 11, 131153.CrossRefGoogle ScholarPubMed
Soares, S, Silva, MS, Garcia-Estevez, I, GroBmann, P, Bras, N, Brandao, E, Mateus, N, de Freitas, V, Behrens, M and Meyerfof, W 2018. Human bitter taste receptors are activated by different classes of polyphenols. Journal of Agricultural and Food Chemistry 66, 88148823.CrossRefGoogle ScholarPubMed
Steensels, S, Cools, L, Avau, B, Vancleef, L, Farre, R, Verbeke, K and Depoortere, I 2016. Supplementation of oligofructose, but not sucralose, decreases high-fat diet induced body weight gain in mice independent of gustducin-mediated gut hormone release. Molecular Nutrition and Food Research 61, 1600716. doi: 10.1002/mnfr.201600716 CrossRefGoogle Scholar
Steensels, S and Depoortere, I 2018. Chemoreceptors in the gut. Annual Review of Physiology 80, 117141.CrossRefGoogle Scholar
Steinert, RE, Gerspach, AC, Gutmann, H, Asarian, L, Drewe, J and Beglinger, C 2011a. The functional involvement of gut-expressed sweet taste receptors in glucose-stimulated secretion of glucagon-like peptide-1 (GLP-1) and peptide YY (PYY). Clinical Nutrition 30, 524532.CrossRefGoogle Scholar
Steinert, RE, Frey, F, Topfer, A, Drewe, J and Beglinger, C 2011b. Effects of carbohydrate sugars and artificial sweeteners on appetite and the secretion of gastrointestinal satiety peptides. British Journal of Nutrition 105, 13201328.CrossRefGoogle Scholar
Stevens, C.E 1988. Comparative physiology of the vertebrate digestive system. Cambridge University Press, Cambridge, NY, USA.Google Scholar
Stewart, JE, Newman, LP and Keast, RSJ 2011. Oral sensitivity to oleic acid is associated with fat intake and body mass index. Clinical Nutrition 30, 838844.CrossRefGoogle ScholarPubMed
Sukumaran, SK, Yee, KK, Iwata, S, Kotha, R, Quezada-Calvillo, R, Nichols, BL, Mohan, S, Pinto, BM, Shigemura, N and Ninomiya, Y 2016. Taste cell-expressed α-glucosidase enzymes contribute to gustatory responses to disaccharides. Proceedings of National Academy of Science 113, 60356040.CrossRefGoogle ScholarPubMed
Sundaresan, S, Shahid, R, Riehl, TE, Chandra, R, Nassir, F, Stenson, WF, Liddle, RA and Abumrad, NA 2013. CD36-dependent signaling mediates fatty acid-induced gut release of secretin and cholecystokinin. The Federation of American Societies for Experimental Biology Journal 27, 11911202.CrossRefGoogle ScholarPubMed
Sung, MM, Kim, TT, Denou, E, Soltys, CM, Hamza, SM, Byme, NJ, Masson, G, Park, H, Wishart, DS, Madsen, KL, Shertzer, JD and Dyck, JR 2017. Improved glucose homeostasis in obese mice treated with resveratrol is associated with alterations in the gut microbiome. Diabetes 66, 418425.CrossRefGoogle ScholarPubMed
Swartz, TD, Duca, FA, de Wouters, T, Sakar, Y and Covasa, M 2012. Up-regulation of intestinal type 1 taste receptor 3 and sodium glucose luminal transporter-1 expression and increased sucrose intake in mice lacking gut microbiota. British Journal of Nutrition 107, 621630.CrossRefGoogle ScholarPubMed
Tedo Perez, MG 2009. The umami taste in pigs: L-amino acid preferences and in vitro recognition by the receptor dimer pT1r1/pT1r3 expressed in porcine taster and non-taste tissues. PhD Thesis, The Autonomous University of Barcelona, Barcelona, Spain.Google Scholar
Tepper, BJ, Koelliker, Y, Zhao, L, Ullrich, NV, Lanzara, C, D’Adam, P, Ferrara, A, Ulivi, S, Esposito, L and Gasparini, P 2008. Variation in the bitter-taste receptor gene TAS2R38, and adiposity in a genetically isolated population in Southern Italy. Obesity 16, 22892295.CrossRefGoogle Scholar
Tinti, JM, Glaser, D, Wanner, M and Nofre, C 2000. Comparison of gustatory responses to AA s in pigs and in humans. Lebensmittel-Wissenschaft and Technologie 33, 578583.CrossRefGoogle Scholar
Toguri, C 2008. Genetic Variation in CD36 and Dietary Fat Intake. M.Sc. Dissertation, University of Toronto, Toronto, Canada.Google Scholar
Valente, C, Alvarez, L, Marques, PI, Gusmão, L, Amorim, A, Seixas, S and Prata, MJ 2018. Genes from the TAS1R and TAS2R families of taste receptors: looking for signatures of their adaptive role in human evolution. Genome Biology and Evolution 10, 1139–1115.CrossRefGoogle ScholarPubMed
Vancleef, L, Van Den Broeck, T, Thijs, T, Steensels, S, Briand, L, Tack, J and Depoortere, I 2015. Chemosensory signalling pathways involved in sensing of AA s by the ghrelin cell. Scientific Reports 5, 15725. doi: 10.1038/srep15725.CrossRefGoogle Scholar
van der Wielen, N, van Avesaat, M, de Wit, NJ, Vogels, JT, Troost, F, Masclee, A, Koopmans, SJ, van der Meulen, J, Boekschoten, MV, Muller, M, Hendriks, HF, Witkamp, RF and Meijerink, J 2014. Cross-species comparison of genes related to nutrient sensing mechanisms expressed along the intestine. PLoS ONE 9, e107531. doi: 10.1371/journal.pone.0107531.CrossRefGoogle ScholarPubMed
Vigors, S, Sweeney, T, O’Shea, CJ, Browne, JA, and O’Doherty, JV 2014. Improvements in growth performance, bone mineral status and nutrient digestibility in pigs following the dietary inclusion of phytase are accompanied by modifications in intestinal nutrient transporter gene expression. British Journal of Nutrition 112, 688697.CrossRefGoogle ScholarPubMed
von Moltke, J, Ji, M, Liang, HE and Locksley, RM 2016. Tuft-cell-derived IL-25 regulates an intestinal ILC2-epithelial response circuit. Nature 529, 221225.CrossRefGoogle ScholarPubMed
Wang, J, Fu, M, Navarro, M and Roura, E 2017. A double-choice model to quantify negative preference to bitterness in pigs. Animal Production Science 57, 24222422.CrossRefGoogle Scholar
Wang, J, Wu, X, Simonavicius, N, Tian, H and Ling, L 2006. Medium-chain fatty acids as ligands for orphan G protein-coupled receptor GPR84. The Journal of Biological Chemistry 281, 3445734464.CrossRefGoogle Scholar
Wang, Q, Liszt, KI, Deloose, E, Canovai, E, Thijs, T, Farre, R, Ceulemans, LJ, Lannoo, M, Tack, J and Depoortere, I 2019. Obesity alters adrenergic and chemosensory signaling pathways that regulate ghrelin secretion in the human gut. The FASEB Journal 33, 49074920.CrossRefGoogle ScholarPubMed
Weatherburn, DH 2015. Role of short chain fatty acid receptors in the gastrointestinal tract and their potential involvement in appetite control. PhD Thesis, University of Liverpool, Liverpool, Great Britain.Google Scholar
Wellendorph, P, Johansen, LD and Brauner-Osborne, H 2010. Molecular pharmacology of promiscuous seven transmembrane receptors sensing organic nutrients. Molecular Pharmacology 76, 453465.CrossRefGoogle Scholar
Widmayer, P, Kusumakshi, S, Hägele, FA, Boehm, U and Breer, H 2017. Expression of the fatty acid receptors GPR84 and GPR120 and cytodifferentiation of epithelial cells in the gastric mucosa of mouse pups in the course of dietary transition. Frontiers in Physiology 8, 601. doi: 10.3389/fphys.2017.00601 CrossRefGoogle ScholarPubMed
Young, SH, Rey, O, Sternini, C and Rozengurt, E 2010. Amino acid sensing by enteroendocrine STC-1 cells: role of the Na+-coupled neutral amino acid transporter 2. American Journal of Physiology. Cell Physiology 298, 14011413.CrossRefGoogle ScholarPubMed
Zhang, C, Huang, Y, Jiang, Y, Mulpuri, N, Wei, L, Hamelberg, D, Brown, EM and Yang, JJ 2014. Identification of an l-phenylalanine binding site enhancing the cooperative responses of the calcium-sensing receptor to calcium. Journal of Biological Chemistry 289, 52965309.CrossRefGoogle Scholar
Zhang, G, Hasek, LY, Lee, BH and Hamaker, BR 2015. Gut feedback mechanisms and food intake: a physiological approach to slow carbohydrate bioavailability. Food and Function 6, 10721089.CrossRefGoogle ScholarPubMed
Zhao, A, Urban, JF, Anthony, RM, Sun, R, Stiltz, J, van Rooijen, N, Wynn, TA, Gause, WC, Shea-Donohue, T 2008. Th2 cytokine-induced alterations in intestinal smooth muscle function depend on alternatively activated macrophages. Gastroenterology 135, 21725.e1. doi: 10.1053/j.gastro.2008.03.077.CrossRefGoogle ScholarPubMed
Figure 0

Table 1. Main simple carbohydrates receptors and transporters known to be involved in GIT sensing in humans and pigs1

Figure 1

Table 2. Main AA receptors and transceptors known to be involved in GIT sensing in humans and pigs1

Figure 2

Table 3. Main FFA receptors and transceptors known to be involved in GIT sensing in humans and pigs1

Supplementary material: File

Roura et al. supplementary material

Roura et al. supplementary material 1

Download Roura et al. supplementary material(File)
File 34.3 KB