Sie sind auf Seite 1von 13

Downloaded from http://gut.bmj.com/ on April 8, 2016 - Published by group.bmj.

com
Gut Online First, published on October 16, 2013 as 10.1136/gutjnl-2013-305112
Recent advances in basic science

Taste receptors of the gut: emerging roles in health


and disease
Inge Depoortere

Correspondence to ABSTRACT
Professor Inge Depoortere, Gut Recent progress in unravelling the nutrient-sensing Key messages
Peptide Research Lab,
Translational Research Center
mechanisms in the taste buds of the tongue has triggered
for Gastrointestinal Disorders, studies on the existence and role of chemosensory cells in
▸ The gut tastes what we eat—bitter, sweet,
University of Leuven, the gut. Indeed, the gastrointestinal tract is the key
Gasthuisberg O&N1, box 701, umami, fat—in much the same way as the
interface between food and the human body and can
Leuven 3000, Belgium; inge. tongue through the use of similar taste
sense basic tastes in much the same way as the tongue,
depoortere@med.kuleuven.be receptors and chemosensory signalling
through the use of similar G-protein-coupled taste
pathways
Received 29 July 2013 receptors. These receptors ‘taste’ the luminal content and
Revised 16 September 2013
▸ In health, taste receptors sense nutrients from
transmit signals that regulate nutrient transporter
Accepted 25 September 2013 either a luminal or blood-borne direction and
expression and nutrient uptake, and also the release of
transmit signals that control the secretion of
gut hormones and neurotransmitters involved in the
gut hormones and the expression of nutrient
regulation of energy and glucose homeostasis. Hence,
transporters to maintain energy and glucose
they play a prominent role in the communication between
homeostasis and gastrointestinal function
the lumen, epithelium, smooth muscle cells, afferent
▸ In disease, disturbances or adaptations in the
nerve fibres and the brain to trigger adaptive responses
expression or sensitivity of these taste receptors
that affect gastrointestinal function, food intake and
and their signalling pathways may affect
glucose metabolism. This review summarises how sensing
digestive behaviour and metabolism
of nutrients by taste receptors along the gut plays a key
▸ This is a new and emerging field, future studies
role in the process of digestion, and how disturbances or
aimed at a better understanding of how the
adaptations of these chemosensory signalling pathways
sensing of nutrients in the gut is finely tuned
may contribute to the induction or resolution of a number
by different taste receptors in health and
of pathological conditions related to diabetes, obesity, or
disease, should help us to define novel drug
diet-induced symptom generation in irritable bowel
targets
syndrome. Targeting these receptors may represent a
promising novel route for the treatment of a number of
these diseases.

TASTE RECEPTORS AND GUSTATORY


INTRODUCTION FUNCTION
We choose to eat for many reasons, including to The taste system detects compounds that elicit at
satisfy our hunger, to invite pleasant sensory least five perceptual qualities: sweet, umami, bitter,
experiences, and as a social ritual. However, we sour and salty. The latter two are largely mediated
need to eat to acquire nutrients essential for life by ion channels, and will not be discussed further
and health. Not surprisingly, the digestive system here. By contrast, sweet, umami and bitter-tasting
contains a diverse array of detectors that help to stimuli are detected by members of two GPCR fam-
regulate ingestive decisions, impact nutrient assimi- ilies, the taste 1 receptor family (TAS1R) and the
lation, avoid or neutralise toxins, and elicit taste 2 receptor family (TAS2R), that are expressed
complex neural and endocrine responses that affect in subpopulations of taste bud cells (figure 1).
metabolism, gastrointestinal (GI) transit, satiation Subtypes of the TAS1R family heterodimerise to
and satiety. Strikingly, many of the G protein- detect sweet (TAS1R2-TAS13) and umami
coupled receptors (GPCRs) that detect nutrients (TAS1R1-TAS1R3).2 3 Animals that lack these subu-
and toxins in the oral cavity and function as taste nits are deficient in their ability to taste umami and/
receptors there, also subserve important functions or sweet stimuli.4 The presence of multiple binding
throughout the GI tract. Activation of these recep- sites on these receptors can explain the synergistic
tors triggers the release of neurotransmitters (eg, effects of 50 -ribonucleotides and glutamate in
ATP) that will excite primary sensory afferent fibres umami taste, or the ability of many chemically
and interact with neighbouring presynaptic cells to diverse compounds–including sugars, non-caloric
relay information to the hindbrain.1 A similar sweeteners, D-amino acids and some proteins–to
system of highly orchestrated interactions also oper- elicit a sweet taste. Consistent with their proposed
ates in the gut and further points to the functional evolutionary need to detect foods rich in nutrients,
similarity of lingual and intestinal cells. I will the umami and sweet receptors are low-affinity
To cite: Depoortere I. Gut
Published Online First: review the state of our knowledge about taste receptors, with EC50s in the millimolar range for
[please include Day Month receptor function along the entirety of the alimen- their most common natural ligands.
Year] doi:10.1136/gutjnl- tary canal and discuss the implications of these Alternatively, we are quite sensitive to bitter-
2013-305112 functions for human physiology and disease. tasting compounds. While this is important for

Copyright
Depoortere I.Article author
Gut 2013;0:1–12. (or their employer) 2013. Produced by BMJ Publishing Group Ltd (& BSG) under licence.
doi:10.1136/gutjnl-2013-305112 1
Downloaded from http://gut.bmj.com/ on April 8, 2016 - Published by group.bmj.com

Recent advances in basic science

Figure 1 Simplified model of the taste GPCR signalling pathways involved in chemosensing by taste cells of the tongue. Subtypes of the TAS1R
family heterodimerize to detect sweet (TAS1R2-TAS13) and umami (TAS1R1-TAS1R3) while bitter is detected by 25 subtypes of the TAS2R family.
Medium-chain and long-chain fatty acids are detected by FFAR1 and GPR120. Taste receptor binding leads to activation of gustatory G-proteins,
release of intracellular Ca2+, activation of TRPM5, depolarisation, activation of voltage-gated Na+ channels (VGNC) and release of ATP which
activates purinergic receptors on afferent nerve fibres leading to taste perception. ATP, adenosine triphosphate; FFAR1, free fatty acid receptor 1;
GPCR, G-protein coupled receptor; GPR120, G-protein coupled receptor 120; PX-1, pannexin 1-hemichannel; TAS1R, taste receptor type 1; TAS1R1,
taste receptor type 1 member 1; TAS1R2, taste receptor type 1 member 2; TAS1R3, taste receptor type 1 member 3; TAS2R, taste receptor type 2;
TRPM5, transient receptor potential cation channel M5; VGNC, voltage-gated Na+ channel.

avoiding potential toxins, it also reduces the oral tolerance for


many common pharmaceuticals.5 Bitter tastants are recognised Table 1 Overview of the different taste receptors involved in
by TAS2Rs.6 The human genome encodes 25 different TAS2Rs, nutrient sensing
which fall into three functional categories: specialists that
Sweet taste receptors/transporters
respond to one or two compounds; generalists that are highly
TAS1R2-TAS1R3 Taste receptor, type 1, member 2-taste
promiscuous in their stimulus responses; and those that are receptor, type 1, member 3
intermediate in their selectivity.7 Many compounds can activate SGLT1 Sodium-dependent glucose transporter 1
more than one receptor, and activation thresholds can vary GLUT Glucose transporter
from millimolar to nanomolar levels. TAS2R polymorphisms are Amino acid/peptide taste receptors
quite common in human populations, and can have pronounced TAS1R1-TAS1R3 (umami, Taste receptor, type 1, member 1-taste
effects on the ability of individuals to detect certain bitter aliphatic amino acids) receptor, type 1, member 3
compounds.8 CaSR (aromatic amino acids) Calcium sensing receptor
Other GPCRs have been implicated in the detection of gusta- GPRC6A (basic and small G-protein coupled receptor family C group 6
tory stimuli. For example, metabotropic glutamate receptors, neutral amino acids) member A
specifically mGluR1 and mGluR4, may contribute to amino mGluR (umami) Metabotropic glutamate receptor
acid taste.9 The Ca2+-sensing receptor (CaSR) and GPRC6A GPR92 (protein hydrolysates) G-protein coupled receptor 92
have been proposed to mediate Ca2+ taste and also amino acid Bitter taste receptors
taste in extraoral tissues such as the gut.10 11 Two receptors for TAS2R Taste receptor type 2
long-chain fatty acids (LCFAs), FFAR1 (a.k.a., GPR40) and Free fatty acid receptors
GPR120 have been localised to taste cells and contribute to oro- Long-chain/medium-chain fatty acids
sensory responses to fats (whether or not fat can be considered GPR120 G-protein coupled receptor 120
as the sixth taste is the subject of ongoing investigation).12 FFAR1 (also known as GPR40) Free fatty acid receptor 1
Additionally, receptors for short-chain fatty acids (SCFA), Short-chain fatty acids
FFAR2 and FFAR3, have been detected outside the oral cavity.11 FFAR2 (also known as GPR43) Free fatty acid receptor 2
An as yet unidentified receptor may mediate the taste of polysac- FFAR3 (also known as GPR41) Free fatty acid receptor 3
charides. An overview of the different taste receptors involved CaSR, calcium sensing receptor; FFAR1, free fatty acid receptor 1; FFAR2, free fatty
in nutrient sensing is represented in table 1. acid receptor 2, FFAR3, free fatty acid receptor 3; GLUT, glucose transporter; GPR92,
G-protein coupled receptor 92; GPR120, G-protein coupled receptor 92; GPRC6A,
TAS1Rs and TAS2Rs are found in Type II taste cells, a mor- G-protein coupled receptor family C group 6; mGluR, metabotropic glutamate
phological class of microvillar taste bud cells. Activation of a receptor; SGLT1, sodium-dependent glucose transporter 1; TAS1R1, taste receptor
gustatory G-protein (eg, gustducin) coupled to these GPCRs type 1 member 1; TAS1R2, taste receptor type 1 member 2; TAS1R3, taste receptor
type 1 member 3; TAS2R, taste receptor type 2.
results in the activation of phospholipase C β2 leading to

2 Depoortere I. Gut 2013;0:1–12. doi:10.1136/gutjnl-2013-305112


Downloaded from http://gut.bmj.com/ on April 8, 2016 - Published by group.bmj.com

Recent advances in basic science

inositoltriphosphate-mediated release of intracellular Ca2+ and expressed on taste cells may be targets of circulating peptides
activation of the transient receptor potential cation channel M5 produced in the gut, adipose tissue, or other tissues. For
(TRPM5) (figure 1). These events induce membrane depolarisa- example, systemic leptin has been shown to decrease sweet taste
tion, action potential generation, and the release of ATP, which responsiveness through actions on leptin receptors present on a
acts on purinergic receptors to activate gustatory afferents subset of taste cells.16 Thus, postingestive nutrient responses
leading to activation of brain centres involved in taste percep- could feed back onto the peripheral gustatory apparatus to
tion. Sweet, umami and bitter taste receptors are differentially modulate its responsiveness to subsequent foods.
expressed within the Type II taste cells.13 This segregation is a
key component of taste quality coding and hedonic discrimin- Nutrient sensing in the stomach
ation in the gustatory periphery. The extent to which these Proximal stomach
various taste transduction components are colocalised in gut Upon ingestion of food, the proximal stomach relaxes to accom-
cells is unclear, but surely has consequences for how they modate large amounts of food without increasing intragastric
impact GI function. pressure. Tension-sensitive mechanoreceptors are activated by
the arrival of food and relay their information via the vagus to
ROLE OF TASTE RECEPTORS IN NUTRIENT DETECTION AND the hindbrain to induce sensations of satiation.17 Nutrient
RESPONSE DURING DIGESTION sensing in the stomach has been considered to be a less import-
‘Gut’ peptides produced in taste cells of the tongue ant factor, but the role of chemosensory receptors should not be
Taste cells also express a number of bioactive peptides best ignored. Indeed, recent studies showed that intragastric adminis-
known for their roles in metabolism, feeding and satiety, includ- tration of the bitter agonist, denatonium benzoate (DB), inhib-
ing: glucagon-like peptide-1 (GLP-1), glucagon, neuropeptide Y, ited gastric accommodation to nutrient drink infusion and
peptide YY (PYY), cholecystokinin (CCK), vasoactive intestinal tended to increase satiation scores.18 The contractile response
peptide and ghrelin.14 The functions of these peptides in taste induced by DB was mimicked in vitro in smooth muscle strips
buds are not fully understood, but at least some may act to from the mouse fundus and was partially mediated via the gusta-
modulate the responsiveness of the peripheral gustatory appar- tory G-protein subunit, α-gustducin, and involved the release of
atus to certain taste stimuli. Interestingly, stimuli representing Ca2+ from intracellular and extracellular stores.19
different taste qualities promote the release of different peptide The proximal stomach is also the major site of production of
combinations, suggesting that these peptides could also play a the orexigenic hormone, ghrelin, which is considered as a physio-
role in quality coding.15 logical meal initiator.20 How the reduction of plasma ghrelin
Cognate receptors for all the peptides expressed in taste cells during a meal might participate in the induction of satiation is
can be found on taste cells or on the adjacent afferent nerve incompletely explored. The fact that ghrelin levels are suppressed
fibres.14 Therefore, these peptides almost certainly have auto- strongly by ingested proteins, weakly by proteins and biphasically
crine and paracrine functions within the taste bud and, in some by carbohydrates, suggests that the ghrelin P/D1 cells contain the
cases, could function as cotransmitters with ATP. However, it is machinery to sense nutrients.21 Recent immunofluorescence
unclear whether taste bud peptides also reach the bloodstream studies confirmed that the ghrelin cell is colocalised with the gus-
and exert endocrine effects. Furthermore, peptide receptors tatory G-proteins, α-gustducin and α-transducin, the sweet and

Figure 2 Schematic overview of the expression of taste receptors in different type of endocrine cells along the gut that control the release of
hormones in response to nutrients. CaSR, calcium sensing receptor; FFAR1, free fatty acid receptor 1; FFAR2, free fatty acid receptor 2; FFAR3, fatty
acid receptor 3; GPR92, G-protein coupled receptor 92; GPRC6A, G-protein coupled receptor family C group 6 member A; LCFA, long-chain fatty
acids; TAS1R1, taste receptor type 1 member 1; TAS1R2, taste receptor type 1 member 2; TAS1R3, taste receptor type 1 member 3; TAS2R, taste
receptor type 2.

Depoortere I. Gut 2013;0:1–12. doi:10.1136/gutjnl-2013-305112 3


Downloaded from http://gut.bmj.com/ on April 8, 2016 - Published by group.bmj.com

Recent advances in basic science

umami receptor subunit, TAS1R3, and the free fatty acid-sensing Whether any stimulus that suppresses or induces gastric phase 3
receptor, GPR120.22–24 It remains to be investigated whether the activity inhibits or stimulates hunger, respectively, requires
postprandial suppression of ghrelin by nutrients is indeed further investigation. Furthermore, recent studies showed that
mediated via these receptors. However, the first functional gastric phase 3 activity was less frequently observed in obese
studies showed that α-gustducin is involved in the effect of bitter patients, and was associated with fewer hunger peaks suggesting
compounds on ghrelin release and in sensing of medium-chain that the absence of generation of gastric MMCs may be
fatty acids in the diet necessary for the octanoylation of pro-anorexigenic and may also represent a compensatory mech-
ghrelin.22 23 Additionally, GPR120 seems to play a role in the anism to reduce hunger feelings in obesity.33 The motilin
lipid-sensing cascade of the ghrelin cell.23 The distribution of agonist, erythromycin, known to induce gastric phase 3,
taste receptors involved in nutrient sensing in endocrine cells restored the association between gastric phase 3 and hunger
along the gut is summarised in figure 2. peaks in obese patients.

Distal stomach Nutrient sensing in the small intestine


The arrival of food in the stomach stimulates the gastric phase The small intestine is the major site of digestion and absorption
of acid, pepsinogen and mucus production. The two principal in the GI tract. In the small intestine, carbohydrates, fats and
triggers are distension of the stomach, which initiates local protein digestion products, as well as osmolarity changes and
myenteric and vago-vagal reflexes, and the chemical content of physical distention activate inhibitory neural and endocrine
the food. In the stomach, sensing of protein breakdown pro- pathways that signal the stomach to delay emptying and to help
ducts is important as they activate the release of two important limit ingestion by enhancing gastric mechanoreceptor stimula-
regulators of pepsinogen and acid secretion: gastrin (G-cells) tion. However, experiments demonstrated that intestinal nutri-
and somatostatin (D-cells). Indeed, peptone has been shown to ent infusions inhibit food intake during sham feeding when
affect gastrin and somatostatin secretion in rats.25 Recent studies gastric contents were drained via an open gastric cannula to
showed that mouse and pig antral G-cells and a subpopulation obviate the gastric filling effects on food intake.34 Thus, while
of D-cells express GPR92, a receptor activated by dietary intestinal signals and gastric emptying interact to control food
protein hydrolysates.26 In both endocrine cell types, GPR92 is intake, a delay of gastric emptying is not required for intestinal
coupled to different signalling pathways. It is therefore conceiv- signals to elicit satiation. The small intestine also sends endo-
able that GPR92 may play an important role in adjusting the crine satiety signals to the hypothalamus, either directly, via the
release of gastrin and somatostatin in response to protein digests bloodstream and across the incomplete blood-brain barrier to
in the chyme. In addition, G and D-cells express the amino acid the arcuate nucleus, or indirectly, through the activation of the
taste receptor GPRC6A and the CaSR that act in concert with vagus nerve. It is clear that the vagal-brainstem-hypothalamic
each other to sense a broad spectrum of amino acids ranging pathway plays a major role in the effect of the satiation hor-
from basic and small neutral (GPRC6A) to aromatic (L-Phe) mones, CCK and GLP-1, on food intake. Their receptors are
amino acids and Ca2+.27 28 The stimulation of gastrin after expressed on vagal afferents, they increase vagal afferent firing
gavage with L-Phe, peptone, Ca2+ and a neutralising buffer was and induce cfos expression in the nucleus of the solitary tract
abolished in CaSR−/− mice suggesting a regulatory role for this after intraperitoneal injection.35–39 Furthermore the inhibition
receptor in gastrin release.29 The TAS1R family is not important of food intake by exogenous GLP-1 and CCK is blocked by
for G-cell sensing of protein and amino acids since TAS1R3 is vagotomy and brainstem-hypothalamic pathway
not expressed on G-cells and the gastrin response to peptone is transectioning.40 41
not abolished in TAS1R1/3−/− mice.28 29 Additionally, L-Phe can
induce acid secretion independent of hormonal stimulation via Duodenum
activation of the CaSR on parietal cells.30 In the duodenum, fat, protein hydrolysates and amino acids
In the distal stomach, the chyme is mixed with the digestive stimulate the secretion of CCK from I-cells. This hormone then
secretions and ground by powerful peristaltic contractions. The acts to slow gastric emptying and increase satiety. In STC-1
process of emptying starts and the rate is dependent on the cells, a mouse enteroendocrine-like cell line, LCFAs induce
gastric volume and the chemical nature of the gastric contents. CCK secretion through the lipid sensor GPR120 and the trans-
The major control mechanism for gastric emptying involves duction channel TRPM5.42 43 However, cell lines are incom-
duodenal gastric feedback and the emphasis shifts towards pletely validated as accurate models of native I-cells.
satiety mechanisms. Establishing cultures from endocrine cells remained a challenge
When the meal is emptied from the stomach, the upper GI for many years but the generation of transgenic mice with I cell-
motility pattern changes from a digestive to an interdigestive specific expression of a fluorescent protein, allowed the isolation
pattern: the migrating motor complex, characterised by a group of a pure I-cell population by fluorescence-activated cell sorting.
of strong phasic contractions migrating distally. Phase 3 contrac- Native I-cells are also immunoreactive for FFAR1, and the effect
tions, the most vigorous contractions of the migrating motor of linolenic acid on the secretion of CCK from isolated I-cells
complex (MMC), originating in the stomach, are considered as was abolished in cells from FFAR1−/− mice.44 45 Thus, GPR120
a hunger signal. Arrival of nutrients will disrupt this pattern. and FFAR1 appear to mediate lipid-induced CCK secretion.
Infusion of high doses of amino acids in healthy volunteers Amino acids and protein hydrolysates similarly appear to use
shortens the duration of the MMC length and suppresses antral multiple receptor types to stimulate CCK secretion. In humans,
phase 3 activity.31 Intragastric administration of the bitter com- administration of L-Phe increased CCK secretion and reduced
pound, DB, in the fasted state was shown to decrease antral but food intake.46 Two amino acid-responsive receptors,
not duodenal motility and to shift the origin of phase 3 from TAS1R1-TAS1R3 and the CaSR, are expressed in I-cells, and
the antrum to the duodenum. This was accompanied by a sig- implicated in CCK release.47 For example, Phe-dependent,
nificant decrease in hunger scores.32 In both cases, the mechan- Leu-dependent and Glu-dependent CCK release is dependent
isms involved are unknown but may involve activation of taste on TAS1R1-TAS1R3, while the CaSR mediates Phe-induced and
receptors on endocrine cells, smooth muscle cells and/or nerves. Trp-induced responses in mouse intestinal tissue explants.47

4 Depoortere I. Gut 2013;0:1–12. doi:10.1136/gutjnl-2013-305112


Downloaded from http://gut.bmj.com/ on April 8, 2016 - Published by group.bmj.com

Recent advances in basic science

Studies in vitro (STC-1 cells) and in native I-cells also showed secretion from mouse jejunal and ileal explants, and/or mouse
an important role for the CaSR in the effects of aromatic amino (GLUTag) and human (NCI-H716) enteroendocrine cell
acids on CCK secretion.48–50 Protein hydrolysates stimulate the lines.56 58 59 Third, glucose-stimulated GLP-1 secretion is nearly
release of CCK via GPR92 in STC-cells.51 or fully abolished in α-gustducin−/− and TAS1R2−/− and
Bitter tastants can also induce CCK release from STC-1 cells TAS1R3−/− mice.56 58 Fourth, sweet taste receptor inhibitors, or
by affecting Ca2+ influx.52 53 RNA silencing of α-gustducin, reduce sucralose-stimulated
Glucose-induced CCK secretion in humans, although perhaps GLP-1 secretion from GLUTag and NCI-H716 cells.56 59
not a major phenomenon, was unaffected by the sweet taste Finally, the sweet taste receptor inhibitor, lactisole, reduced sys-
receptor inhibitor lactisole.54 Immunolocalisation studies con- temic levels of GLP-1 and PYY after intragastric or intraduode-
firmed that the sweet taste receptor-specific subunit, TAS1R2, is nal administration of glucose in humans.54 55
not coexpressed with CCK.47 Several studies also indicated an important role for the
Na+-glucose cotransporter SGLT1 and the ATP-sensitive K+
Jejunum-ileum (KATP) channel. For example, the KATP channel blocker tolbuta-
GLP-1 and glucose-dependent insulinotropic polypeptide (GIP) mide enhances GLP-1 secretion from GLUTag cells or from iso-
are incretin hormones that augment insulin secretion after inges- lated L-cells from upper small intestine or colon.60 61
tion of a meal. They are secreted from L-cells and K-cells, Furthermore, immunoreactivity for one KATP channel subunit,
respectively. The mechanisms by which L-cells couple glucose Kir6.2, has been localised to L-cells.62 By contrast, a related
detection to GLP-1 secretion have been controversial, but some KATP channel blocker, glibenclamide, had no effect on GLP-1
clarity has begun to emerge (figure 3). Several lines of evidence secretion from mouse ileum explants, but did induce GLP-1
support that the sweet taste receptor, TAS1R2-TAS1R3, and the secretion from colonic explants, suggesting that this channel is
taste G-protein, α-gustducin, are required for glucose-stimulated involved in GLP-1 secretion from L-cells of the large, but not
GLP-1 secretion from the small intestine. First, these three pro- small intestine.58 The evidence for a role of SGLT1 is more
teins are present in human and rodent L-cells of the small intes- intriguing, as SGLT1−/− mice exhibit reduced GLP-1 and GIP
tine.55–57 Second, several sweet taste receptor agonists — levels after glucose gavage.63 A role for SGLT1 in
sucrose, glucose, fructose and sucralose — can elicit GLP-1 glucose-induced GLP-1 release has been confirmed in GLUTag
cells and primary L-cells.61 64 65
A physiological relationship exists between L-cells and entero-
cytes. SGLT1 and another glucose transporter, GLUT2, are
rapidly upregulated in enterocytes in response to the presence
of dietary sugars or sweeteners. However, their expression and/
or translocation is downregulated in TAS1R3−/− and
α-gustducin−/− mice, suggesting a strong correlation between
taste receptor activation in L-cells and the modulation of
glucose transporter expression in enterocytes, likely through the
actions of GLP-2.59 66 67 Cats which cannot taste sugars do not
upregulate SGLT1 expression in response to increased dietary
carbohydrate levels.68 69
It is unclear whether FFAR1 or GPR120 play the more
important role in L-cell and K-cell fatty acid sensing. Both
receptors are expressed in L-cells.44 70 Oral administration of
α-linolenic acid promoted GLP-1 secretion in vivo.70 In STC-1
cells, however, siRNA against GPR120, but not FFAR1,
impaired α-linolenic acid-induced GLP-1 secretion.
Nevertheless, the secretion of GIP and GLP-1 in response to
acute, oral fat diet administration was reduced in FFAR1−/−
mice and was associated with a concomitant reduction in insulin
secretion and glucose clearance.44
Bitter ligands stimulate GLP-1 secretion from TAS2R-expressing
enteroendocrine cell lines (STC-1 and NCI-H716), suggesting that
bitter taste receptors could have an impact on glucose and insulin
regulation.52 71 In fact, in an Amish Family Diabetes Study, a func-
tionally comprised TAS2R was associated with disturbed glucose
Figure 3 Mechanisms involved in glucose-stimulated GLP-1 release. homeostasis.71
Glucose can stimulate gut peptide secretion by (1) binding to the sweet GPRC6A was recently shown to act as an amino acid sensor
taste receptor heterodimer, TAS1R2-TAS1R3, coupling via the G-protein, in GLUTag that enhances GLP-1 secretion.72
gustducin, to increase intracellular Ca2+ resulting in vesicle fusion and
consequent release of GLP-1; (2) Na+-coupled glucose transport via
SGLT1 which will depolarise the membrane resulting in the opening of Nutrient sensing in the colon
voltage-gated Ca2+ channels and influx of Ca2+. Additionally, transport
The main function of the colon is to store food residues, secrete
of glucose into the cell via SGLT1 will lead to increased metabolism
and closure of K ATP-sensitive channels. ATP, adenosine triphosphate; mucus and absorb remaining water and electrolytes from the
GLP-1, glucagon-like peptide; KATP, ATP-sensitive potassium channel; food residue before defecation. The colon exhibits segmental
PLCβ2, phospholipase C β2; TAS1R2, taste receptor type 1 member 2; movements (haustrations) and phasic propulsive contractions.
TAS1R3, taste receptor type 1 member 3; SGLT1, sodium-dependent The intestinal flora performs fermentation reactions that
glucose transporter 1. produce SCFA and flatus.

Depoortere I. Gut 2013;0:1–12. doi:10.1136/gutjnl-2013-305112 5


Downloaded from http://gut.bmj.com/ on April 8, 2016 - Published by group.bmj.com

Recent advances in basic science

SCFAs ( propionate, acetate and butyrate) are sensed in the increased blood glucose and decreased insulin levels. The
lumen by the free fatty acid receptors, FFAR2 and FFAR3.73 In TAS1R3−/− phenotype was less severe after an intraperitoneal
the human colon, FFAR2 immunoreactive cells were colocalised injection of glucose, indicating that some, but not all, of the
with PYY-containing L-cells but not with 5-HT containing endo- glucose dysregulation results from the disruption of normal gut
crine cells.74 In the rat colon, 5-HT-containing mucosal mast physiology. These findings are somewhat at variance with the
cells were immunoreactive for FFAR2.75 FFAR3-positive cells in studies in patients with type 2 diabetes where a dysregulation of
the human colon were less abundantly present than FFAR2 cells TAS1R2 but not of TAS1R3 was observed during a luminal
and contained PYY but not 5-HT or FFAR2.76 α-gustducin Is glucose challenge and resulted in an increased glucose
colocalised with several fatty acid receptors present on endo- absorption.
crine cells in the mouse colon and is required for It is clear that the regulation of sweet taste receptors and
SCFA-mediated GLP-1 release from mucosal explants.77 The glucose transporters during different glycemic conditions is
effect of SCFA on the release of GLP-1 was also confirmed in complex. More studies using different models and conditions
isolated primary L-cells from mouse colon. In FFAR2−/− and are therefore warranted.
FFAR3−/− mice reduced SCFA-induced GLP-1 secretion was
observed in vitro and in vivo together with a parallel impair- Gastric bypass
ment of glucose tolerance.78 Gastric bypass surgery is one of the most effective methods for
The functional role of TAS1R3 in L-cells of the human colon inducing a marked and sustained weight loss in morbidly obese
is not clear.55 GLP-1-secreting cells of the colon and rectum are patients. This procedure also results in amelioration or even
normally not responsive to sugars, suggesting no role for the complete remission of type 2 diabetes mellitus independent
sweet taste receptor in hindgut GLP-1 secretion. Even when from weight loss.84 The mechanisms involved are incompletely
these cells become glucose-responsive in association with carbo- understood but involve restriction, malabsorption and humoral
hydrate malabsorption, for instance after gastric bypass surgery, changes. Indeed, bypass surgery decreases the release of the
this glucose sensitivity is sweet taste receptor independent (the orexigenic hormone, ghrelin, and increases the release of sati-
identity of the glucose sensor is unknown). ation hormones PYY and GLP-1. After gastric bypass surgery,
the contact of nutrients with much of the stomach, duodenum
CONTRIBUTIONS OF TASTE RECEPTORS TO and part of the jejunum is bypassed. It can be hypothesised that
PATHOPHYSIOLOGY: POSSIBLE TARGETS FOR THERAPY? nutrient sensors on the ghrelin cell are isolated from contact
Glucose homeostasis and diabetes with nutrients thereby probably affecting ghrelin release and,
Young et al79 80 were the first to compare the intestinal levels of consequently, energy and glucose homeostasis. Additionally, the
sweet taste receptors in patients with well-controlled type 2 dia- rapid delivery of undigested nutrients to the lower small intes-
betes to those without diabetes. Absolute levels of TAS1R2, tine may affect the regulation of taste receptors and/or glucose
TAS1R3, α-gustducin or TRPM5 transcripts in the duodenum transporters on L-cells, resulting in enhanced release of PYY and
of healthy people, or patients with type 2 diabetes, were GLP-1. Indeed, duodenal–jejunal bypass surgery in a rat model
unaffected by acute variations in glycaemia during fasting. of type 2 diabetes decreased SGLT1 mRNA and
However, the intestinal sweet taste receptor system was highly SGLT1-mediated transport in jejunum distal to the duodenojeju-
responsive to changes in luminal glucose. During euglycemia, nostomy and improved diabetes independent from body weight
intraduodenal glucose infusion increased TAS1R2 transcript loss.85 A similar observation was made in non-obese mice after
levels in both groups but during hyperglycaemia TAS1R2 duodenojejunal bypass surgery.86 Sweet taste receptors regulate
mRNA expression decreased in healthy volunteers but not in SGLT1 and a significant decrease in TAS1R2 and TAS1R3
diabetics. Levels of TAS1R3 did not change significantly. The protein levels in the alimentary limb after gastric bypass have
TAS1R2 dysregulation in patients with type 2 diabetes may been demonstrated in rats.87 These results suggest that TAS1R3
potentially increase the risk of postprandial hyperglycaemia antagonists or SGLT1 inhibitors may represent a promising
since they exhibited increased glucose absorption during hyper- target for the treatment of obesity. In fact, LX4211, a dual intes-
glycaemia, as evidenced by an increase in the glucose absorption tinal SGLT1/renal SGLT2 inhibitor increased GLP-1 levels and
marker 3-O-methyl-glucose (3-OMG), compared with healthy PYY levels in patients with type 2 diabetes and improved blood
subjects. As SGLT1 is responsible for the active transport of glucose levels and, therefore, mimicked the effects of gastric
luminal 3-OMG, these studies confirm that TAS1R2 regulates bypass surgery.88 These results are inconsistent with studies
glucose absorption via SGLT1. The positive association found showing that SGLT1 is required for glucose-mediated GLP-1
between luminal glucose-induced changes in some sweet taste release by L-cells in vitro.61 63 However, due to the carbohy-
receptor transcripts and the secretion of GLP-1 and GIP under- drate malabsorption induced by SGLT1 inhibition, L-cells in the
scores that sweet taste receptors do have a regulatory role in the colon also become glucose sensitive and increase GLP-1 levels.89
release of incretins. Although these findings are supported by in The authors suggested that this may be mediated by SCFAs pro-
vitro studies, in vivo studies either in rodents or in humans duced by colonic fermentation of unabsorbed glucose that inter-
failed to show an effect of artificial sweeteners on the release of act with FFARs on L-cells to induce GLP-1 and PYY release. A
satiation hormones, thereby questioning the role of sweet taste KATP channel-dependent mechanism involving an unknown
receptors in incretin release.56 58 81–83 Nevertheless, chronic glucose sensor has also been suggested in glucose-induced
exposure of high-fat fed diabetic mice with the artificial sweet- hindgut GLP-1 secretion during carbohydrate malabsorption.58
ener, oligofructose, increased GLP-1 levels. Thus, the relative After gastric bypass surgery, patients show a decreased prefer-
importance of glucose transporters versus sweet taste receptors ence for sweet and fatty foods, but the factors involved are
in the release of incretins remains a matter of debate. unclear. The decision of what to eat is modulated by taste, olfac-
Glucose and insulin levels are remarkably normal in tion and oral textural perception. Taste, in particular, has an
TAS1R2−/− and TAS1R3−/− mice, despite the presence of sweet important input into food preference because it has direct
taste ageusia in both genotypes. However, after an oral glucose effects on brain reward circuits that drive eating. It has been sug-
challenge TAS1R3−/− mice, but not TAS1R2−/− mice, showed gested that the acuity for sweet taste increases after gastric

6 Depoortere I. Gut 2013;0:1–12. doi:10.1136/gutjnl-2013-305112


Downloaded from http://gut.bmj.com/ on April 8, 2016 - Published by group.bmj.com

Recent advances in basic science

bypass, potentially leading to increased intensity of percep- mechanisms of action have been put forward for the FODMAP
tion.90 It remains to be investigated whether endocrine cells in diet: (1) FODMAPs are poorly absorbed in the small intestine
the gut also show changes in sweet taste sensitivity after gastric and are osmotically active, thereby drawing fluid through the
bypass surgery. Roux en-Y gastric bypass (RYGB) could further large bowel leading to diarrhoea97; (2) fermentation of the
reset the food reward system through changes in the release of FODMAPs in the colon will generate gases that may be incorpo-
gut hormones, known to modify activity in brain reward rated in volatile end-products.98 The increase in fluid and gas
systems and dopaminergic signalling, but also known to modu- components will lead to luminal distension and may, in the pres-
late taste sensitivity in taste buds.91 92 Indeed, a recent study ence of visceral hypersensitivity, induce bloating, flatulence,
showed that obese patients, after RYGB, had lower brain- abdominal pain and motility disturbances.
hedonic responses to food than patients after gastric banding.93 In this review, we speculate that alterations in nutrient-sensing
Additionally, aversive conditioning during the early postsurgical mechanisms may also play a role in symptom generation in
period where unpleasant feelings (eg, dumping) associated with patients with IBS.
particular foods can lead to conditioning of food aversion, may
also play a role. Altered short-chain fatty acid sensing?
Fermentation of FODMAPs, such as fructans, in the colon will
Irritable bowel syndrome (IBS): FODMAPS and gluten also result in the production of SCFAs, known to affect local
sensitivity ion secretion99–101 and colonic motility.102 103 Indeed, intralum-
Patients with irritable bowel syndrome (IBS) claim that diet plays inal application of SCFA in the rat proximal colon accelerated
a major role in triggering GI symptoms.94 The most common colonic transit by inducing the release of 5-HT that stimulates
approaches to manage food intolerance in IBS include: (1) the 5-HT3 receptors located on vagal sensory fibres, resulting in
low-FODMAP (fermentable, oligosaccharides disaccharides, muscle contraction via a vagal reflex pathway involving the
monosaccharides and polyols) diet, (2) the gluten-free diet, (3) release of ACh from the myenteric plexus (figure 4).104 105 The
the elimination diet for food chemicals. How foods trigger func- role of 5-HT in SCFAs-induced contractions was confirmed in
tional gut symptoms is unclear. contractility studies with smooth muscle strips in vitro.106 In
fact, use of 5-HT3 antagonists has been shown to improve
The low-FODMAP diet symptoms in patients with IBS.107 We hypothesise that FFAR2
Recent trials showed that dietary FODMAPs, especially fructose receptors on mucosal mast cells mediate the effect of SCFA on
and fructans, are dietary triggers for symptom generation, and 5-HT release, since enterochromaffin cells and smooth muscle
that restricting their level of intake might lead to durable cells do not contain FFARs (figure 4).74 75 Cremon et al108
symptom improvement in patients with IBS.95 96 Two showed that 5-HT release from mucosal biopsy specimens of

Figure 4 Proposed model for a role of altered short-chain fatty acid sensing that might contribute to symptom generation in patients with IBS in
response to ingestion of FODMAPs. Bacterial overgrowth in the small intestine of patients with IBS may result in the generation of short-chain fatty
acids (SCFAs) that may increase the sensitivity of FFAR2/3 on I-cells. The resulting increased release of CCK is known to alter intestinal motor activity
and to reduce pain thresholds in patients with IBS. In the colon, fermentation of fructans leads to the production of gases and SCFAs. Increased
FFAR2/3 sensing may enhance the secretion of GLP-1 and PYY from L-cells and 5-HT from mast cells, and may modulate visceral sensation and
motility by activation of extrinsic sensory neurons. ACh, acetylcholine; CCK, cholecystokinin; CCK1R, cholecystokinin 1 receptor; FFAR2/3, free fatty
acid receptor 2/3; FODMAPs, fermentable, oligosaccharides, disaccharides, monosaccharides and polyols; GLP-1, glucagon-like peptide 1; GLP-1 R,
glucagon-like peptide 1 receptor; 5-HT, 5-hydroxytryptamine; 5HT3, 5-hydroxytryptamine 3 receptor; IBS, irritable bowel syndrome; PYY, peptide YY;
SCFA, short-chain fatty acids; Y2R, Y2 receptor.

Depoortere I. Gut 2013;0:1–12. doi:10.1136/gutjnl-2013-305112 7


Downloaded from http://gut.bmj.com/ on April 8, 2016 - Published by group.bmj.com

Recent advances in basic science

patients with IBS was increased and correlated with mast cell FFAR3 and transporters to affect ion secretion and GI motility
counts and the severity of the abdominal pain. It can be by releasing hormones and neurotransmitters. Studies are war-
hypothesised that in patients with IBS, alterations in the sensitiv- ranted to investigate whether alterations in the number or sensi-
ity or number of free fatty acid receptors involved in the tivity of these FFARs occur in patients with IBS. If so, FFAR2
SCFA-induced release of 5-HT from mast cells, may play an and FFAR3 antagonists, currently under development, may
important role. therefore become promising tools for the treatment of IBS.118
As already outlined, FFAR2 and FFAR3 sense SCFAs to affect Since SCFAs induce neutrophil chemotaxis through FFAR2, a
the release of gut peptides. Patients with IBS have increased low FODMAP diet or FFAR2 antagonists may, therefore, also
fasting and postprandial CCK levels.109 Isolated duodenal I-cells help to control inflammation in subsets of patients with IBS
are highly enriched in FFAR2 and FFAR3 mRNA transcripts.105 with low-grade inflammation.119
Under normal conditions these cells mainly sense circulating
SCFAs. The concept that small intestinal bacterial overgrowth is Altered carbohydrate sensing?
a major pathogenic mechanism underlying IBS is still a matter Effects observed in TAS1R3−/− and GLUT5−/− mice mimic the
of debate.110–112 Nevertheless, in those patients with IBS with effects observed in patients with IBS. Due to duodenal carbohy-
bacterial overgrowth, luminal SCFA may also be sensed by the drate malabsorption, both genotypes display a distended prox-
I-cells and increase CCK secretion (figure 4). Infusion of CCK imal colon with the development of gas pockets as a result of
in patients with IBS can lead to excessive intestinal motor activ- subsequent fermentation by intestinal microflora.58 120
ity and reduced pain thresholds.113 114 Additionally, the increased carbohydrate content in the colon of
The postprandial PYY response is significantly increased in TAS1R3−/− mice, induced robust glucose-induced GLP-1 secre-
hypersensitive compared to normosensitive patients with IBS.109 tion from the colon which may induce alterations in transit
We propose that FFAR2 activation by SCFAs might be an import- (figure 5). Fructose interacts with TAS1R3 and GLUT5.58 120
ant trigger for the release PYY/GLP-1 from colonic L-cells (figure We hypothesise that the fructose malabsorption in some patients
4). Indeed, feeding rats a fructo-oligosaccharide-enriched diet with IBS may be due to alterations in the basal expression of
selectively induced the proliferation of FFAR2-positive TAS1R3 and GLUT5 or in their regulation in response to
L-cells.115 Additionally, colonic infusion of SCFA stimulated PPY dietary fructose. Therefore, future studies are warranted to
release in rats. Immunoneutralisation of circulating PYY abolished investigate the mechanisms of altered glucose sensing in patients
the effect of SCFA on colonic motility while the effect was mim- with IBS.
icked by exogenous PYY infusion.116 In FFAR3−/− mice, charac-
terised by a decrease in PYY expression, intestinal transit rate was The gluten-free diet
increased.117 Gluten is a well known trigger for GI symptoms in the setting
In conclusion, the low FODMAP diet might also improve of coeliac disease. The existence of gluten intolerance was
symptoms by reducing the levels of SCFAs that act via FFAR2, demonstrated in patients with IBS without celiac disease.121

Figure 5 Possible role of changes in carbohydrate sensing that may contribute to symptom generation in patients with IBS in response to
ingestion of FODMAPs. Decreased expression or sensitivity of sweet taste receptors and/or concomitant alterations in GLUT5 expression may
contribute to fructose malabsorption in patients with IBS. Due to the increased carbohydrate content in the colon, the L-cells become glucose
sensitive through a sweet taste receptor-independent pathway involving closure of KATP channels and influx of Ca2+ through voltage-gated Ca2+
channels. The resulting increase in GLP-1 release may induce changes in transit due to the ileal brake effect. ATP, adenosine triphosphate;
FODMAPs, fermentable oligosaccharides, disaccharides, monosaccharides and polyols; GLP-1, glucagon-like peptide 1; GLUT2, glucose transporter 2;
GLUT5, glucose transporter 5; KATP, ATP-sensitive potassium channel; TAS1R2, taste receptor type 1 member 2; TAS1R3, taste receptor type 1
member 3; SGLT1, sodium-dependent glucose transporter.

8 Depoortere I. Gut 2013;0:1–12. doi:10.1136/gutjnl-2013-305112


Downloaded from http://gut.bmj.com/ on April 8, 2016 - Published by group.bmj.com

Recent advances in basic science

However, the same authors recently reported that gluten might completion of ingestion. Furthermore the scores for nausea and
not be a specific trigger of functional gut symptoms once pain were related directly to the increased plasma CCK concen-
dietary FODMAPs are reduced.122 Another controlled trial of trations in patients with functional dyspepsia. It is known that
gluten-free diet showed that gluten alters small bowel perme- exogenous CCK administration can mimic dyspeptic symp-
ability in diarrhoea-predominant patients with IBS, particularly toms.135 It is therefore conceivable that similar to patients with
in HLA-DQ2/8-positive patients.123 This may result in greater IBS, who frequently overlap with functional dyspepsia patients,
fluid flux toward the lumen and may elicit immune responses alterations in the expression or sensitivity of free fatty acid
that affect afferent nerves resulting in hypersensitivity. receptors on endocrine cells may render them more sensitive to
dietary fat and exacerbate symptoms.136 Diet-intervention
Altered glutamate sensing? studies, similar to what has been performed in patients with
Glutamic acid (Glu) and proline together account for one half IBS, are warranted to investigate the role of nutrients and,
or more of the peptide-bound amino acids in gluten. Ingestion hence, of taste receptors in symptom generation in patients with
of a protein diet rich in L-Glu does not lead to appreciable functional dyspepsia.
changes in plasma glutamate concentrations, since glutamate is
extensively metabolised by enterocytes and does not reach the
portal vein.124 Thus, once proteins such as wheat are digested, CONCLUSIONS AND PERSPECTIVES
dietary Glu may stimulate Glu sensors (e.g TAS1R -TAS1R3, The observation, and now compelling evidence, for the pres-
CaSR and metabotropic glutamate receptors (mGluR)) in the ence and function of taste receptors in extraoral tissues offers
stomach and intestine producing local effects on gut function. exciting new possibilities for targeted therapeutics in the battle
For example, in dogs, intragastric, but not intraduodenal, against diseases of and involving the GI tract. Several taste
administration of monosodium glutamate (MSG) stimulated receptor families involved in nutrient sensing in the gut offer
gastric emptying and induced phasic non-propagating contrac- potential advantages as drug targets.
tions in the upper gut which were blocked by vagotomy.125
Contradictory results have been reported concerning the effect
of MSG on gastric emptying in humans.126 127 Sweet taste receptors
Among 20 natural amino acids investigated, only L-Glu This long history of sweetener chemistry suggests that new com-
evoked firing of afferent fibres of the vagal gastric branch in pounds of exceptional specificity and high efficacy could be
rats. The effect is mediated via metabotropic L-Glu receptors designed. The beneficial effects of artificial sweeteners in the
that induce the release of 5-HT from mucosal cells interacting control of body weight and glucose homeostasis are still a
with 5-HT3 receptors on afferent fibres.128 Additionally, intra- matter of debate, but advances in our understanding of the
gastric infusion of Glu activated brain regions that are directly sweet taste receptor biology may therefore help to design new
or indirectly targeted by these vagal inputs.129 Nevertheless, the artificial sweeteners with favourable effects.137 Indeed, in con-
effect of glutamate on vagal afferent firing seems not to be trast with in vitro studies, in vivo studies either in rodents or in
straightforward, since inhibitory effects of exogenous and humans failed to show an effect of artificial sweeteners on the
endogenous glutamate have been reported on vagal afferent release of satiation hormones. 56 58 81–83 Advances in our under-
mechanosensitivity involving group II (mGluR2 and 3) and standing of the sweet taste receptor biology may therefore help
group III mGluR (mGlu R4, 6, 7, 8).130 to design new artificial sweeteners with favourable effects.
In view of the beneficial effects that are observed with a However, their effectiveness may be complicated by the
gluten-free diet in patients with IBS, we hypothesise that altera- complex interaction between sweet taste receptors and glucose
tions in luminal glutamate sensing may occur that affect vagal transporters that cross-regulate each other’s expression.
nerve activity resulting in altered brain–gut interactions and Additionally, the evidence provided for altered sweet taste recep-
symptom generation. tor control in type 2 diabetes may open new opportunities for
drugs that interfere with sweet taste receptor regulation.
The elimination diet for food chemicals In gastric bypass patients, additional studies should help to
The elimination diet involves restriction of common food aller- provide insight to what the effect is of shunting nutrients to
gens, specific chemical substances in foods or medications that more distal regions of the gut in the control of taste receptor
contain these chemicals. expression/glucose transporter regulation, and the release of
Coffee is commonly reported as a trigger for symptoms in hormones involved in the regulation of energy and glucose
patients with IBS.131 It is not clear whether salicylates or caf- homeostasis. It is tempting to speculate that targeting these
feine in coffee are the triggers but both components taste bitter receptors/transporters may mimic the effects of gastric bypass
and could alter gut function via TAS2Rs. surgery in a non-surgical manner. In fact, ongoing trials with
SGLT1 inhibitors already show promising results.88
Functional dyspepsia
According to the Rome III classification, functional dyspepsia
can be divided into two categories: postprandial distress syn- Bitter taste receptors
drome and epigastric pain syndrome, suggesting that at least in Evidence suggests that bitter agonists could be considered as
some patients the disorder is related to food ingestion. While good targets to reduce hunger and motility.18 22 32 However,
most of the patients report that their symptoms are triggered the design of new bitter drugs may be complicated by the fact
within 30 min after meal ingestion, few studies have been per- that in humans, 25 receptor subtypes exist, each with different
formed to evaluate the role of specific foods.132 Particularly, ligand selectivities and a different distribution pattern.
meals containing fat seem to induce or exacerbate symptoms.133 Nevertheless, there are also some opportunities. Thousands of
Pilichiewicz et al134 reported that ingestion of a high-fat meal, plant-derived bitter tastants and metabolites are available that
but not of a high-carbohydrate meal, was associated with a sub- could be tested in appropriate physiological models to select
stantially greater increase in nausea and pain immediately after those with favourable therapeutic profiles.

Depoortere I. Gut 2013;0:1–12. doi:10.1136/gutjnl-2013-305112 9


Downloaded from http://gut.bmj.com/ on April 8, 2016 - Published by group.bmj.com

Recent advances in basic science

Amino acid and free fatty acid receptors 21 Foster-Schubert KE, Overduin J, Prudom CE, et al. Acyl and total ghrelin are
Recent evidence suggests that food choice (low FODMAPs, suppressed strongly by ingested proteins, weakly by lipids, and biphasically by
carbohydrates. J Clin Endocrinol Metab 2008;93:1971–9.
gluten-free diet) is a key management strategy for the treatment 22 Janssen S, Laermans J, Verhulst PJ, et al. Bitter taste receptors and
of symptoms in patients with IBS. These studies underscore alpha-gustducin regulate the secretion of ghrelin with functional effects on food
claims made for many years by alternative practitioners. The intake and gastric emptying. Proc Natl Acad Sci U S A 2011;108:2094–9.
mechanisms involved are not completely understood, but indir- 23 Janssen S, Laermans J, Iwakura H, et al. Sensing of fatty acids for octanoylation of
ghrelin involves a gustatory G-protein. PLoS One 2012;7:e40168.
ect evidence suggests that altered carbohydrate sensing, exacer-
24 Hass N, Schwarzenbacher K, Breer H. T1R3 is expressed in brush cells and
bated sensing of SCFAs by FFARs or of Glu by amino acid ghrelin-producing cells of murine stomach. Cell Tissue Res 2010;339:493–504.
receptors may be involved. Determining alterations in basal 25 Saffouri B, DuVal JW, Makhlouf GM. Stimulation of gastrin secretion in vitro by
expression levels of these receptors or in their regulation in intraluminal chemicals: regulation by intramural cholinergic and noncholinergic
response to a diet may help to provide a rationale for the use of neurons. Gastroenterology 1984;87:557–61.
26 Haid D, Widmayer P, Voigt A, et al. Gustatory sensory cells express a receptor
FFAR antagonists or Glu receptor antagonists in these patients responsive to protein breakdown products (GPR92). Histochem Cell Biol
and may help to predict whether a patient will be responsive to 2013;140:137–45.
the diet or not. Similar strategies may be useful in patients with 27 Haid DC, Jordan-Biegger C, Widmayer P, et al. Receptors responsive to protein
functional dyspepsia. breakdown products in g-cells and d-cells of mouse, swine and human. Front
Physiol 2012;3:65.
It is clear that the existence and functional role of taste recep-
28 Haid D, Widmayer P, Breer H. Nutrient sensing receptors in gastric endocrine cells.
tors in the gut is a new and fascinating field of research that J Mol Histol 2011;42:355–64.
may lead to the development of new therapeutic drugs that may 29 Feng J, Petersen CD, Coy DH, et al. Calcium-sensing receptor is a physiologic
benefit from the progress made in our understanding of taste multimodal chemosensor regulating gastric G-cell growth and gastrin secretion.
receptor function in the tongue. Proc Natl Acad Sci U S A 2010;107:17791–6.
30 Busque SM, Kerstetter JE, Geibel JP, et al. L-type amino acids stimulate gastric
Acknowledgements The author wishes to thank S. Munger for providing helpful acid secretion by activation of the calcium-sensing receptor in parietal cells. Am J
discussion. Physiol Gastrointest Liver Physiol 2005;289:G664–9.
31 Gielkens HA, van den Biggelaar A, Vecht J, et al. Effect of intravenous amino
Competing interests None. acids on interdigestive antroduodenal motility and small bowel transit time. Gut
Provenance and peer review Commissioned; externally peer reviewed. 1999;44:240–5.
32 Deloose E, Corsetti M, Van Oudenhove L, et al. In man intragastric administration
of the bitter compound denatonium benzoate decreases hunger and the occurence
REFERENCES of gastric phase III in the fasting state. Gastroenterology 2013;144(suppl 1):S-548.
1 Roper SD. Taste buds as peripheral chemosensory processors. Semin Cell Dev Biol 33 Deloose E, Janssens J, Lannoo M, et al. Obesity causes a switch in the origin of
2013;24:71–9. phase III contractions of the migrating complex to reduce hunger feelings.
2 Nelson G, Hoon MA, Chandrashekar J, et al. Mammalian sweet taste receptors. Neurogastroenterol Motil 2012;24:31.
Cell 2001;106:381–90. 34 Gibbs J, Maddison SP, Rolls ET. Satiety role of the small intestine examined in
3 Nelson G, Chandrashekar J, Hoon MA, et al. An amino-acid taste receptor. Nature sham-feeding rhesus monkeys. J Comp Physiol Psychol 1981;95:1003–15.
2002;416:199–202. 35 Broberger C, Holmberg K, Shi TJ, et al. Expression and regulation of
4 Zhao GQ, Zhang Y, Hoon MA, et al. The receptors for mammalian sweet and cholecystokinin and cholecystokinin receptors in rat nodose and dorsal root
umami taste. Cell 2003;115:255–66. ganglia. Brain Res 2001;903:128–40.
5 Clark AA, Liggett SB, Munger SD. Extraoral bitter taste receptors as mediators of 36 Blackshaw LA, Grundy D. Effects of cholecystokinin (CCK-8) on two classes of
off-target drug effects. Faseb J 2012;26:4827–31. gastroduodenal vagal afferent fibre. J Auton Nerv Syst 1990;31:191–201.
6 Chandrashekar J, Mueller KL, Hoon MA, et al. T2Rs function as bitter taste 37 Gaisano GG, Park SJ, Daly DM, et al. Glucagon-like peptide-1 inhibits
receptors. Cell 2000;100:703–11. voltage-gated potassium currents in mouse nodose ganglion neurons.
7 Meyerhof W, Batram C, Kuhn C, et al. The molecular receptive ranges of human Neurogastroenterol Motil 2010;22:470–9, e111.
TAS2R bitter taste receptors. Chem Senses 2010;35:157–70. 38 Zittel TT, Glatzle J, Kreis ME, et al. C-fos protein expression in the nucleus of the
8 Behrens M, Meyerhof W. Bitter taste receptors and human bitter taste perception. solitary tract correlates with cholecystokinin dose injected and food intake in rats.
Cell Mol Life Sci 2006;63:1501–9. Brain Res 1999;846:1–11.
9 Chaudhari N, Pereira E, Roper SD. Taste receptors for umami: the case for multiple 39 Baggio LL, Huang Q, Brown TJ, et al. A recombinant human glucagon-like peptide
receptors. Am J Clin Nutr 2009;90:738S–42S. (GLP)-1-albumin protein (albugon) mimics peptidergic activation of GLP-1
10 Bystrova MF, Romanov RA, Rogachevskaja OA, et al. Functional expression of the receptor-dependent pathways coupled with satiety, gastrointestinal motility, and
extracellular-Ca2+-sensing receptor in mouse taste cells. J Cell Sci glucose homeostasis. Diabetes 2004;53:2492–500.
2010;123:972–82. 40 Abbott CR, Monteiro M, Small CJ, et al. The inhibitory effects of peripheral
11 Janssen S, Depoortere I. Nutrient sensing in the gut: new roads to therapeutics? administration of peptide YY(3–36) and glucagon-like peptide-1 on food intake
Trends Endocrinol Metab 2013;24:92–100. are attenuated by ablation of the vagal-brainstem-hypothalamic pathway. Brain
12 Cartoni C, Yasumatsu K, Ohkuri T, et al. Taste preference for fatty acids is Res 2005;1044:127–31.
mediated by GPR40 and GPR120. J Neurosci 2010;30:8376–82. 41 Crawley JN, Kiss JZ, Mezey E. Bilateral midbrain transections block the behavioral
13 Chandrashekar J, Hoon MA, Ryba NJ, et al. The receptors and cells for effects of cholecystokinin on feeding and exploration in rats. Brain Res
mammalian taste. Nature 2006;444:288–94. 1984;322:316–21.
14 Dotson CD, Geraedts MC, Munger SD. Peptide regulators of peripheral taste 42 Tanaka T, Katsuma S, Adachi T, et al. Free fatty acids induce cholecystokinin
function. Semin Cell Dev Biol 2013;24:232–9. secretion through GPR120. Naunyn Schmiedebergs Arch Pharmacol
15 Geraedts MC, Munger SD. Gustatory stimuli representing different perceptual 2008;377:523–7.
qualities elicit distinct patterns of neuropeptide secretion from taste buds. 43 Shah BP, Liu P, Yu T, et al. TRPM5 is critical for linoleic acid-induced CCK
J Neurosci 2013;33:7559–64. secretion from the enteroendocrine cell line, STC-1. Am J Physiol Cell Physiol
16 Kawai K, Sugimoto K, Nakashima K, et al. Leptin as a modulator of sweet taste 2012;302:C210–9.
sensitivities in mice. Proc Natl Acad Sci U S A 2000;97:11044–9. 44 Edfalk S, Steneberg P, Edlund H. Gpr40 is expressed in enteroendocrine cells and
17 Janssen P, Vanden Berghe P, Verschueren S, et al. Review article: the role of mediates free fatty acid stimulation of incretin secretion. Diabetes
gastric motility in the control of food intake. Aliment Pharmacol Ther 2008;57:2280–7.
2011;33:880–94. 45 Liou AP, Lu X, Sei Y, et al. The G-protein-coupled receptor GPR40 directly
18 Andrews CN, Verschueren S, Janssen P, et al. The bitter taste receptor agonist mediates long-chain fatty acid-induced secretion of cholecystokinin.
denatonium benzoate alters intragastric pressure profiles during nutrient drink test Gastroenterology 2011;140:903–12.
in healthy volunteers. Gastroenterology 2013;144(suppl 1):S-549. 46 Ballinger AB, Clark ML. L-phenylalanine releases cholecystokinin (CCK) and is
19 Avau B, Thijs T, Laermans J, et al. Endocrine and smooth muscle responses of the associated with reduced food intake in humans: evidence for a physiological role
bitter agonist, denatonium benzoate, in the stomach. Neurogastroenterol Motil of CCK in control of eating. Metabolism 1994;43:735–8.
2012;24:153, A378. 47 Daly K, Al-Rammahi M, Moran A, et al. Sensing of amino acids by the
20 Kojima M, Hosoda H, Date Y, et al. Ghrelin is a growth-hormone-releasing gut-expressed taste receptor T1R1-T1R3 stimulates CCK secretion. Am J Physiol
acylated peptide from stomach. Nature 1999;402:656–60. Gastrointest Liver Physiol 2013;304:G271–82.

10 Depoortere I. Gut 2013;0:1–12. doi:10.1136/gutjnl-2013-305112


Downloaded from http://gut.bmj.com/ on April 8, 2016 - Published by group.bmj.com

Recent advances in basic science

48 Hira T, Nakajima S, Eto Y, et al. Calcium-sensing receptor mediates 76 Tazoe H, Otomo Y, Karaki S, et al. Expression of short-chain fatty acid receptor
phenylalanine-induced cholecystokinin secretion in enteroendocrine STC-1 cells. GPR41 in the human colon. Biomed Res 2009;30:149–56.
Febs J 2008;275:4620–6. 77 Li Y, Kokrashvili Z, Mosinger B, et al. Gustducin couples fatty acid receptors to
49 Liou AP, Sei Y, Zhao X, et al. The extracellular calcium-sensing receptor is required GLP-1 release in colon. Am J Physiol Endocrinol Metab 2013;304:E651–60.
for cholecystokinin secretion in response to L-phenylalanine in acutely isolated 78 Tolhurst G, Heffron H, Lam YS, et al. Short-chain fatty acids stimulate
intestinal I cells. Am J Physiol Gastrointest Liver Physiol 2011;300:G538–46. glucagon-like peptide-1 secretion via the G-protein-coupled receptor FFAR2.
50 Wang Y, Chandra R, Samsa LA, et al. Amino acids stimulate cholecystokinin Diabetes 2012;61:364–71.
release through the Ca2+-sensing receptor. Am J Physiol Gastrointest Liver Physiol 79 Young RL, Chia B, Isaacs NJ, et al. Disordered control of intestinal sweet taste
2011;300:G528–37. receptor expression and glucose absorption in type 2 diabetes. Diabetes
51 Choi S, Lee M, Shiu AL, et al. GPR93 activation by protein hydrolysate induces 2013;62:3532–41.
CCK transcription and secretion in STC-1 cells. Am J Physiol Gastrointest Liver 80 Young RL, Sutherland K, Pezos N, et al. Expression of taste molecules in the upper
Physiol 2007;292:G1366–75. gastrointestinal tract in humans with and without type 2 diabetes. Gut 2009;58:337–46.
52 Jeon TI, Zhu B, Larson JL, et al. SREBP-2 regulates gut peptide secretion through 81 Ford HE, Peters V, Martin NM, et al. Effects of oral ingestion of sucralose on gut
intestinal bitter taste receptor signaling in mice. J Clin Invest 2008;118:3693–700. hormone response and appetite in healthy normal-weight subjects. Eur J Clin Nutr
53 Chen MC, Wu SV, Reeve JR Jr., et al. Bitter stimuli induce Ca2+ signaling and 2011;65:508–13.
CCK release in enteroendocrine STC-1 cells: role of L-type voltage-sensitive Ca2+ 82 Steinert RE, Frey F, Topfer A, et al. Effects of carbohydrate sugars and artificial
channels. Am J Physiol Cell Physiol 2006;291:C726–39. sweeteners on appetite and the secretion of gastrointestinal satiety peptides. Br J
54 Gerspach AC, Steinert RE, Schonenberger L, et al. The role of the gut sweet taste Nutr 2011;105:1320–8.
receptor in regulating GLP-1, PYY, and CCK release in humans. Am J Physiol 83 Ma J, Bellon M, Wishart JM, et al. Effect of the artificial sweetener, sucralose, on
Endocrinol Metab 2011;301:E317–25. gastric emptying and incretin hormone release in healthy subjects. Am J Physiol
55 Steinert RE, Gerspach AC, Gutmann H, et al. The functional involvement of Gastrointest Liver Physiol 2009;296:G735–9.
gut-expressed sweet taste receptors in glucose-stimulated secretion of 84 Rubino F, Schauer PR, Kaplan LM, et al. Metabolic surgery to treat type 2 diabetes:
glucagon-like peptide-1 (GLP-1) and peptide YY (PYY). Clin Nutr clinical outcomes and mechanisms of action. Annu Rev Med 2010;61:393–411.
2011;30:524–32. 85 Jurowich CF, Rikkala PR, Thalheimer A, et al. Duodenal-Jejunal Bypass Improves
56 Jang HJ, Kokrashvili Z, Theodorakis MJ, et al. Gut-expressed gustducin and taste Glycemia and Decreases SGLT1-Mediated Glucose Absorption in rats with
receptors regulate secretion of glucagon-like peptide-1. Proc Natl Acad Sci U S A streptozotocin-induced type 2 diabetes. Ann Surg 2013;258:89–97.
2007;104:15069–74. 86 Yan S, Sun F, Li Z, et al. Reduction of intestinal electrogenic glucose absorption
57 Rozengurt N, Wu SV, Chen MC, et al. Colocalization of the alpha-subunit of after duodenojejunal bypass in a mouse mγδodel. Obes Surg 2013;23:1361–9.
gustducin with PYY and GLP-1 in L cells of human colon. Am J Physiol 87 Bueter M, Miras AD, Chichger H, et al. Alterations of sucrose preference after
Gastrointest Liver Physiol 2006;291:G792–802. Roux-en-Y gastric bypass. Physiol Behav 2011;104:709–21.
58 Geraedts MC, Takahashi T, Vigues S, et al. Transformation of postingestive glucose 88 Zambrowicz B, Ding ZM, Ogbaa I, et al. Effects of LX4211, a dual SGLT1/SGLT2
responses after deletion of sweet taste receptor subunits or gastric bypass surgery. inhibitor, plus sitagliptin on postprandial active GLP-1 and glycemic control in type
Am J Physiol Endocrinol Metab 2012;303:E464–74. 2 diabetes. Clin Ther 2013;35:273–85, e7.
59 Margolskee RF, Dyer J, Kokrashvili Z, et al. T1R3 and gustducin in gut sense 89 Powell DR, Smith M, Greer J, et al. LX4211 increases serum glucagon-like peptide
sugars to regulate expression of Na+-glucose cotransporter 1. Proc Natl Acad Sci 1 and peptide YY levels by reducing sodium/glucose cotransporter 1
U S A 2007;104:15075–80. (SGLT1)-mediated absorption of intestinal glucose. J Pharmacol Exp Ther
60 Reimann F, Gribble FM. Glucose-sensing in glucagon-like peptide-1-secreting cells. 2013;345:250–9.
Diabetes 2002;51:2757–63. 90 Miras AD, le Roux CW. Bariatric surgery and taste: novel mechanisms of weight
61 Reimann F, Habib AM, Tolhurst G, et al. Glucose sensing in L cells: a primary cell loss. Curr Opin Gastroenterol 2010;26:140–5.
study. Cell Metab 2008;8:532–9. 91 Batterham RL, ffytche DH, Rosenthal JM, et al. PYY modulation of cortical and
62 Nielsen LB, Ploug KB, Swift P, et al. Co-localisation of the Kir6.2/SUR1 channel hypothalamic brain areas predicts feeding behaviour in humans. Nature
complex with glucagon-like peptide-1 and glucose-dependent insulinotrophic 2007;450:106–9.
polypeptide expression in human ileal cells and implications for glycaemic control 92 Martin B, Dotson CD, Shin YK, et al. Modulation of taste sensitivity by GLP-1
in new onset type 1 diabetes. Eur J Endocrinol 2007;156:663–71. signaling in taste buds. Ann N Y Acad Sci 2009;1170:98–101.
63 Gorboulev V, Schurmann A, Vallon V, et al. Na(+)-D-glucose cotransporter SGLT1 93 Scholtz S, Miras AD, Chhina N, et al. Obese patients after gastric bypass surgery
is pivotal for intestinal glucose absorption and glucose-dependent incretin have lower brain-hedonic responses to food than after gastric banding. Gut
secretion. Diabetes 2012;61:187–96. Published Online First: 20 Aug 2013. doi:10.1136/gutjnl-2013-305008.
64 Gribble FM, Williams L, Simpson AK, et al. A novel glucose-sensing mechanism 94 Monsbakken KW, Vandvik PO, Farup PG. Perceived food intolerance in subjects
contributing to glucagon-like peptide-1 secretion from the GLUTag cell line. with irritable bowel syndrome– etiology, prevalence and consequences. Eur J Clin
Diabetes 2003;52:1147–54. Nutr 2006;60:667–72.
65 Parker HE, Adriaenssens A, Rogers G, et al. Predominant role of active versus 95 Shepherd SJ, Gibson PR. Fructose malabsorption and symptoms of irritable bowel
facilitative glucose transport for glucagon-like peptide-1 secretion. Diabetologia syndrome: guidelines for effective dietary management. J Am Diet Assoc
2012;55:2445–55. 2006;106:1631–9.
66 Mace OJ, Affleck J, Patel N, et al. Sweet taste receptors in rat small intestine 96 Shepherd SJ, Parker FC, Muir JG, et al. Dietary triggers of abdominal symptoms in
stimulate glucose absorption through apical GLUT2. J Physiol 2007;582:379–92. patients with irritable bowel syndrome: randomized placebo-controlled evidence.
67 Daly K, Al-Rammahi M, Arora DK, et al. Expression of sweet receptor components Clin Gastroenterol Hepatol 2008;6:765–71.
in equine small intestine: relevance to intestinal glucose transport. Am J Physiol 97 Barrett JS, Gearry RB, Muir JG, et al. Dietary poorly absorbed, short-chain
Regul Integr Comp Physiol 2012;303:R199–208. carbohydrates increase delivery of water and fermentable substrates to the
68 Li X, Li W, Wang H, et al. Pseudogenization of a sweet-receptor gene accounts for proximal colon. Aliment Pharmacol Ther 2010;31:874–82.
cats’ indifference toward sugar. PLoS Genet 2005;1:27–35. 98 Ong DK, Mitchell SB, Barrett JS, et al. Manipulation of dietary short chain
69 Buddington RK, Chen JW, Diamond JM. Dietary regulation of intestinal carbohydrates alters the pattern of gas production and genesis of symptoms in
brush-border sugar and amino acid transport in carnivores. Am J Physiol irritable bowel syndrome. J Gastroenterol Hepatol 2010;25:1366–73.
1991;261:R793–801. 99 Yajima T. Luminal propionate-induced secretory response in the rat distal colon in
70 Hirasawa A, Tsumaya K, Awaji T, et al. Free fatty acids regulate gut incretin vitro. J Physiol 1988;403:559–75.
glucagon-like peptide-1 secretion through GPR120. Nat Med 2005;11:90–4. 100 Ruppin H, Bar-Meir S, Soergel KH, et al. Absorption of short-chain fatty acids by
71 Dotson CD, Zhang L, Xu H, et al. Bitter taste receptors influence glucose the colon. Gastroenterology 1980;78:1500–7.
homeostasis. PloS one 2008;3:e3974. 101 Binder HJ, Mehta P. Short-chain fatty acids stimulate active sodium and chloride
72 Oya M, Kitaguchi T, Pais R, et al. The G protein-coupled receptor family C group 6 absorption in vitro in the rat distal colon. Gastroenterology 1989;96:989–96.
subtype A (GPRC6A) receptor is involved in amino acid-induced glucagon-like 102 Tazoe H, Otomo Y, Kaji I, et al. Roles of short-chain fatty acids receptors, GPR41
peptide-1 secretion from GLUTag cells. J Biol Chem 2013;288:4513–21. and GPR43 on colonic functions. J Physiol Pharmacol 2008;59(Suppl 2):251–62.
73 Brown AJ, Goldsworthy SM, Barnes AA, et al. The Orphan G protein-coupled 103 Cherbut C, Aube AC, Blottiere HM, et al. Effects of short-chain fatty acids on
receptors GPR41 and GPR43 are activated by propionate and other short chain gastrointestinal motility. Scand J Gastroenterol Suppl 1997;222:58–61.
carboxylic acids. J Biol Chem 2003;278:11312–19. 104 Fukumoto S, Tatewaki M, Yamada T, et al. Short-chain fatty acids stimulate colonic
74 Karaki S, Tazoe H, Hayashi H, et al. Expression of the short-chain fatty acid transit via intraluminal 5-HT release in rats. Am J Physiol Regul Integr Comp
receptor, GPR43, in the human colon. J Mol Histol 2008;39:135–42. Physiol 2003;284:R1269–76.
75 Karaki S, Mitsui R, Hayashi H, et al. Short-chain fatty acid receptor, GPR43, is 105 Sykaras AG, Demenis C, Case RM, et al. Duodenal enteroendocrine I-cells contain
expressed by enteroendocrine cells and mucosal mast cells in rat intestine. Cell mRNA transcripts encoding key endocannabinoid and fatty acid receptors. PLoS
Tissue Res 2006;324:353–60. One 2012;7:e42373.

Depoortere I. Gut 2013;0:1–12. doi:10.1136/gutjnl-2013-305112 11


Downloaded from http://gut.bmj.com/ on April 8, 2016 - Published by group.bmj.com

Recent advances in basic science

106 Mitsui R, Ono S, Karaki S, et al. Neural and non-neural mediation of 122 Biesiekierski JR, Peters SL, Newnham ED, et al. No effects of gluten in patients
propionate-induced contractile responses in the rat distal colon. Neurogastroenterol with self-reported non-celiac gluten sensitivity after dietary reduction of
Motil 2005;17:585–94. fermentable, poorly absorbed, short-chain carbohydrates. Gastroenterology
107 Ford AC, Brandt LJ, Young C, et al. Efficacy of 5-HT3 antagonists and 5-HT4 2013;145:320–8, e3.
agonists in irritable bowel syndrome: systematic review and meta-analysis. Am J 123 Vazquez-Roque MI, Camilleri M, Smyrk T, et al. A controlled trial of gluten-free
Gastroenterol 2009;104:1831–43; quiz 44. diet in patients with irritable bowel syndrome-diarrhea: effects on bowel frequency
108 Cremon C, Carini G, Wang B, et al. Intestinal serotonin release, sensory neuron and intestinal function. Gastroenterology 2013;144:903–11, e3.
activation, and abdominal pain in irritable bowel syndrome. Am J Gastroenterol 124 Reeds PJ, Burrin DG, Stoll B, et al. Intestinal glutamate metabolism. J Nutr
2011;106:1290–8. 2000;130:978S–82S.
109 Van Der Veek PP, Biemond I, Masclee AA. Proximal and distal gut hormone 125 Toyomasu Y, Mochiki E, Yanai M, et al. Intragastric monosodium L-glutamate
secretion in irritable bowel syndrome. Scand J Gastroenterol 2006;41:170–7. stimulates motility of upper gut via vagus nerve in conscious dogs. Am J Physiol
110 Pimentel M, Chow EJ, Lin HC. Eradication of small intestinal bacterial overgrowth Regul Integr Comp Physiol 2010;298:R1125–35.
reduces symptoms of irritable bowel syndrome. Am J Gastroenterol 2000;95:3503–6. 126 Zai H, Kusano M, Hosaka H, et al. Monosodium L-glutamate added to a
111 Vanner S. The small intestinal bacterial overgrowth. Irritable bowel syndrome high-energy, high-protein liquid diet promotes gastric emptying. Am J Clin Nutr
hypothesis: implications for treatment. Gut 2008;57:1315–21. 2009;89:431–5.
112 Posserud I, Stotzer PO, Bjornsson ES, et al. Small intestinal bacterial overgrowth in 127 Boutry C, Matsumoto H, Airinei G, et al. Monosodium glutamate raises antral
patients with irritable bowel syndrome. Gut 2007;56:802–8. distension and plasma amino acid after a standard meal in humans. Am J Physiol
113 van der Schaar PJ, van Hoboken E, Ludidi S, et al. Effect of cholecystokinin on Gastrointest Liver Physiol 2011;300:G137–45.
rectal motor and sensory function in patients with irritable bowel syndrome and 128 Uneyama H, Niijima A, San Gabriel A, et al. Luminal amino acid sensing in the rat
healthy controls. Colorectal Dis 2013;15:e29–34. gastric mucosa. Am J Physiol Gastrointest Liver Physiol 2006;291:G1163–70.
114 Kellow JE, Phillips SF, Miller LJ, et al. Dysmotility of the small intestine in irritable 129 Kondoh T, Torii K. Brain activation by umami substances via gustatory and visceral
bowel syndrome. Gut 1988;29:1236–43. signaling pathways, and physiological significance. Biol Pharm Bull
115 Kaji I, Karaki S, Tanaka R, et al. Density distribution of free fatty acid receptor 2 2008;31:1827–32.
(FFA2)-expressing and GLP-1-producing enteroendocrine L cells in human and rat 130 Page AJ, Young RL, Martin CM, et al. Metabotropic glutamate receptors inhibit
lower intestine, and increased cell numbers after ingestion of mechanosensitivity in vagal sensory neurons. Gastroenterology 2005;128:402–10.
fructo-oligosaccharide. J Mol Histol 2011;42:27–38. 131 Simren M, Mansson A, Langkilde AM, et al. Food-related gastrointestinal
116 Cherbut C, Ferrier L, Roze C, et al. Short-chain fatty acids modify colonic motility symptoms in the irritable bowel syndrome. Digestion 2001;63:108–15.
through nerves and polypeptide YY release in the rat. Am J Physiol 1998;275: 132 Bisschops R, Karamanolis G, Arts J, et al. Relationship between symptoms and
G1415–22. ingestion of a meal in functional dyspepsia. Gut 2008;57:1495–503.
117 Samuel BS, Shaito A, Motoike T, et al. Effects of the gut microbiota on host 133 Mullan A, Kavanagh P, O’Mahony P, et al. Food and nutrient intakes and eating
adiposity are modulated by the short-chain fatty-acid binding G protein-coupled patterns in functional and organic dyspepsia. Eur J Clin Nutr 1994;48:97–105.
receptor, Gpr41. Proc Natl Acad Sci USA 2008;105:16767–72. 134 Pilichiewicz AN, Feltrin KL, Horowitz M, et al. Functional dyspepsia is associated
118 Bindels LB, Dewulf EM, Delzenne NM. GPR43/FFA2: physiopathological relevance with a greater symptomatic response to fat but not carbohydrate, increased fasting
and therapeutic prospects. Trends Pharmacol Sci 2013;34:226–32. and postprandial CCK, and diminished PYY. Am J Gastroenterol
119 Sina C, Gavrilova O, Forster M, et al. G protein-coupled receptor 43 is essential for 2008;103:2613–23.
neutrophil recruitment during intestinal inflammation. J Immunol 2009;183:7514–22. 135 Chua AS, Keeling PW. Cholecystokinin hyperresponsiveness in functional
120 Barone S, Fussell SL, Singh AK, et al. Slc2a5 (Glut5) is essential for the absorption dyspepsia. World J Gastroenterol 2006;12:2688–93.
of fructose in the intestine and generation of fructose-induced hypertension. J Biol 136 Barbera R, Feinle C, Read NW. Abnormal sensitivity to duodenal lipid infusion in
Chem 2009;284:5056–66. patients with functional dyspepsia. Eur J Gastroenterol Hepatol 1995;7:1051–7.
121 Biesiekierski JR, Newnham ED, Irving PM, et al. Gluten causes gastrointestinal 137 Raben A, Richelsen B. Artificial sweeteners: a place in the field of functional
symptoms in subjects without celiac disease: a double-blind randomized foods? Focus on obesity and related metabolic disorders. Curr Opin Clin Nutr
placebo-controlled trial. Am J Gastroenterol 2011;106:508–14; quiz 15. Metab Care 2012;15:597–604.

12 Depoortere I. Gut 2013;0:1–12. doi:10.1136/gutjnl-2013-305112


Downloaded from http://gut.bmj.com/ on April 8, 2016 - Published by group.bmj.com

Taste receptors of the gut: emerging roles in


health and disease
Inge Depoortere

Gut published online October 16, 2013

Updated information and services can be found at:


http://gut.bmj.com/content/early/2013/10/16/gutjnl-2013-305112

These include:

References This article cites 136 articles, 50 of which you can access for free at:
http://gut.bmj.com/content/early/2013/10/16/gutjnl-2013-305112#BIBL

Email alerting Receive free email alerts when new articles cite this article. Sign up in the
service box at the top right corner of the online article.

Topic Articles on similar topics can be found in the following collections


Collections GUT Recent advances in basic science (80)
Irritable bowel syndrome (326)

Notes

To request permissions go to:


http://group.bmj.com/group/rights-licensing/permissions

To order reprints go to:


http://journals.bmj.com/cgi/reprintform

To subscribe to BMJ go to:


http://group.bmj.com/subscribe/

Das könnte Ihnen auch gefallen