Sei sulla pagina 1di 22

Downloaded from gut.bmj.com on March 24, 2014 - Published by group.bmj.com Downloaded from gut.bmj.

.com on March 24, 2014 - Published by group.bmj.com


179 179







Recent advances in basic science


Taste receptors of the gut: emerging roles in health
and disease
Inge Depoortere
Correspondence to
Professor Inge Depoortere, Gut
Peptide Research Lab,
Translational Research Center
for Gastrointestinal Disorders,
University of Leuven,
Gasthuisberg O&N1, box 701,
Leuven 3000, Belgium; inge.
depoortere@med.kuleuven.be

Received 29 July 2013
Revised 16 September 2013
Accepted 25 September 2013
Published Online First
16 October 2013














































To cite: Depoortere I. Gut
2014;63:179190.
ABSTRACT
Recent progress in unravelling the nutrient-sensing
mechanisms in the taste buds of the tongue has triggered
studies on the existence and role of chemosensory cells in
the gut. Indeed, the gastrointestinal tract is the key interface
between food and the human body and can sense basic
tastes in much the same way as the tongue, through the
use of similar G-protein-coupled taste receptors. These
receptors taste the luminal content and transmit signals
that regulate nutrient transporter expression and nutrient
uptake, and also the release of gut hormones and
neurotransmitters involved in the regulation of energy and
glucose homeostasis. Hence,
they play a prominent role in the communication between
the lumen, epithelium, smooth muscle cells, afferent
nerve bres and the brain to trigger adaptive responses that
affect gastrointestinal function, food intake and glucose
metabolism. This review summarises how sensing of
nutrients by taste receptors along the gut plays a key role in
the process of digestion, and how disturbances or
adaptations of these chemosensory signalling pathways may
contribute to the induction or resolution of a number of
pathological conditions related to diabetes, obesity, or diet-
induced symptom generation in irritable bowel syndrome.
Targeting these receptors may represent a promising novel
route for the treatment of a number of these diseases.



INTRODUCTION
We choose to eat for many reasons, including to
satisfy our hunger, to invite pleasant sensory
experiences, and as a social ritual. However, we
need to eat to acquire nutrients essential for life
and health. Not surprisingly, the digestive system
contains a diverse array of detectors that help to
regulate ingestive decisions, impact nutrient assimi-
lation, avoid or neutralise toxins, and elicit
complex neural and endocrine responses that affect
metabolism, gastrointestinal (GI) transit, satiation
and satiety. Strikingly, many of the G protein-
coupled receptors (GPCRs) that detect nutrients
and toxins in the oral cavity and function as taste
receptors there, also subserve important functions
throughout the GI tract. Activation of these recep-
tors triggers the release of neurotransmitters (eg,
ATP) that will excite primary sensory afferent bres
and interact with neighbouring presynaptic cells to
relay information to the hindbrain.
1
A similar
system of highly orchestrated interactions also oper-
ates in the gut and further points to the functional
similarity of lingual and intestinal cells. I will
review the state of our knowledge about taste
receptor function along the entirety of the alimen-
tary canal and discuss the implications of these
functions for human physiology and disease.
Downloaded from gut.bmj.com on March 24, 2014 - Published by group.bmj.com Downloaded from gut.bmj.com on March 24, 2014 - Published by group.bmj.com
180 180







Key messages

The gut
tastes what
we eat
bitter, sweet,
umami,
fatin much
the same
way as the
tongue
through the
use of
similar taste
receptors
and
chemosensor
y signalling
pathways
In health, taste
receptors sense
nutrients from
either a luminal
or blood-borne
direction and
transmit signals
that control the
secretion of gut
hormones and
the expression
of nutrient
transporters to
maintain energy
and glucose
homeostasis
and
gastrointestinal
function
In disease,
disturbances or
adaptations in
the expression or
sensitivity of
these taste
receptors and
their signalling
pathways may
affect digestive
behaviour and
metabolism
This is a new and
emerging eld,
future studies
aimed at a better
understanding of
how the sensing
of nutrients in
the gut is nely tuned
by different taste receptors in health and
disease, should help us to dene novel
drug targets




TASTE RECEPTORS AND
GUSTATORY FUNCTION
The taste system detects compounds that elicit
at least ve perceptual qualities: sweet, umami,
bitter, sour and salty. The latter two are largely
mediated by ion channels, and will not be
discussed further here. By contrast, sweet, umami
and bitter-tasting stimuli are detected by members
of two GPCR fam- ilies, the taste 1 receptor
family (TAS1R) and the taste 2 receptor family
(TAS2R), that are expressed in subpopulations of
taste bud cells ( gure 1). Subtypes of the
TAS1R family heterodimerise to detect sweet
(TAS1R2-TAS13) and umami (TAS1R1-
TAS1R3).
2 3
Animals that lack these subu- nits are
decient in their ability to taste umami and/ or
sweet stimuli.
4
The presence of multiple binding
sites on these receptors can explain the
synergistic effects of 5
0
-ribonucleotides and
glutamate in umami taste, or the ability of
many chemically diverse compoundsincluding
sugars, non-caloric sweeteners, D-amino acids
and some proteinsto elicit a sweet taste.
Consistent with their proposed evolutionary need
to detect foods rich in nutrients, the umami and
sweet receptors are low-afnity receptors, with
EC
50
s in the millimolar range for their most
common natural ligands.
Alternatively, we are quite sensitive to
bitter- tasting compounds. While this is
important for
Downloaded from gut.bmj.com on March 24, 2014 - Published by group.bmj.com Downloaded from gut.bmj.com on March 24, 2014 - Published by group.bmj.com
181 181







Recent advances in basic science



Figure 1 Simplied 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 Ca
2+
, activation of TRPM5, depolarisation, activation of voltage-gated Na
+
channels (VGNC) and release of ATP which
activates purinergic receptors on afferent nerve bres 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
by TAS2Rs.
6
The human genome encodes 25 different TAS2Rs,
which fall into three functional categories: specialists that
respond to one or two compounds; generalists that are highly
promiscuous in their stimulus responses; and those that are
intermediate in their selectivity.
7
Many compounds can activate
more than one receptor, and activation thresholds can vary
from millimolar to nanomolar levels. TAS2R polymorphisms are

Table 1 Overview of the different taste receptors involved in
nutrient sensing

Sweet taste receptors/transporters
TAS1R2-TAS1R3 Taste receptor, type 1, member 2-taste
receptor, type 1, member 3
SGLT1 Sodium-dependent glucose transporter 1
GLUT Glucose transporter
Amino acid/peptide taste receptors
quite common in human populations, and can have pronounced
effects on the ability of individuals to detect certain bitter
TAS1R1-TAS1R3 (umami,
aliphatic amino acids)
Taste receptor, type 1, member 1-taste
receptor, type 1, member 3
compounds.
8
CaSR (aromatic amino acids) Calcium sensing receptor
Other GPCRs have been implicated in the detection of gusta-
tory stimuli. For example, metabotropic glutamate receptors,
GPRC6A (basic and small
neutral amino acids)
G-protein coupled receptor family C group 6
member A
specically mGluR1 and mGluR4, may contribute to amino
acid taste.
9
The Ca
2+
-sensing receptor (CaSR) and GPRC6A
have been proposed to mediate Ca
2+
taste and also amino acid
taste in extraoral tissues such as the gut.
10 11
Two receptors for
long-chain fatty acids (LCFAs), FFAR1 (a.k.a., GPR40) and
GPR120 have been localised to taste cells and contribute to oro-
sensory responses to fats (whether or not fat can be considered
as the sixth taste is the subject of ongoing investigation).
12

Additionally, receptors for short-chain fatty acids (SCFA),
FFAR2 and FFAR3, have been detected outside the oral cavity.
11

An as yet unidentied receptor may mediate the taste of polysac-
charides. An overview of the different taste receptors involved
in nutrient sensing is represented in table 1.
TAS1Rs and TAS2Rs are found in Type II taste cells, a mor-
phological class of microvillar taste bud cells. Activation of a
gustatory G-protein (eg, gustducin) coupled to these GPCRs
results in the activation of phospholipase C 2 leading to
mGluR (umami) Metabotropic glutamate receptor
GPR92 ( protein hydrolysates) G-protein coupled receptor 92
Bitter taste receptors
TAS2R Taste receptor type 2
Free fatty acid receptors
Long-chain/medium-chain fatty acids
GPR120 G-protein coupled receptor 120
FFAR1 (also known as GPR40) Free fatty acid receptor 1
Short-chain fatty acids
FFAR2 (also known as GPR43) Free fatty acid receptor 2
FFAR3 (also known as GPR41) Free fatty acid receptor 3

CaSR, calcium sensing receptor; FFAR1, free fatty acid receptor 1; FFAR2, free fatty
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,
G-protein coupled receptor family C group 6; mGluR, metabotropic glutamate
receptor; SGLT1, sodium-dependent glucose transporter 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.
Downloaded from gut.bmj.com on March 24, 2014 - Published by group.bmj.com Downloaded from gut.bmj.com on March 24, 2014 - Published by group.bmj.com
182 182







Recent advances in basic science
inositoltriphosphate-mediated release of intracellular Ca
2+
and
activation of the transient receptor potential cation channel M5
(TRPM5) ( gure 1). These events induce membrane depolarisa-
tion, action potential generation, and the release of ATP, which
acts on purinergic receptors to activate gustatory afferents
leading to activation of brain centres involved in taste percep-
tion. Sweet, umami and bitter taste receptors are differentially
expressed within the Type II taste cells.
13
This segregation is a
key component of taste quality coding and hedonic discrimin-
ation in the gustatory periphery. The extent to which these
various taste transduction components are colocalised in gut
cells is unclear, but surely has consequences for how they
impact GI function.

ROLE OF TASTE RECEPTORS IN NUTRIENT DETECTION AND
RESPONSE DURING DIGESTION
Gut peptides produced in taste cells of the tongue
Taste cells also express a number of bioactive peptides best
known for their roles in metabolism, feeding and satiety, includ-
ing: glucagon-like peptide-1 (GLP-1), glucagon, neuropeptide Y,
peptide YY (PYY), cholecystokinin (CCK), vasoactive intestinal
peptide and ghrelin.
14
The functions of these peptides in taste
buds are not fully understood, but at least some may act to
modulate the responsiveness of the peripheral gustatory appar-
atus to certain taste stimuli. Interestingly, stimuli representing
different taste qualities promote the release of different peptide
combinations, suggesting that these peptides could also play a
role in quality coding.
15

Cognate receptors for all the peptides expressed in taste cells
can be found on taste cells or on the adjacent afferent nerve
bres.
14
Therefore, these peptides almost certainly have auto-
crine and paracrine functions within the taste bud and, in some
cases, could function as cotransmitters with ATP. However, it is
unclear whether taste bud peptides also reach the bloodstream
and exert endocrine effects. Furthermore, peptide receptors
expressed on taste cells may be targets of circulating peptides
produced in the gut, adipose tissue, or other tissues. For
example, systemic leptin has been shown to decrease sweet taste
responsiveness through actions on leptin receptors present on a
subset of taste cells.
16
Thus, postingestive nutrient responses
could feed back onto the peripheral gustatory apparatus to
modulate its responsiveness to subsequent foods.

Nutrient sensing in the stomach
Proximal stomach
Upon ingestion of food, the proximal stomach relaxes to accom-
modate large amounts of food without increasing intragastric
pressure. Tension-sensitive mechanoreceptors are activated by
the arrival of food and relay their information via the vagus to
the hindbrain to induce sensations of satiation.
17
Nutrient
sensing in the stomach has been considered to be a less import-
ant factor, but the role of chemosensory receptors should not be
ignored. Indeed, recent studies showed that intragastric adminis-
tration of the bitter agonist, denatonium benzoate (DB), inhib-
ited gastric accommodation to nutrient drink infusion and
tended to increase satiation scores.
18
The contractile response
induced by DB was mimicked in vitro in smooth muscle strips
from the mouse fundus and was partially mediated via the gusta-
tory G-protein subunit, -gustducin, and involved the release of
Ca
2+
from intracellular and extracellular stores.
19

The proximal stomach is also the major site of production of
the orexigenic hormone, ghrelin, which is considered as a physio-
logical meal initiator.
20
How the reduction of plasma ghrelin
during a meal might participate in the induction of satiation is
incompletely explored. The fact that ghrelin levels are suppressed
strongly by ingested proteins, weakly by proteins and biphasically
by carbohydrates, suggests that the ghrelin P/D1 cells contain the
machinery to sense nutrients.
21
Recent immunouorescence
studies conrmed that the ghrelin cell is colocalised with the gus-
tatory G-proteins, -gustducin and -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
Downloaded from gut.bmj.com on March 24, 2014 - Published by group.bmj.com Downloaded from gut.bmj.com on March 24, 2014 - Published by group.bmj.com
183 183






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.
Downloaded from gut.bmj.com on March 24, 2014 - Published by group.bmj.com Downloaded from gut.bmj.com on March 24, 2014 - Published by group.bmj.com
184 184







Recent advances in basic science

umami receptor subunit, TAS1R3, and the free fatty acid-sensing
receptor, GPR120.
2224
It remains to be investigated whether the
postprandial suppression of ghrelin by nutrients is indeed
mediated via these receptors. However, the rst functional
studies showed that -gustducin is involved in the effect of bitter
compounds on ghrelin release and in sensing of medium-chain
fatty acids in the diet necessary for the octanoylation of
ghrelin.
22 23
Additionally, GPR120 seems to play a role in the
lipid-sensing cascade of the ghrelin cell.
23
The distribution of
taste receptors involved in nutrient sensing in endocrine cells
along the gut is summarised in gure 2.

Distal stomach
The arrival of food in the stomach stimulates the gastric phase
of acid, pepsinogen and mucus production. The two principal
triggers are distension of the stomach, which initiates local
myenteric and vago-vagal reexes, and the chemical content of
the food. In the stomach, sensing of protein breakdown pro-
ducts is important as they activate the release of two important
regulators of pepsinogen and acid secretion: gastrin (G-cells)
and somatostatin (D-cells). Indeed, peptone has been shown to
affect gastrin and somatostatin secretion in rats.
25
Recent studies
showed that mouse and pig antral G-cells and a subpopulation
of D-cells express GPR92, a receptor activated by dietary
protein hydrolysates.
26
In both endocrine cell types, GPR92 is
coupled to different signalling pathways. It is therefore conceiv-
able that GPR92 may play an important role in adjusting the
release of gastrin and somatostatin in response to protein digests
in the chyme. In addition, G and D-cells express the amino acid
taste receptor GPRC6A and the CaSR that act in concert with
each other to sense a broad spectrum of amino acids ranging
from basic and small neutral (GPRC6A) to aromatic (L-Phe)
amino acids and Ca
2+
.
27 28
The stimulation of gastrin after
gavage with L-Phe, peptone, Ca
2+
and a neutralising buffer was
abolished in CaSR
/
mice suggesting a regulatory role for this
receptor in gastrin release.
29
The TAS1R family is not important
for G-cell sensing of protein and amino acids since TAS1R3 is
not expressed on G-cells and the gastrin response to peptone is
not abolished in TAS1R1/3
/
mice.
28 29
Additionally, L-Phe can
induce acid secretion independent of hormonal stimulation via
activation of the CaSR on parietal cells.
30

In the distal stomach, the chyme is mixed with the digestive
secretions and ground by powerful peristaltic contractions. The
process of emptying starts and the rate is dependent on the
gastric volume and the chemical nature of the gastric contents.
The major control mechanism for gastric emptying involves
duodenal gastric feedback and the emphasis shifts towards
satiety mechanisms.
When the meal is emptied from the stomach, the upper GI
motility pattern changes from a digestive to an interdigestive
pattern: the migrating motor complex, characterised by a group
of strong phasic contractions migrating distally. Phase 3 contrac-
tions, the most vigorous contractions of the migrating motor
complex (MMC), originating in the stomach, are considered as
a hunger signal. Arrival of nutrients will disrupt this pattern.
Infusion of high doses of amino acids in healthy volunteers
shortens the duration of the MMC length and suppresses antral
phase 3 activity.
31
Intragastric administration of the bitter com-
pound, DB, in the fasted state was shown to decrease antral but
not duodenal motility and to shift the origin of phase 3 from
the antrum to the duodenum. This was accompanied by a sig-
nicant decrease in hunger scores.
32
In both cases, the mechan-
isms involved are unknown but may involve activation of taste
receptors on endocrine cells, smooth muscle cells and/or nerves.
Downloaded from gut.bmj.com on March 24, 2014 - Published by group.bmj.com Downloaded from gut.bmj.com on March 24, 2014 - Published by group.bmj.com
185 185






Whether any stimulus that suppresses or induces gastric phase 3
activity inhibits or stimulates hunger, respectively, requires
further investigation. Furthermore, recent studies showed that
gastric phase 3 activity was less frequently observed in obese
patients, and was associated with fewer hunger peaks suggesting
that the absence of generation of gastric MMCs may be
pro-anorexigenic and may also represent a compensatory mech-
anism to reduce hunger feelings in obesity.
33
The motilin
agonist, erythromycin, known to induce gastric phase 3,
restored the association between gastric phase 3 and hunger
peaks in obese patients.

Nutrient sensing in the small intestine
The small intestine is the major site of digestion and absorption
in the GI tract. In the small intestine, carbohydrates, fats and
protein digestion products, as well as osmolarity changes and
physical distention activate inhibitory neural and endocrine
pathways that signal the stomach to delay emptying and to help
limit ingestion by enhancing gastric mechanoreceptor stimula-
tion. However, experiments demonstrated that intestinal nutri-
ent infusions inhibit food intake during sham feeding when
gastric contents were drained via an open gastric cannula to
obviate the gastric lling effects on food intake.
34
Thus, while
intestinal signals and gastric emptying interact to control food
intake, a delay of gastric emptying is not required for intestinal
signals to elicit satiation. The small intestine also sends endo-
crine satiety signals to the hypothalamus, either directly, via the
bloodstream and across the incomplete blood-brain barrier to
the arcuate nucleus, or indirectly, through the activation of the
vagus nerve. It is clear that the vagal-brainstem-hypothalamic
pathway plays a major role in the effect of the satiation hor-
mones, CCK and GLP-1, on food intake. Their receptors are
expressed on vagal afferents, they increase vagal afferent ring
and induce cfos expression in the nucleus of the solitary tract
after intraperitoneal injection.
3539
Furthermore the inhibition
of food intake by exogenous GLP-1 and CCK is blocked by
vagotomy and brainstem-hypothalamic pathway
transectioning.
40 41


Duodenum
In the duodenum, fat, protein hydrolysates and amino acids
stimulate the secretion of CCK from I-cells. This hormone then
acts to slow gastric emptying and increase satiety. In STC-1
cells, a mouse enteroendocrine-like cell line, LCFAs induce
CCK secretion through the lipid sensor GPR120 and the trans-
duction channel TRPM5.
42 43
However, cell lines are incom-
pletely validated as accurate models of native I-cells.
Establishing cultures from endocrine cells remained a challenge
for many years but the generation of transgenic mice with I cell-
specic expression of a uorescent protein, allowed the isolation
of a pure I-cell population by uorescence-activated cell sorting.
Native I-cells are also immunoreactive for FFAR1, and the effect
of linolenic acid on the secretion of CCK from isolated I-cells
was abolished in cells from FFAR1
/
mice.
44 45
Thus, GPR120
and FFAR1 appear to mediate lipid-induced CCK secretion.
Amino acids and protein hydrolysates similarly appear to use
multiple receptor types to stimulate CCK secretion. In humans,
administration of L-Phe increased CCK secretion and reduced
food intake.
46
Two amino acid-responsive receptors,
TAS1R1-TAS1R3 and the CaSR, are expressed in I-cells, and
implicated in CCK release.
47
For example, Phe-dependent,
Leu-dependent and Glu-dependent CCK release is dependent
on TAS1R1-TAS1R3, while the CaSR mediates Phe-induced and
Trp-induced responses in mouse intestinal tissue explants.
47
Downloaded from gut.bmj.com on March 24, 2014 - Published by group.bmj.com Downloaded from gut.bmj.com on March 24, 2014 - Published by group.bmj.com
186 186







Recent advances in basic science

Studies in vitro (STC-1 cells) and in native I-cells also showed
an important role for the CaSR in the effects of aromatic amino
acids on CCK secretion.
4850
Protein hydrolysates stimulate the
release of CCK via GPR92 in STC-cells.
51

Bitter tastants can also induce CCK release from STC-1 cells
by affecting Ca
2+
inux.
52 53

Glucose-induced CCK secretion in humans, although perhaps
not a major phenomenon, was unaffected by the sweet taste
receptor inhibitor lactisole.
54
Immunolocalisation studies con-
rmed that the sweet taste receptor-specic subunit, TAS1R2, is
not coexpressed with CCK.
47


Jejunum-ileum
GLP-1 and glucose-dependent insulinotropic polypeptide (GIP)
are incretin hormones that augment insulin secretion after inges-
tion of a meal. They are secreted from L-cells and K-cells,
respectively. The mechanisms by which L-cells couple glucose
detection to GLP-1 secretion have been controversial, but some
clarity has begun to emerge ( gure 3). Several lines of evidence
support that the sweet taste receptor, TAS1R2-TAS1R3, and the
taste G-protein, -gustducin, are required for glucose-stimulated
GLP-1 secretion from the small intestine. First, these three pro-
teins are present in human and rodent L-cells of the small intes-
tine.
5557
Second, several sweet taste receptor agonists
sucrose, glucose, fructose and sucralose can elicit GLP-1



Figure 3 Mechanisms involved in glucose-stimulated GLP-1 release.
Glucose can stimulate gut peptide secretion by (1) binding to the sweet
taste receptor heterodimer, TAS1R2-TAS1R3, coupling via the G-protein,
gustducin, to increase intracellular Ca
2+
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
secretion from mouse jejunal and ileal explants, and/or mouse
(GLUTag) and human (NCI-H716) enteroendocrine cell
lines.
56 58 59
Third, glucose-stimulated GLP-1 secretion is nearly
or fully abolished in -gustducin
/
and TAS1R2
/
and
TAS1R3
/
mice.
56 58
Fourth, sweet taste receptor inhibitors, or
RNA silencing of -gustducin, reduce sucralose-stimulated
GLP-1 secretion from GLUTag and NCI-H716 cells.
56 59

Finally, the sweet taste receptor inhibitor, lactisole, reduced sys-
temic levels of GLP-1 and PYY after intragastric or intraduode-
nal administration of glucose in humans.
54 55

Several studies also indicated an important role for the
Na
+
-glucose cotransporter SGLT1 and the ATP-sensitive K
+
(K
ATP
) channel. For example, the K
ATP
channel blocker tolbuta-
mide enhances GLP-1 secretion from GLUTag cells or from iso-
lated L-cells from upper small intestine or colon.
60 61

Furthermore, immunoreactivity for one K
ATP
channel subunit,
Kir6.2, has been localised to L-cells.
62
By contrast, a related
K
ATP
channel blocker, glibenclamide, had no effect on GLP-1
secretion from mouse ileum explants, but did induce GLP-1
secretion from colonic explants, suggesting that this channel is
involved in GLP-1 secretion from L-cells of the large, but not
small intestine.
58
The evidence for a role of SGLT1 is more
intriguing, as SGLT1
/
mice exhibit reduced GLP-1 and GIP
levels after glucose gavage.
63
A role for SGLT1 in
glucose-induced GLP-1 release has been conrmed 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
-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
-linolenic acid promoted GLP-1 secretion in vivo.
70
In STC-1
cells, however, siRNA against GPR120, but not FFAR1,
impaired -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
homeostasis.
71

GPRC6A was recently shown to act as an amino acid sensor
in GLUTag that enhances GLP-1 secretion.
72



Nutrient sensing in the colon
voltage-gated Ca
2+
channels and inux of Ca
2+
. 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;
GLP-1, glucagon-like peptide; K
ATP
, ATP-sensitive potassium channel;
PLC2, phospholipase C 2; TAS1R2, taste receptor type 1 member 2;
TAS1R3, taste receptor type 1 member 3; SGLT1, sodium-dependent
glucose transporter 1.
Downloaded from gut.bmj.com on March 24, 2014 - Published by group.bmj.com Downloaded from gut.bmj.com on March 24, 2014 - Published by group.bmj.com
187 187






mucus and absorb remaining water and electrolytes from the
food residue before defecation. The colon exhibits segmental
movements (haustrations) and phasic propulsive
contractions. The intestinal ora performs fermentation
reactions that produce SCFA and atus.
Downloaded from gut.bmj.com on March 24, 2014 - Published by group.bmj.com Downloaded from gut.bmj.com on March 24, 2014 - Published by group.bmj.com
188 188







Recent advances in basic science

SCFAs ( propionate, acetate and butyrate) are sensed in the
lumen by the free fatty acid receptors, FFAR2 and FFAR3.
73
In
the human colon, FFAR2 immunoreactive cells were colocalised
with PYY-containing L-cells but not with 5-HT containing endo-
crine cells.
74
In the rat colon, 5-HT-containing mucosal mast
cells were immunoreactive for FFAR2.
75
FFAR3-positive cells in
the human colon were less abundantly present than FFAR2 cells
and contained PYY but not 5-HT or FFAR2.
76
-gustducin Is
colocalised with several fatty acid receptors present on endo-
crine cells in the mouse colon and is required for
SCFA-mediated GLP-1 release from mucosal explants.
77
The
effect of SCFA on the release of GLP-1 was also conrmed in
isolated primary L-cells from mouse colon. In FFAR2
/
and
FFAR3
/
mice reduced SCFA-induced GLP-1 secretion was
observed in vitro and in vivo together with a parallel impair-
ment of glucose tolerance.
78

The functional role of TAS1R3 in L-cells of the human colon
is not clear.
55
GLP-1-secreting cells of the colon and rectum are
normally not responsive to sugars, suggesting no role for the
sweet taste receptor in hindgut GLP-1 secretion. Even when
these cells become glucose-responsive in association with carbo-
hydrate malabsorption, for instance after gastric bypass surgery,
this glucose sensitivity is sweet taste receptor independent (the
identity of the glucose sensor is unknown).

CONTRIBUTIONS OF TASTE RECEPTORS TO
PATHOPHYSIOLOGY: POSSIBLE TARGETS FOR THERAPY?
Glucose homeostasis and diabetes
Young et al
79 80
were the rst to compare the intestinal levels of
sweet taste receptors in patients with well-controlled type 2 dia-
betes to those without diabetes. Absolute levels of TAS1R2,
TAS1R3, -gustducin or TRPM5 transcripts in the duodenum
of healthy people, or patients with type 2 diabetes, were
unaffected by acute variations in glycaemia during fasting.
However, the intestinal sweet taste receptor system was highly
responsive to changes in luminal glucose. During euglycemia,
intraduodenal glucose infusion increased TAS1R2 transcript
levels in both groups but during hyperglycaemia TAS1R2
mRNA expression decreased in healthy volunteers but not in
diabetics. Levels of TAS1R3 did not change signicantly. The
TAS1R2 dysregulation in patients with type 2 diabetes may
potentially increase the risk of postprandial hyperglycaemia
since they exhibited increased glucose absorption during hyper-
glycaemia, as evidenced by an increase in the glucose absorption
marker 3-O-methyl-glucose (3-OMG), compared with healthy
subjects. As SGLT1 is responsible for the active transport of
luminal 3-OMG, these studies conrm that TAS1R2 regulates
glucose absorption via SGLT1. The positive association found
between luminal glucose-induced changes in some sweet taste
receptor transcripts and the secretion of GLP-1 and GIP under-
scores that sweet taste receptors do have a regulatory role in the
release of incretins. Although these ndings are supported by in
vitro studies, in vivo studies either in rodents or in humans
failed to show an effect of articial sweeteners on the release of
satiation hormones, thereby questioning the role of sweet taste
receptors in incretin release.
56 58 8183
Nevertheless, chronic
exposure of high-fat fed diabetic mice with the articial sweet-
ener, oligofructose, increased GLP-1 levels. Thus, the relative
importance of glucose transporters versus sweet taste receptors
in the release of incretins remains a matter of debate.
Glucose and insulin levels are remarkably normal in
TAS1R2
/
and TAS1R3
/
mice, despite the presence of sweet
taste ageusia in both genotypes. However, after an oral glucose
challenge TAS1R3
/
mice, but not TAS1R2
/
mice, showed
Downloaded from gut.bmj.com on March 24, 2014 - Published by group.bmj.com Downloaded from gut.bmj.com on March 24, 2014 - Published by group.bmj.com
189 189






increased blood glucose and decreased insulin levels. The
TAS1R3
/
phenotype was less severe after an intraperitoneal
injection of glucose, indicating that some, but not all, of the
glucose dysregulation results from the disruption of normal gut
physiology. These ndings are somewhat at variance with the
studies in patients with type 2 diabetes where a dysregulation of
TAS1R2 but not of TAS1R3 was observed during a luminal
glucose challenge and resulted in an increased glucose
absorption.
It is clear that the regulation of sweet taste receptors and
glucose transporters during different glycemic conditions is
complex. More studies using different models and conditions
are therefore warranted.

Gastric bypass
Gastric bypass surgery is one of the most effective methods for
inducing a marked and sustained weight loss in morbidly obese
patients. This procedure also results in amelioration or even
complete remission of type 2 diabetes mellitus independent
from weight loss.
84
The mechanisms involved are incompletely
understood but involve restriction, malabsorption and humoral
changes. Indeed, bypass surgery decreases the release of the
orexigenic hormone, ghrelin, and increases the release of sati-
ation hormones PYY and GLP-1. After gastric bypass surgery,
the contact of nutrients with much of the stomach, duodenum
and part of the jejunum is bypassed. It can be hypothesised that
nutrient sensors on the ghrelin cell are isolated from contact
with nutrients thereby probably affecting ghrelin release and,
consequently, energy and glucose homeostasis. Additionally, the
rapid delivery of undigested nutrients to the lower small intes-
tine may affect the regulation of taste receptors and/or glucose
transporters on L-cells, resulting in enhanced release of PYY and
GLP-1. Indeed, duodenaljejunal bypass surgery in a rat model
of type 2 diabetes decreased SGLT1 mRNA and
SGLT1-mediated transport in jejunum distal to the duodenojeju-
nostomy and improved diabetes independent from body weight
loss.
85
A similar observation was made in non-obese mice after
duodenojejunal bypass surgery.
86
Sweet taste receptors regulate
SGLT1 and a signicant decrease in TAS1R2 and TAS1R3
protein levels in the alimentary limb after gastric bypass have
been demonstrated in rats.
87
These results suggest that TAS1R3
antagonists or SGLT1 inhibitors may represent a promising
target for the treatment of obesity. In fact, LX4211, a dual intes-
tinal SGLT1/renal SGLT2 inhibitor increased GLP-1 levels and
PYY levels in patients with type 2 diabetes and improved blood
glucose levels and, therefore, mimicked the effects of gastric
bypass surgery.
88
These results are inconsistent with studies
showing that SGLT1 is required for glucose-mediated GLP-1
release by L-cells in vitro.
61 63
However, due to the carbohy-
drate malabsorption induced by SGLT1 inhibition, L-cells in the
colon also become glucose sensitive and increase GLP-1 levels.
89

The authors suggested that this may be mediated by SCFAs pro-
duced by colonic fermentation of unabsorbed glucose that inter-
act with FFARs on L-cells to induce GLP-1 and PYY release. A
K
ATP
channel-dependent mechanism involving an unknown
glucose sensor has also been suggested in glucose-induced
hindgut GLP-1 secretion during carbohydrate malabsorption.
58

After gastric bypass surgery, patients show a decreased prefer-
ence for sweet and fatty foods, but the factors involved are
unclear. The decision of what to eat is modulated by taste, olfac-
tion and oral textural perception. Taste, in particular, has an
important input into food preference because it has direct
effects on brain reward circuits that drive eating. It has been sug-
gested that the acuity for sweet taste increases after gastric
Downloaded from gut.bmj.com on March 24, 2014 - Published by group.bmj.com Downloaded from gut.bmj.com on March 24, 2014 - Published by group.bmj.com
190 190







Recent advances in basic science

bypass, potentially leading to increased intensity of percep-
tion.
90
It remains to be investigated whether endocrine cells in
the gut also show changes in sweet taste sensitivity after gastric
bypass surgery. Roux en-Y gastric bypass (RYGB) could further
reset the food reward system through changes in the release of
gut hormones, known to modify activity in brain reward
systems and dopaminergic signalling, but also known to modu-
late taste sensitivity in taste buds.
91 92
Indeed, a recent study
showed that obese patients, after RYGB, had lower brain-
hedonic responses to food than patients after gastric banding.
93

Additionally, aversive conditioning during the early postsurgical
period where unpleasant feelings (eg, dumping) associated with
particular foods can lead to conditioning of food aversion, may
also play a role.

Irritable bowel syndrome (IBS): FODMAPS and gluten
sensitivity
Patients with irritable bowel syndrome (IBS) claim that diet plays
a major role in triggering GI symptoms.
94
The most common
approaches to manage food intolerance in IBS include: (1) the
low-FODMAP ( fermentable, oligosaccharides disaccharides,
monosaccharides and polyols) diet, (2) the gluten-free diet, (3)
the elimination diet for food chemicals. How foods trigger func-
tional gut symptoms is unclear.

The low-FODMAP diet
Recent trials showed that dietary FODMAPs, especially fructose
and fructans, are dietary triggers for symptom generation, and
that restricting their level of intake might lead to durable
symptom improvement in patients with IBS.
95 96
Two
mechanisms of action have been put forward for the FODMAP
diet: (1) FODMAPs are poorly absorbed in the small intestine
and are osmotically active, thereby drawing uid through the
large bowel leading to diarrhoea
97
; (2) fermentation of the
FODMAPs in the colon will generate gases that may be incorpo-
rated in volatile end-products.
98
The increase in uid and gas
components will lead to luminal distension and may, in the pres-
ence of visceral hypersensitivity, induce bloating, atulence,
abdominal pain and motility disturbances.
In this review, we speculate that alterations in nutrient-sensing
mechanisms may also play a role in symptom generation in
patients with IBS.

Altered short-chain fatty acid sensing?
Fermentation of FODMAPs, such as fructans, in the colon will
also result in the production of SCFAs, known to affect local
ion secretion
99101
and colonic motility.
102 103
Indeed, intralum-
inal application of SCFA in the rat proximal colon accelerated
colonic transit by inducing the release of 5-HT that stimulates
5-HT
3
receptors located on vagal sensory bres, resulting in
muscle contraction via a vagal reex pathway involving the
release of ACh from the myenteric plexus ( gure 4).
104 105
The
role of 5-HT in SCFAs-induced contractions was conrmed in
contractility studies with smooth muscle strips in vitro.
106
In
fact, use of 5-HT
3
antagonists has been shown to improve
symptoms in patients with IBS.
107
We hypothesise that FFAR2
receptors on mucosal mast cells mediate the effect of SCFA on
5-HT release, since enterochromafn cells and smooth muscle
cells do not contain FFARs ( gure 4).
74 75
Cremon et al
108
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,
Downloaded from gut.bmj.com on March 24, 2014 - Published by group.bmj.com Downloaded from gut.bmj.com on March 24, 2014 - Published by group.bmj.com
191 191






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.
Downloaded from gut.bmj.com on March 24, 2014 - Published by group.bmj.com Downloaded from gut.bmj.com on March 24, 2014 - Published by group.bmj.com
192 192







Recent advances in basic science

patients with IBS was increased and correlated with mast cell
counts and the severity of the abdominal pain. It can be
hypothesised that in patients with IBS, alterations in the sensitiv-
ity or number of free fatty acid receptors involved in the
SCFA-induced release of 5-HT from mast cells, may play an
important role.
As already outlined, FFAR2 and FFAR3 sense SCFAs to affect
the release of gut peptides. Patients with IBS have increased
fasting and postprandial CCK levels.
109
Isolated duodenal I-cells
are highly enriched in FFAR2 and FFAR3 mRNA transcripts.
105

Under normal conditions these cells mainly sense circulating
SCFAs. The concept that small intestinal bacterial overgrowth is
a major pathogenic mechanism underlying IBS is still a matter
of debate.
110112
Nevertheless, in those patients with IBS with
bacterial overgrowth, luminal SCFA may also be sensed by the
I-cells and increase CCK secretion ( gure 4). Infusion of CCK
in patients with IBS can lead to excessive intestinal motor activ-
ity and reduced pain thresholds.
113 114

The postprandial PYY response is signicantly increased in
hypersensitive compared to normosensitive patients with IBS.
109

We propose that FFAR2 activation by SCFAs might be an import-
ant trigger for the release PYY/GLP-1 from colonic L-cells ( gure
4). Indeed, feeding rats a fructo-oligosaccharide-enriched diet
selectively induced the proliferation of FFAR2-positive
L-cells.
115
Additionally, colonic infusion of SCFA stimulated PPY
release in rats. Immunoneutralisation of circulating PYY abolished
the effect of SCFA on colonic motility while the effect was mim-
icked by exogenous PYY infusion.
116
In FFAR3
/
mice, charac-
terised by a decrease in PYY expression, intestinal transit rate was
increased.
117

In conclusion, the low FODMAP diet might also improve
symptoms by reducing the levels of SCFAs that act via FFAR2,
FFAR3 and transporters to affect ion secretion and GI motility
by releasing hormones and neurotransmitters. Studies are war-
ranted to investigate whether alterations in the number or sensi-
tivity of these FFARs occur in patients with IBS. If so, FFAR2
and FFAR3 antagonists, currently under development, may
therefore become promising tools for the treatment of IBS.
118

Since SCFAs induce neutrophil chemotaxis through FFAR2, a
low FODMAP diet or FFAR2 antagonists may, therefore, also
help to control inammation in subsets of patients with IBS
with low-grade inammation.
119


Altered carbohydrate sensing?
Effects observed in TAS1R3
/
and GLUT5
/
mice mimic the
effects observed in patients with IBS. Due to duodenal carbohy-
drate malabsorption, both genotypes display a distended prox-
imal colon with the development of gas pockets as a result of
subsequent fermentation by intestinal microora.
58 120

Additionally, the increased carbohydrate content in the colon of
TAS1R3
/
mice, induced robust glucose-induced GLP-1 secre-
tion from the colon which may induce alterations in transit
( gure 5). Fructose interacts with TAS1R3 and GLUT5.
58 120

We hypothesise that the fructose malabsorption in some patients
with IBS may be due to alterations in the basal expression of
TAS1R3 and GLUT5 or in their regulation in response to
dietary fructose. Therefore, future studies are warranted to
investigate the mechanisms of altered glucose sensing in patients
with IBS.

The gluten-free diet
Gluten is a well known trigger for GI symptoms in the setting
of coeliac disease. The existence of gluten intolerance was
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 K
ATP
channels and inux of Ca
2+
through voltage-gated Ca
2+
channels. The resulting increase in GLP-1 release may induce changes in transit due to the ileal brake effect. ATP, adenosine triphosphate;
Downloaded from gut.bmj.com on March 24, 2014 - Published by group.bmj.com Downloaded from gut.bmj.com on March 24, 2014 - Published by group.bmj.com
193 193






FODMAPs, fermentable oligosaccharides, disaccharides, monosaccharides and polyols; GLP-1, glucagon-like peptide 1; GLUT2, glucose transporter 2;
GLUT5, glucose transporter 5; K
ATP
, ATP-sensitive potassium channel; TAS1R2, taste receptor type 1 member 2; TAS1R3, taste receptor type 1
member 3; SGLT1, sodium-dependent glucose transporter.
Downloaded from gut.bmj.com on March 24, 2014 - Published by group.bmj.com Downloaded from gut.bmj.com on March 24, 2014 - Published by group.bmj.com
194 194







Recent advances in basic science

However, the same authors recently reported that gluten might
not be a specic trigger of functional gut symptoms once
dietary FODMAPs are reduced.
122
Another controlled trial of
gluten-free diet showed that gluten alters small bowel perme-
ability in diarrhoea-predominant patients with IBS, particularly
in HLA-DQ2/8-positive patients.
123
This may result in greater
uid ux toward the lumen and may elicit immune responses
that affect afferent nerves resulting in hypersensitivity.

Altered glutamate sensing?
Glutamic acid (Glu) and proline together account for one half
or more of the peptide-bound amino acids in gluten. Ingestion
of a protein diet rich in L-Glu does not lead to appreciable
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,
dietary Glu may stimulate Glu sensors (e.g TAS1R -TAS1R3,
CaSR and metabotropic glutamate receptors (mGluR)) in the
stomach and intestine producing local effects on gut function.
For example, in dogs, intragastric, but not intraduodenal,
administration of monosodium glutamate (MSG) stimulated
gastric emptying and induced phasic non-propagating contrac-
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

Among 20 natural amino acids investigated, only L-Glu
evoked ring of afferent bres of the vagal gastric branch in
rats. The effect is mediated via metabotropic L-Glu receptors
that induce the release of 5-HT from mucosal cells interacting
with 5-HT
3
receptors on afferent bres.
128
Additionally, intra-
gastric infusion of Glu activated brain regions that are directly
or indirectly targeted by these vagal inputs.
129
Nevertheless, the
effect of glutamate on vagal afferent ring seems not to be
straightforward, since inhibitory effects of exogenous and
endogenous glutamate have been reported on vagal afferent
mechanosensitivity involving group II (mGluR2 and 3) and
group III mGluR (mGlu R4, 6, 7, 8).
130

In view of the benecial effects that are observed with a
gluten-free diet in patients with IBS, we hypothesise that altera-
tions in luminal glutamate sensing may occur that affect vagal
nerve activity resulting in altered braingut interactions and
symptom generation.

The elimination diet for food chemicals
The elimination diet involves restriction of common food aller-
gens, specic chemical substances in foods or medications that
contain these chemicals.
Coffee is commonly reported as a trigger for symptoms in
patients with IBS.
131
It is not clear whether salicylates or caf-
feine in coffee are the triggers but both components taste bitter
and could alter gut function via TAS2Rs.

Functional dyspepsia
According to the Rome III classication, functional dyspepsia
can be divided into two categories: postprandial distress syn-
drome and epigastric pain syndrome, suggesting that at least in
some patients the disorder is related to food ingestion. While
most of the patients report that their symptoms are triggered
within 30 min after meal ingestion, few studies have been per-
formed to evaluate the role of specic foods.
132
Particularly,
meals containing fat seem to induce or exacerbate symptoms.
133

Pilichiewicz et al
134
reported that ingestion of a high-fat meal,
but not of a high-carbohydrate meal, was associated with a sub-
stantially greater increase in nausea and pain immediately after
Downloaded from gut.bmj.com on March 24, 2014 - Published by group.bmj.com Downloaded from gut.bmj.com on March 24, 2014 - Published by group.bmj.com
195 195






completion of ingestion. Furthermore the scores for nausea and
pain were related directly to the increased plasma CCK concen-
trations in patients with functional dyspepsia. It is known that
exogenous CCK administration can mimic dyspeptic symp-
toms.
135
It is therefore conceivable that similar to patients with
IBS, who frequently overlap with functional dyspepsia patients,
alterations in the expression or sensitivity of free fatty acid
receptors on endocrine cells may render them more sensitive to
dietary fat and exacerbate symptoms.
136
Diet-intervention
studies, similar to what has been performed in patients with
IBS, are warranted to investigate the role of nutrients and,
hence, of taste receptors in symptom generation in patients with
functional dyspepsia.


CONCLUSIONS AND PERSPECTIVES
The observation, and now compelling evidence, for the pres-
ence and function of taste receptors in extraoral tissues offers
exciting new possibilities for targeted therapeutics in the battle
against diseases of and involving the GI tract. Several taste
receptor families involved in nutrient sensing in the gut offer
potential advantages as drug targets.


Sweet taste receptors
This long history of sweetener chemistry suggests that new com-
pounds of exceptional specicity and high efcacy could be
designed. The benecial effects of articial sweeteners in the
control of body weight and glucose homeostasis are still a
matter of debate, but advances in our understanding of the
sweet taste receptor biology may therefore help to design new
articial sweeteners with favourable effects.
137
Indeed, in con-
trast with in vitro studies, in vivo studies either in rodents or in
humans failed to show an effect of articial sweeteners on the
release of satiation hormones.
56 58 8183
Advances in our under-
standing of the sweet taste receptor biology may therefore help
to design new articial sweeteners with favourable effects.
However, their effectiveness may be complicated by the
complex interaction between sweet taste receptors and glucose
transporters that cross-regulate each others expression.
Additionally, the evidence provided for altered sweet taste recep-
tor control in type 2 diabetes may open new opportunities for
drugs that interfere with sweet taste receptor regulation.
In gastric bypass patients, additional studies should help to
provide insight to what the effect is of shunting nutrients to
more distal regions of the gut in the control of taste receptor
expression/glucose transporter regulation, and the release of
hormones involved in the regulation of energy and glucose
homeostasis. It is tempting to speculate that targeting these
receptors/transporters may mimic the effects of gastric bypass
surgery in a non-surgical manner. In fact, ongoing trials with
SGLT1 inhibitors already show promising results.
88



Bitter taste receptors
Evidence suggests that bitter agonists could be considered as
good targets to reduce hunger and motility.
18 22 32
However,
the design of new bitter drugs may be complicated by the fact
that in humans, 25 receptor subtypes exist, each with different
ligand selectivities and a different distribution pattern.
Nevertheless, there are also some opportunities. Thousands of
plant-derived bitter tastants and metabolites are available that
could be tested in appropriate physiological models to select
those with favourable therapeutic proles.
Downloaded from gut.bmj.com on March 24, 2014 - Published by group.bmj.com Downloaded from gut.bmj.com on March 24, 2014 - Published by group.bmj.com
196 196






1

2
as
2013;24:719.
et al. taste
33 et al. a in the of
of the to feelings.
2012;24:31.

3
2001;106:38190.
et al. taste Nature
34 of the in
1981;95:100315.

2002;416:199202.
35 C, et al. and of
4 et al. and
taste. 2003;115:25566.

and in and
2001;903:12840.
5 taste as of
2012;26:482731.
36 of on two of
1990;31:191201.
6 et al. as taste
37 et al. inhibits

7
2000;100:70311.
C, C, et al. of human

in neurons.
e111.

taste 2010;35:15770.
38 et al. in the of the
8 taste and taste perception.
2006;63:15019.

and in
1999;846:111.
9 the multiple
39 Q, et al. peptide

2009;90:738S42S.

of
10 et al. of the
in taste

and
2004;53:2492500.

11
2010;123:97282.
in the gut: to therapeutics?
40 et al. of peripheral
of and on intake

12
2013;24:92100.
C, et al.

are of the
2005;1044:12731.

and 2010;30:837682.
41 the
13 et al. and
taste. 2006;444:28894.

of on and in
1984;322:31621.
14 of taste
42 et al. cholecystokinin

15
2013;24:2329.
perceptual


2008;377:5237.

of taste buds.
43 et al.

16
2013;33:755964.
et al. as a of taste

the
2012;302:C2109.

in 2000;97:110449.
44 in and
17 et al. the of
in the of

of Diabetes
2008;57:22807.

2011;33:88094.
45 et al.
18 et al. taste agonist
test

of cholecystokinin.
2011;140:90312.

in 1):S-549.
46 and
19 et al. and of the
in the

in a
of in of 1994;43:7358.

A378.
47 et al. of the
20 et al. a growth-hormone-releasing

taste

1999;402:65660.

2013;304:G27182.


Recent advances in basic science

Amino acid and free fatty acid receptors
Recent evidence suggests that food choice (low FODMAPs,
gluten-free diet) is a key management strategy for the treatment
of symptoms in patients with IBS. These studies underscore
claims made for many years by alternative practitioners. The
mechanisms involved are not completely understood, but indir-
ect evidence suggests that altered carbohydrate sensing, exacer-
bated sensing of SCFAs by FFARs or of Glu by amino acid
receptors may be involved. Determining alterations in basal
expression levels of these receptors or in their regulation in
response to a diet may help to provide a rationale for the use of
FFAR antagonists or Glu receptor antagonists in these patients
and may help to predict whether a patient will be responsive to
the diet or not. Similar strategies may be useful in patients with
functional dyspepsia.
It is clear that the existence and functional role of taste recep-
tors in the gut is a new and fascinating eld of research that
may lead to the development of new therapeutic drugs that may
benet from the progress made in our understanding of taste
receptor function in the tongue.

Acknowledgements The author wishes to thank S. Munger for providing helpful
discussion.
Competing interests None.
Provenance and peer review Commissioned; externally peer reviewed.


REFERENCES
21 Foster-Schubert KE, Overduin J, Prudom CE, et al. Acyl and total ghrelin are
suppressed strongly by ingested proteins, weakly by lipids, and biphasically by
carbohydrates. J Clin Endocrinol Metab 2008;93:19719.
22 Janssen S, Laermans J, Verhulst PJ, et al. Bitter taste receptors and
alpha-gustducin regulate the secretion of ghrelin with functional effects on food
intake and gastric emptying. Proc Natl Acad Sci U S A 2011;108:20949.
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.
24 Hass N, Schwarzenbacher K, Breer H. T1R3 is expressed in brush cells and
ghrelin-producing cells of murine stomach. Cell Tissue Res 2010;339:493504.
25 Saffouri B, DuVal JW, Makhlouf GM. Stimulation of gastrin secretion in vitro by
intraluminal chemicals: regulation by intramural cholinergic and noncholinergic
neurons. Gastroenterology 1984;87:55761.
26 Haid D, Widmayer P, Voigt A, et al. Gustatory sensory cells express a receptor
responsive to protein breakdown products (GPR92). Histochem Cell Biol
2013;140:13745.
27 Haid DC, Jordan-Biegger C, Widmayer P, et al. Receptors responsive to protein
breakdown products in g-cells and d-cells of mouse, swine and human. Front
Physiol 2012;3:65.
28 Haid D, Widmayer P, Breer H. Nutrient sensing receptors in gastric endocrine cells.
J Mol Histol 2011;42:35564.
29 Feng J, Petersen CD, Coy DH, et al. Calcium-sensing receptor is a physiologic
multimodal chemosensor regulating gastric G-cell growth and gastrin secretion.
Proc Natl Acad Sci U S A 2010;107:177916.
30 Busque SM, Kerstetter JE, Geibel JP, et al. L-type amino acids stimulate gastric
acid secretion by activation of the calcium-sensing receptor in parietal cells. Am J
Physiol Gastrointest Liver Physiol 2005;289:G6649.
31 Gielkens HA, van den Biggelaar A, Vecht J, et al. Effect of intravenous amino
acids on interdigestive antroduodenal motility and small bowel transit time. Gut
1999;44:2405.
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
of gastric phase III in the fasting state. Gastroenterology 2013;144(suppl 1):S-548.
Downloaded from gut.bmj.com on March 24, 2014 - Published by group.bmj.com Downloaded from gut.bmj.com on March 24, 2014 - Published by group.bmj.com
197 197







Recent advances in basic science

48

Hira T, Nakajima S, Eto Y, et al. Calcium-sensing receptor mediates
phenylalanine-induced cholecystokinin secretion in enteroendocrine STC-1 cells.
Febs J 2008;275:46206.

76

77

Tazoe H, Otomo Y, Karaki S, et al. Expression of short-chain fatty acid receptor
GPR41 in the human colon. Biomed Res 2009;30:14956.
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:E65160.

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:G53846.

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;61:36471.

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:G52837.

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;62:353241.

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:G136675.

gastrointestinal tract in humans with and without type 2 diabetes. Gut 2009;58:33746.
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;118:3693700.

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
CCK release in enteroendocrine STC-1 cells: role of L-type voltage-sensitive Ca2+

82
2011;65:50813.
Steinert RE, Frey F, Topfer A, et al. Effects of carbohydrate sugars and articial

channels. Am J Physiol Cell Physiol 2006;291:C72639.

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
receptor in regulating GLP-1, PYY, and CCK release in humans. Am J Physiol

83
Nutr 2011;105:13208.
Ma J, Bellon M, Wishart JM, et al. Effect of the articial sweetener, sucralose, on

Endocrinol Metab 2011;301:E31725.

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:G7359.

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;61:393411.

2011;30:52432. 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;258:8997.

2007;104:1506974. 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 model. Obes Surg 2013;23:13619.

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:G792802.

Roux-en-Y gastric bypass. Physiol Behav 2011;104:70921.
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:E46474.

2 diabetes. Clin Ther 2013;35:27385, 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;104:1507580.

(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;345:2509.

Diabetes 2002;51:275763. 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;26:1405.

study. Cell Metab 2008;8:5329. 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;450:1069.

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;156:66371.

signaling in taste buds. Ann N Y Acad Sci 2009;1170:98101.
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;61:18796.

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;52:114754.

Nutr 2006;60:66772.
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

66
2012;55:244555.
Mace OJ, Afeck J, Patel N, et al. Sweet taste receptors in rat small intestine

96
2006;106:16319.
Shepherd SJ, Parker FC, Muir JG, et al. Dietary triggers of abdominal symptoms in

stimulate glucose absorption through apical GLUT2. J Physiol 2007;582:37992.

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;6:76571.

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:R199208.

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;31:87482.

cats indifference toward sugar. PLoS Genet 2005;1:2735. 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;25:136673.

1991;261:R793801. 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;403:55975.

71
glucagon-like peptide-1 secretion through GPR120. Nat Med 2005;11:904.
Dotson CD, Zhang L, Xu H, et al. Bitter taste receptors inuence glucose
100 Ruppin H, Bar-Meir S, Soergel KH, et al. Absorption of short-chain fatty acids by
the colon. Gastroenterology 1980;78:15007.

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;96:98996.

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;288:451321.

and GPR43 on colonic functions. J Physiol Pharmacol 2008;59(Suppl 2):25162.
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;222:5861.

carboxylic acids. J Biol Chem 2003;278:1131219. 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;39:13542.

Physiol 2003;284:R126976.
Downloaded from gut.bmj.com on March 24, 2014 - Published by group.bmj.com Downloaded from gut.bmj.com on March 24, 2014 - Published by group.bmj.com
198 198






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;324:35360.

One 2012;7:e42373.
Downloaded from gut.bmj.com on March 24, 2014 - Published by group.bmj.com Downloaded from gut.bmj.com on March 24, 2014 - Published by group.bmj.com
199 199







Recent advances in basic science

106 Mitsui R, Ono S, Karaki S, et al. Neural and non-neural mediation of
propionate-induced contractile responses in the rat distal colon. Neurogastroenterol
Motil 2005;17:58594.
107 Ford AC, Brandt LJ, Young C, et al. Efcacy of 5-HT3 antagonists and 5-HT4
agonists in irritable bowel syndrome: systematic review and meta-analysis. Am J
Gastroenterol 2009;104:183143; quiz 44.
108 Cremon C, Carini G, Wang B, et al. Intestinal serotonin release, sensory neuron
activation, and abdominal pain in irritable bowel syndrome. Am J Gastroenterol
2011;106:12908.
109 Van Der Veek PP, Biemond I, Masclee AA. Proximal and distal gut hormone
secretion in irritable bowel syndrome. Scand J Gastroenterol 2006;41:1707.
110 Pimentel M, Chow EJ, Lin HC. Eradication of small intestinal bacterial overgrowth
reduces symptoms of irritable bowel syndrome. Am J Gastroenterol 2000;95:35036.
111 Vanner S. The small intestinal bacterial overgrowth. Irritable bowel syndrome
hypothesis: implications for treatment. Gut 2008;57:131521.
112 Posserud I, Stotzer PO, Bjornsson ES, et al. Small intestinal bacterial overgrowth in
patients with irritable bowel syndrome. Gut 2007;56:8028.
113 van der Schaar PJ, van Hoboken E, Ludidi S, et al. Effect of cholecystokinin on
rectal motor and sensory function in patients with irritable bowel syndrome and
healthy controls. Colorectal Dis 2013;15:e2934.
114 Kellow JE, Phillips SF, Miller LJ, et al. Dysmotility of the small intestine in irritable
bowel syndrome. Gut 1988;29:123643.
115 Kaji I, Karaki S, Tanaka R, et al. Density distribution of free fatty acid receptor 2
(FFA2)-expressing and GLP-1-producing enteroendocrine L cells in human and rat
lower intestine, and increased cell numbers after ingestion of
fructo-oligosaccharide. J Mol Histol 2011;42:2738.
116 Cherbut C, Ferrier L, Roze C, et al. Short-chain fatty acids modify colonic motility
through nerves and polypeptide YY release in the rat. Am J Physiol 1998;275:
G141522.
117 Samuel BS, Shaito A, Motoike T, et al. Effects of the gut microbiota on host
adiposity are modulated by the short-chain fatty-acid binding G protein-coupled
receptor, Gpr41. Proc Natl Acad Sci USA 2008;105:1676772.
118 Bindels LB, Dewulf EM, Delzenne NM. GPR43/FFA2: physiopathological relevance
and therapeutic prospects. Trends Pharmacol Sci 2013;34:22632.
119 Sina C, Gavrilova O, Forster M, et al. G protein-coupled receptor 43 is essential for
neutrophil recruitment during intestinal inammation. J Immunol 2009;183:751422.
120 Barone S, Fussell SL, Singh AK, et al. Slc2a5 (Glut5) is essential for the absorption
of fructose in the intestine and generation of fructose-induced hypertension. J Biol
Chem 2009;284:505666.
121 Biesiekierski JR, Newnham ED, Irving PM, et al. Gluten causes gastrointestinal
symptoms in subjects without celiac disease: a double-blind randomized
placebo-controlled trial. Am J Gastroenterol 2011;106:50814; quiz 15.
122 Biesiekierski JR, Peters SL, Newnham ED, et al. No effects of gluten in patients
with self-reported non-celiac gluten sensitivity after dietary reduction of
fermentable, poorly absorbed, short-chain carbohydrates. Gastroenterology
2013;145:3208, e3.
123 Vazquez-Roque MI, Camilleri M, Smyrk T, et al. A controlled trial of gluten-free
diet in patients with irritable bowel syndrome-diarrhea: effects on bowel frequency
and intestinal function. Gastroenterology 2013;144:90311, e3.
124 Reeds PJ, Burrin DG, Stoll B, et al. Intestinal glutamate metabolism. J Nutr
2000;130:978S82S.
125 Toyomasu Y, Mochiki E, Yanai M, et al. Intragastric monosodium L-glutamate
stimulates motility of upper gut via vagus nerve in conscious dogs. Am J Physiol
Regul Integr Comp Physiol 2010;298:R112535.
126 Zai H, Kusano M, Hosaka H, et al. Monosodium L-glutamate added to a
high-energy, high-protein liquid diet promotes gastric emptying. Am J Clin Nutr
2009;89:4315.
127 Boutry C, Matsumoto H, Airinei G, et al. Monosodium glutamate raises antral
distension and plasma amino acid after a standard meal in humans. Am J Physiol
Gastrointest Liver Physiol 2011;300:G13745.
128 Uneyama H, Niijima A, San Gabriel A, et al. Luminal amino acid sensing in the rat
gastric mucosa. Am J Physiol Gastrointest Liver Physiol 2006;291:G116370.
129 Kondoh T, Torii K. Brain activation by umami substances via gustatory and visceral
signaling pathways, and physiological signicance. Biol Pharm Bull
2008;31:182732.
130 Page AJ, Young RL, Martin CM, et al. Metabotropic glutamate receptors inhibit
mechanosensitivity in vagal sensory neurons. Gastroenterology 2005;128:40210.
131 Simren M, Mansson A, Langkilde AM, et al. Food-related gastrointestinal
symptoms in the irritable bowel syndrome. Digestion 2001;63:10815.
132 Bisschops R, Karamanolis G, Arts J, et al. Relationship between symptoms and
ingestion of a meal in functional dyspepsia. Gut 2008;57:1495503.
133 Mullan A, Kavanagh P, OMahony P, et al. Food and nutrient intakes and eating
patterns in functional and organic dyspepsia. Eur J Clin Nutr 1994;48:97105.
134 Pilichiewicz AN, Feltrin KL, Horowitz M, et al. Functional dyspepsia is associated
with a greater symptomatic response to fat but not carbohydrate, increased fasting
and postprandial CCK, and diminished PYY. Am J Gastroenterol
2008;103:261323.
135 Chua AS, Keeling PW. Cholecystokinin hyperresponsiveness in functional
dyspepsia. World J Gastroenterol 2006;12:268893.
136 Barbera R, Feinle C, Read NW. Abnormal sensitivity to duodenal lipid infusion in
patients with functional dyspepsia. Eur J Gastroenterol Hepatol 1995;7:10517.
137 Raben A, Richelsen B. Articial sweeteners: a place in the eld of functional
foods? Focus on obesity and related metabolic disorders. Curr Opin Clin Nutr
Metab Care 2012;15:597604.
Downloaded from gut.bmj.com on March 24, 2014 - Published by group.bmj.com







Taste receptors of the gut: emerging roles in
health and disease
Inge Depoortere

Gut 2014 63: 179-190 originally published online October 16, 2013
doi: 10.1136/gutjnl-2013-305112



Updated information and services can be found at:
http://gut.bmj.com/content/63/1/179.full.html




References


Email alerting
service
These include:

This article cites 136 articles, 52 of which can be accessed free at:
http://gut.bmj.com/content/63/1/179.full.html#ref-list-1

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


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

GUT Recent advances in basic science (65 articles)
Irritable bowel syndrome (296 articles)



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/

Potrebbero piacerti anche