Sie sind auf Seite 1von 9

Physiology & Behavior 105 (2011) 6270

Contents lists available at ScienceDirect

Physiology & Behavior


j o u r n a l h o m e p a g e : w w w. e l s ev i e r. c o m / l o c a t e / p h b

Nutrient sensing in the gut: interactions between chemosensory cells, visceral


afferents and the secretion of satiation peptides
Robert E. Steinert, Christoph Beglinger
Clinical Research Center, Department of Biomedicine and Division of Gastroenterology, University Hospital Basel, Basel, Switzerland

a r t i c l e i n f o a b s t r a c t

Article history: The chemical environment of the intestinal lumen and the presence of nutrients in the gut result in the
Received 20 December 2010 secretion of regulatory peptides and in the discharge of intestinal sensory afferent bers. Since afferent nerve
Received in revised form 24 February 2011 terminals do not directly extend into the intestinal lumen, their activation presumably depends on
Accepted 25 February 2011
intermediary steps post-absorptive mechanisms, i.e. direct neuronal activation by absorbed substances and
pre-absorptive mechanisms, i.e. neuronal activation by a secondary substance released from the mucosal
Keywords:
Gut
epithelium.
Gastrointestinal satiation peptides Entero-endocrine cells (EEC) are able to taste the luminal content and to function as chemosensory
Taste receptor transducers to provide the interface between the intestinal lumen and the afferent nerve terminals. Their
Nutrient sensing secretory products mainly peptide hormones are released upon stimulation by nutrients into the
extracellular space of the lamina propria to either act 1) locally in a paracrine fashion to activate afferent
terminals or other cells or 2) in an endocrine fashion via intestinal capillaries and the lymphatic system to
bind to specic receptors at more distant targets.
The chemosensory mechanisms by which EEC sense the intestinal lumen remain, however, poorly
understood. Recent information suggest that taste signaling mechanisms known from the oral epithelium
also operate in the mucosal epithelium. Several nutrient-responsive G-protein coupled receptors (GPCRs)
have been identied in EEC including the sweet-taste responsive T1R2/T1R3 heterodimer or GPR120,
responsive to free fatty acids (FFAs).
This review provides a brief overview on gastrointestinal chemosensory mechanisms and their functional
involvement in the secretion of satiation peptides with a focus on human studies albeit most evidence at
present comes from in vitro or animal studies.
2011 Elsevier Inc. All rights reserved.

1. Introduction The rst evidence that nervism was not the only mechanism
controlling gut functions was provided by Bayliss and Starling in 1902
Intestinal chemosensitivity is a key element in digestive processes [4]. They observed that the presence of protons in the proximal small
and has been observed in mechanisms such as the dependence of intestine elicited a strong stimulation of pancreatic-uid secretion. By
gastric emptying on the chemical nature of nutrients or the fact that cutting all the nerves to the pancreas in their experimental animals,
oral ingestion of glucose triggers more insulin release than a similar they found that this process was not governed by the nervous system
plasma glucose prole delivered intravenously [1,2]. and showed that a substance secreted by the intestinal lining stimulates
One of the rst physiologists who proposed a chemosensitivity the pancreas after being transported via the bloodstream. They named
hypothesis was Pavlov in the 19th century. Based on his nervism, i.e. the secreted compound 'secretin' and used Hardy's term hormone as a
the theory that the nervous system controls the greatest possible general designation for blood-borne chemical messengers.
number of bodily activities, he suggested that sensory nerves In the 1970s Fujita and Kobayashi [5] proposed the intestinal sensor
are exposed to the intestinal lumen and directly catch chemical cell theory, which was further expanded by Grundy [6]. They described
messages of the luminal content at their nerve endings (nerve nutrient-sensing bipolar cells, which contain secretory granules in the
antenna theory) [3]. basolateral portion and cytoplasmatic projections to the intestinal
lumen. These cells were thought to interact with the intestinal lumen
and to release peptide hormones and bioactive amines to transfer
information to other organs via endocrine or vagal pathways.
Corresponding author at: Clinical Research Center, Department of Biomedicine,
Division of Gastroenterology, University Hospital Basel, CH-4031 Basel, Switzerland.
In 1985, N. Mei reviewed the subject of intestinal chemosensitivity
Tel.: +41 612653846; fax: +41 612653847. [7]. Available data demonstrated that (i) changes in the chemical
E-mail address: beglinger@tmr.ch (C. Beglinger). environment of the intestinal lumen (pH, osmolality) and the

0031-9384/$ see front matter 2011 Elsevier Inc. All rights reserved.
doi:10.1016/j.physbeh.2011.02.039
R.E. Steinert, C. Beglinger / Physiology & Behavior 105 (2011) 6270 63

presence of different nutrients (carbohydrates, protein, lipids) lead to function as the transducer to provide the interface between the GI
a discharge of sensory afferent bers; however (ii) vagal ring is not lumen and nerve terminals [15].
due to a direct intraluminal stimulation of nerve terminals. He
suggested that activation of chemosensory nerve terminals may 3. Chemosensory cells
depend on intermediary steps. To date, intestinal chemosensitivity is
thought to involve a highly specialized and complex system of A major morphological characteristic of chemosensory cells is that
primary afferent neurons, intestinal chemosensory cells and the gut they are scattered along the epithelial lining and that they have direct
immune system. access to the luminal content. Cell types that fulll this requirement
This review summarizes important current data on gastrointesti- include (i) enterocytes, (ii) brush cells and (iii) entero-endocrine cells
nal (GI) chemosensory mechanisms and their functional involvement (EEC). All of them have repeatedly been implicated in intestinal
in the secretion of satiation peptides with a focus on human studies chemosensitivity, and afferent nerve terminals have been visualized
albeit most evidence at present comes from in vitro or animal studies. close to these cells [12,23,24]. Brush cells are characterized by a brush
of rigid apical microvilli and, although the function of these cells has
2. Primary afferent neurons long been unknown, recent data suggest that they comprise a diffuse
chemosensory system that covers large areas of the digestive and
Two classes of primary afferent neurons innervate the GI tract: respiratory tract (for review see ([25])).
intrinsic primary afferent neurons (IPANs) and extrinsic (vagal and Most data, however, imply that EEC are the primary chemosensory
spinal) primary afferent neurons. In addition intestinofugal neurons cells. Although they represent less than 1% of the entire epithelial
are part of the afferent limb [8]. population; in total, they constitute the largest endocrine organ
IPANs innervate both, mucosal and muscular layers of the gut wall secreting multiple peptide hormones and bioactive amines. Results
and originate either in the myenteric (Auerbach) or submucosal obtained from experiments in cultured entero-endocrine GLUTag and
(Meissner) plexus. They are part of the enteric nervous system which NCI-H716 cells indicate that EEC are themselves nutrient-sensitive,
generates reexes affecting GI motility, blood ow and water/ albeit indirect sensing pathways (via brush cells or enterocytes) might
electrolyte secretion. Myenteric IPAN's respond to distortion in also be involved [26,27]. Two groups of EEC can be distinguished
external muscles layers whereas Meissner IPANs detect distortion of according to their epithelial localization: open cells with microvilli
the mucosa. Both also respond to changes in luminal chemistry; they extending into the intestinal lumen, and closed cells that do not reach
synapse with each other and form a dense network that commu- the epithelial surface. Open cells directly sense luminal factors whereas
nicates to interneurons to provide the enteric nervous system the closed cells (e.g. enterochromafn-like cells) are regulated by the
information required for the autonomic control of digestion [9,10]. luminal content indirectly through neural and humoral mechanisms.
Extrinsic primary afferent neurons have their cell bodies lying in Over 20 different EEC types have been classied according to their
dorsal root ganglia (spinal primary afferent neurons) and in jugular content of the secretory granules. The different cell types are variously
and nodose ganglia (vagal primary afferent neurons) [10]. They both distributed along the intestinal tract. For example, I-cells are primarily
innervate the entire length of the GI tract; vagal afferents are more present in the duodenal and jejunal mucosa, whereas the majority of
prevalent in the proximal gut and spinal afferents (particularly pelvic) L-cells are located in the distal ileum and colon (Table 1).
predominate the distal gut. Their nerve terminals are distributed
within the gut wall including mucosa and submucosa, muscle and 4. Nutrient sensing and chemosensory mechanisms
enteric ganglia but also in serosa and mesenteric attachments.
Importantly, 80 to 90% of axons in the vagus nerve are afferent The ndings that different macronutrients have different poten-
nerve bers; therefore, the vagus nerve is the major component of cies to modulate the secretion of gut peptides (e.g. CCK, GLP-1 or PYY)
extrinsic afferent pathways [11]. Vagal afferents have been demon- independently of energy densities suggests that regulatory mecha-
strated to be both mechano- and chemo-sensitive. For mechanosen- nisms are of complex design to generate the appropriate secretory
sitivity, sensory nerve bers may function as contact sensors that responses [2830].
detect the movement of luminal content across the mucosal surface
(e.g. mucosal deformation or villi distension) by stretch-activated ion 4.1. Carbohydrates
channels [12]. Chemosensitivity was rst demonstrated more than
50 years ago by Iggo. He measured the discharge of vagal afferent Carbohydrates in nature include the complex plant products starch
bres with electrophysiological recordings and found a variety of and cellulose, the animal starch (glycogen), and various disaccharide
nutrients to activate the vagus from the gut [13]. and monosaccharide sugars. The complex carbohydrates are further
Although the primary afferent nerve terminals in the mucosal
epithelium are closely associated with the lamina propria, they do,
Table 1
however, not project directly into the intestinal lumen. Tracing
Selected entero-endocrine cells: location and secretory products.
studies demonstrate that they never actually enter the epithelial layer
[14,15]. Thus their activation is thought to depend on postabsorptive Cell type Highest density Peptide released
or preabsorptive mechanisms, respectively. Several vagal chemor- G-cells Stomach Gastrin
eceptors have been described including gluco-, amino acid-, lipid-, X-cells Stomach Ghrelin
pH-, and osmo-receptors as well as receptors sensitive to specic ECL-cells Stomach Histamin
Unamed cells Stomach and duodenum Gastrin Releasing Peptide (GRP)
dietary compounds, suggesting a direct post-absorptive activation of
S-cells Duodenum and jejunum Secretin
the vagus by absorbed substances from the lumen [12,15]. In contrast, I-cells Duodenum and jejunum Cholecystokinin (CCK)
in the 1980s Gibbs and Smith discovered that gut hormones particular K-cells Duodenum and jejunum Glucose-dependent insulinotropic
those, like CCK, that act to control food intake and gastric emptying polypeptide (GIP)
N-cells Ileum Neurotensin
also require the vagus in mediating their action suggesting a neuronal
L-cells Ileum and colon Glucagon-like peptides (GLP's),
vagal activation by secondary substances (e.g. gut peptides) released Peptide YY (PYY)
from the mucosal epithelium [16]. To date, numerous receptors for D-cells Entire GI tract Somatostatin
gut peptides and other bioactive molecules have been identied on EC-cells Entire GI tract 5-hydroxytryptamin (5-HT, Serotonin)
afferent bers, including CCK-1 and -2, GLP-1, Y2, GHS1, CB1, GABAB EC-cells, enterochromafn cells; ECL-cells, enterochromafn-like-cells; glucagon-like
receptors [1722] and specialized chemosensory cells are proposed to peptides (GLP's) include GLP-1, GLP-2, oxyntomodulin, glicentin.
64 R.E. Steinert, C. Beglinger / Physiology & Behavior 105 (2011) 6270

broken down by intestinal enzymes to liberate the simple sugars. multiple members of the T2R family in mouse gastric and duodenal
Most studies have focused on glucose as the most important mucosa. The proteins were cloned and sequenced, conrming that
carbohydrate for human metabolism. Different mechanisms are they are identical to known taste receptor sequences in lingual
thought to be involved in glucose sensing by gut EEC. epithelium [45]. In 2005, Dyer et al. [46] reported the expression of
T1R sweet-taste receptors at mRNA and protein levels in mouse small
4.1.1. Glucose transporter activity intestine. In addition, several other taste signalling proteins including
Several studies have documented that the secretion of peptide transducin or TRPM5 were detected in gastric or intestinal mucosa of
hormones may depend on actively absorbed glucose via the sodium- humans and rodents as well as in EEC in culture [4750]. In human
dependent glucose co-transporter (SGLT-1). Gribble et al. [31] duodenal biopsy sections, -gustducin, T1R2 and T1R3 were
identied a glucose sensing-pathway in GLUTag cells, whereby sugars identied by immunohistochemistry at apical projections of cells
trigger membrane depolarization and GLP-1 secretion via electrogenic with shapes characteristic of EEC. -gustducin was shown to be
actions of SGLT-1. In the same experiments, GLP-1 release was colocalized with: (i) GLP-1-expressing entero-endocrine L-cells,
impaired when glucose absorption was blocked with phlorizin, a (ii) GIP-expressing entero-endocrine K-cells, and (iii) GIP and GLP-1
specic SGLT-1 inhibitor. In rats, glucose-triggered GIP secretion was coexpressing entero-endocrine K/L-cells [50]. Independent conrma-
markedly reduced by phlorizin [32]. In humans, slowly and rapidly tion of the expression of -gustducin in K and L-cells came from
digestible carbohydrates differed in their ability to stimulate the experiments using laser capture microdissection followed by RT-PCR
release of GLP-1 and GIP; moreover, the rate of intestinal glucose and the same technique was used to show that -gustducin was not
absorption strongly correlated with GIP plasma concentrations [33]. present in duodenal enterocytes [50].
In addition to SGLT-1, there is evidence that GLUT2, which facilitates In human colonic mucosa, Rozengurt et al. [48] found -gustducin
the glucose transport across the basolateral membrane into the in entero-endocrine L-cells that express PYY and GLP-1. Recent
bloodstream, may be involved in glucose sensing. Although GLP-1 studies also demonstrate coexpression of -gustducin with CCK in
secretion was generally thought to be insensitive to plasma glucose entero-endocrine I-cells [45]. Furthermore, most -gustducin positive
levels, knockout mice models for GLUT2 (GLUT2/) showed cells coexpress chromogranin A, a marker of EEC, but not villin, which
impaired GLP-1 responses to oral glucose [34], suggesting a potential indicates that in humans, -gustducin is present predominantly in
metabolism-dependent pathway to adjust GLP-1 secretion to plasma EEC [48]. In contrast, in rodents, -gustducin immunoreactivity has
glucose concentration. also been documented in intestinal brush cells (which are character-
ized by villin expression) thus, suggesting a role of these cells in
4.1.2. KATP channel dependent glucose sensing chemosensory functions in rodents [5153].
KATP channel dependent glucose sensing is well characterized as
glucose sensors in pancreatic -cells but also in cardiac muscle and in 4.1.3.2. Functional evidence for the involvement of taste receptors in
subsets of neurons in the brain [35,36]. In -cells, the metabolism of glucose sensing. Functional evidence that taste receptor pathways are
glucose leads to the generation of ATP; this triggers the closure of KATP involved in gut peptide secretion stems primarily from results
channels, which leads to membrane depolarization, Ca2+ inux and obtained in intestinal cell lines which have been used extensively to
nally to the release of insulin [37]. It has been suggested that glucose elucidate secretory responses in EEC. EEC models include (i) the
may trigger the release of gut peptides from EEC via similar murine STC-1 and GLUTag cell line, (ii) the rat pancreatic acinar tumor
mechanisms. Glucose-stimulated secretion of GLP-1 from entero- cell line AR42J with neuroendocrine properties and (iii) the human
endocrine GLUTag or NCIH716 cells involves the closure of KATP endocrine intestinal cell lines HuTu-80 and NCI-H716. All of them
channels [31,38,39]; in addition in primary cultures of mouse small express multiple components of taste signaling proteins and secrete
intestine glucose-triggered GIP secretion was dependent on KATP several GI peptides [45,47]. Stimulation of NCI-H716 or GLUTag cells
channel activity [40]. However, in humans, the involvement of KATP with sweet receptor agonists, including glucose, sucrose or sucralose,
channels has been questioned because sulphonylureas, which an articial sweetener, promoted the release of GLP-1 and GIP. More
stimulate insulin release in diabetics by their inhibitory action on importantly, the sweet-taste receptor antagonists lactisole and
KATP channels, do not trigger the release of GLP-1 [38]. Moreover, gurmarin, both blocked the secretion of GLP-1 and GIP from these
Ritzel et al. [41] report that GLP-1 is also secreted in response to non- cells [50,54]. In addition, bitter ligands were shown to be potent
metabolizable sugar analogues such as 3-O-methylglucose suggesting stimulants of CCK release from STC-1 cells [55]. Major support for a
additional mechanisms of glucose sensing. functional role of taste-signaling pathways in the gut is provided by
Jang et al. [50] Knockout mice lacking -gustducin (-gust/) or
4.1.3. Taste receptors T1R3 (T1R3/) were examined for their responses to carbohydrate
Taste receptors were originally described in taste cells of lingual gavage. In contrast to the wild type littermates, the knockout models
epithelium and have been identied as ion channels (salty, sour) and showed no increase in glucose-stimulated secretion of GLP-1 and GIP.
G-protein coupled receptors (GPCRs) responsive to bitter, sweet and In addition, they revealed an impaired release of insulin and elevated
umami [42]. The GPCR type 1 taste receptor (T1R) family includes blood glucose level. Young et al. [56] quantied transcript level of
three distinct subunits (T1R1, T1R2 and T1R3) that form heterodimers T1R2, T1R3, -gustducin and TRPM5 in the upper GI tract of subjects
to mediate either: (i) sweet-taste such as carbohydrate sugars and with and without type II diabetes mellitus. He found that higher
articial sweeteners (T1R2 + T1R3) or, (ii) umami and other savory serum blood glucose concentrations were associated with lower
amino acids (T1R1 + T1R3). The type 2 receptor (T2R) family contains levels of sweet-taste signaling molecule transcripts. He also demon-
over 30 genes encoding different receptor transcripts that mediate strated in mice that jejunal infusions of glucose signicantly
bitter taste, such as plant alkaloids. Recent ndings suggest that the decreased transcript level of T1R2 compared to control infusions
molecular pathways that mediate lingual taste perception also with 2-deoxy-D-glucose or saline and therefore concluded that taste
operate in the gut. transduction in the gut might be under dynamic metabolic and
luminal control [56].
4.1.3.1. Histological evidence for the involvement of taste receptors in We also performed experiments to determine the functional
glucose sensing. In 1996, Hfer et al. [43] rst identied taste receptor- involvement of sweet-taste receptors in glucose-stimulated secre-
like cells in the rat gut by showing expression of -gustducin, a taste tions of GLP-1 and PYY in humans. We used lactisole, a competitive
specic G-protein subunit and marker for chemosensory cells. Wu et inhibitor of the sweet-taste receptor subunit T1R3, to block sweet-
al. [44] demonstrated the presence of transcripts corresponding to taste receptor function in the gut. In parallel to the in vitro data by Jang
R.E. Steinert, C. Beglinger / Physiology & Behavior 105 (2011) 6270 65

et al. [50], we could demonstrate that lactisole attenuated the regular the molecular structure of carbohydrate sugars and their ability to
glucose-stimulated secretion of GLP-1 and PYY in a dose dependent stimulate the release of incretins has been investigated in more detail
manner (Fig. 1). by Sirinek et al. [62] and Shima et al. [63]. In analogy to our ndings,
Taken together, these data suggest that gut-expressed sweet-taste they demonstrate that only glucose and, to a lesser extent, galactose,
receptors may be functionally involved in glucose-stimulated secre- but not fructose or mannose can stimulate the release of GLP-1 and
tion of GI peptides with plasticity of receptor expression to adapt to GIP. They propose a special sugar sensor with specic steric require-
different nutritional situations. ments which may be involved in the secretion of these peptides
[62,63].
4.1.3.3. Limitations. We and other groups investigated whether Taken together, the current data suggest that additional non-taste
equisweet solutions of carbohydrate sugars or articial sweetener chemosensory mechanisms directed towards certain nutrient mole-
have equivalent effects on the release of gut peptides [5760]. cules (e.g. glucose, a major fuel for the body) may exist to modulate
Interestingly, we found that in contrast to the in vitro experiments, the the secretion of gut peptides (Fig. 2). Also, potential energy sensing
equisweet solutions of either glucose, fructose or articial sweetener mechanisms or energy thresholds might exist for the secretory
had different effects on gut peptide secretion: only glucose potently responses, albeit it is unlikely that the release is directly related to the
stimulated the secretion of GLP-1 and PYY; in contrast fructose was caloric load in a dose response manner.
much less effective and articial sweetener had no signicant effect In addition, gut-expressed sweet-taste receptors may have
[57]. We infer from these observations that activation of the taste regulatory function aside from gut peptide secretion. The two main
receptor pathway alone is not sufcient to stimulate GLP-1 and PYY mechanisms of intestinal glucose absorption into enterocytes (SGLT1
secretion. The ndings that fructose was much less effective in and GLUT2) also appear to be regulated by sweet-taste receptor
stimulating peptides secretion than equisweet glucose further signaling. Margolskee et al. [54] showed in experiments in mice that
suggests that different molar loads (25 g fructose vs. 50 g glucose) SGLT-1 is upregulated by a high sugar diet or when fed articial
may affect peptide secretion differentially. However, previous human sweetener; in contrast mice decient in either -gustducin or T1R3
data document that GLP-1 responses to equal doses (75 g) of either failed to increase SGLT-1 expression.
fructose or glucose is less for fructose [61]. The relationship between
4.2. Lipids

Lipids are major stimuli for the secretion of GI peptides but the
mechanisms of fat-triggered hormone release remain largely un-
known. Long-chain free fatty acids (FFA) (chain length NC12) potently
trigger the release of GLP-1, PYY and CCK. In contrast, C11 or shorter
FFA does not, suggesting that a molecular recognition system for FFA
resides within the GI tract [64,65].
One potential mechanism involves the activation of specic GPCRs,
such as GPR40, GPR41, GPR43, GPR119 and GPR120. GPR40 and
GPR120 are responsive to medium and long-chain FFA [6669]. In
contrast, GPR43 and GPR41 are activated by short chain FFAs such as
acetate or butyrate [69]. GPR40 has been identied in EEC in mice
where it colocalizes predominantly with GLP-1 and GIP (less with
CCK, PYY and ghrelin) suggesting that it mediates FFA stimulated
incretin secretion [66]. GPR41 and 43 have been identied in rat
endocrine cells containing PYY and in mucosal mast cells, which
contain 5-hydroxytryptamine (5-HT). This is consistent with data
demonstrating that FFAs stimulate the release of PYY and 5-HT from
rat ileum and colon [70,71]. In addition, GPR41 decient knockout
mice showed impaired expression of PYY and increased intestinal
transit rate [72]. GPR119 is found in rodent and human L- and K-cells
and is responsive to oleoylethanolamide (OEA), an endogenous fatty
acid derivative [73]. Recent studies have reported that OEA and a
novel GPR119 agonists stimulate the secretion of GLP-1 and GIP in
vitro and in vivo in rodents [74,75].
The best evidence for a role of GPCRs in the sensing of intestinal
lipids is presently available for GPR120. This receptor is abundantly
expressed in human and mouse intestine, as well as in STC-1 cells and
its stimulation has been shown to promote the secretion of GLP-1 and
CCK in mice and humans [67,68,76,77]. Tsujimoto and coworkers
showed that in mice GPR120 is colocalized with GLP-1 predominantly
in colonic endocrine cells. In addition, he showed that colonic, but not
duodenal or ileal administration of -linoleic acid induced a
Fig. 1. The functional involvement of gut-expressed sweet-taste receptors in glucose-
signicant increase in circulating levels of GLP-1, and therefore
stimulated secretion of glucagon-like peptide (GLP-1) and peptide tyrosine tyrosine
(PYY) was determined by intragastric infusion of 75 g glucose with or without 150, 300 concluded that lipids contained in food may contribute to the later
and 450 ppm lactisole (a sweet-taste receptor antagonist) in a double blind, 4-way phase of GLP-1 secretion by direct effects on distal L-cells [68,76].
crossover trial including 8 healthy fasted subjects. Lactisole induced a dose-dependent Moreover, FFA-triggered secretion of GLP-1 was markedly reduced in
reduction in glucose-stimulated secretion of GLP-1 and PYY; the reduction was noted GPR120 knockout mice (GPR120/) [68].
with all three concentrations (the 300 ppm dose is not shown for visual clarity). The
area under the curve for the plasma concentration-time prole of GLP-1 was
We also found GPR120 to be expressed mainly in distal segments
signicantly reduced with 450 ppm lactisole (P = 0.043); in contrast, PYY secretion of the human GI tract (Fig. 3), again suggesting that GPR120 is
only trended towards a reduction (P = 0.066) due to the limited number of subjects. implicated in the later phase of GI peptide secretion; perhaps involved
66 R.E. Steinert, C. Beglinger / Physiology & Behavior 105 (2011) 6270

Fig. 2. Summary model depicting the ndings that glucose-stimulated secretion of GLP-1 and PYY can blocked by a sweet-taste antagonist suggesting the functional involvement of
the sweet-taste receptor sweet-pathway in glucose-stimulated secretion of these peptides. In contrast, sweetness from articial sweetener (AS) does not affect peptide secretion
suggesting that the activation of the taste receptor pathway alone is not sufcient to stimulate GLP-1 and PYY secretion.

in the ileal break, a physiological mechanism by which the presence of important for the production of OEA; in addition, it is thought to
nutrient in ileal parts delay gastric emptying and thereby reduce function as specic lipid-taste receptor in lingual epithelium [7981].
nutrient delivery to the small intestine. However, the majority of To what extend FATP4 is involved in GI chemosensitivity, however, is
ingested food does not transit to distal segments of the small intestine, not clear [23]. Finally, the formation of chylomicron components such
but is absorbed within the proximal parts; therefore the physiological as apo A-IV in response to long chain FFA uptake appear to be critical
relevance of these direct effects remains unclear. for the secretion of intestinal CCK suggesting an alternative lipid
Two further proteins that are thought to be involved in lipid sensing pathway [81].
sensing include the fatty acid transport protein (FATP4) and the fatty
acid translocase (FAT CD36). Both are membrane proteins that 4.3. Proteins
facilitate long-chain FFA uptake into mammalian cells [78]. CD36 is
Although the products of protein hydrolysis, i.e. peptones and
amino acids also stimulate secretion of GI peptides from EEC, the exact
mechanisms by which these molecules are detected remain elusive
[82]. Like for glucose- and lipid-sensing, several mechanisms have
been proposed. Choi et al. [83] demonstrated a role for GPR93:
increasing the expression of GPR93 in STC-1 cells increased CCK
mRNA expression and CCK secretion. In addition, it is thought that
glutamine, a potent stimulus for GLP-1 release in GLUTag cells, may
act in part via the sodium-dependent amino-acid transporter SLC38A2
[38,84]. It is proposed that electrogenic nutrient transporters trigger
cell excitability by small inward currents and that this depolarizing
effect contributes to peptide release from EEC. This may also include
the oligopeptide transporting systems PEPT1 and PEPT2 [38,84].
Matsumura et al. [85] showed that transfection of PEPT1 in STC-1 cells
evokes membrane depolarization and di-peptide-stimulated hor-
mone secretion in a pH-dependent manner. Finally, there is evidence
that the extracellular Ca2+ sensing receptor (CaR) may account for
Fig. 3. mRNA expression of GPR120 in different gut segments normalized to 18S
expression. Data represent means SEM of biopsies from 9 healthy subjects. Signi-
sensing mechanisms in the gut. Several studies show CaR expression
cant differences compared to duodenum are indicated by *, p b 0.05; **, p b 0.01; along the small and large intestine and suggest potential roles in
***, p b 0.001. signaling protein availability [86].
R.E. Steinert, C. Beglinger / Physiology & Behavior 105 (2011) 6270 67

5. Interactions between entero-endocrine cells and muscarinic or nicotinic receptors to stimulate the secretion of GLP-1
visceral afferents [100,105,107].
An alternative explanation for the early-phase secretion of GLP-1
The secretory products of EEC are released upon stimulation (e.g. (and here in particular for the more rapid and shorter lived glucose-
food ingestion) by exocytosis at the basolateral membrane into the stimulated GLP-1 release) is that luminal glucose in the proximal gut
interstitial space of the lamina propria. They either interact locally in a directly stimulates GLP-1 secretion [50,108,109]. 1) The time course of
paracrine fashion on primary afferent neurons and other cells, or they glucose-triggered secretion of GLP-1 in humans is consistent with
enter the systemic circulation via capillaries draining into the hepatic glucose reaching the proximal intestine [101]. 2) In vitro studies using
portal vein or more slowly via the lymph to act in an endocrine GLUTag, STC-1 or NCIH716 cell culture models demonstrate that EEC
fashion [23,87]. Because many of the gut peptides are subject to a high are themselves glucose sensitive [26,27], and 3) proximal entero-
liver extraction and a rapid breakdown by proteolytic enzymes, endocrine L-cells have been shown to express different chemosensory
largest concentrations are found very near the site of secretion, i.e. in proteins such as sweet-taste receptors and -gustducin to sense
the lamina propria of the mucosa, the submucosa and in the hepatic glucose [50]. Thus, the limited number of L-cells present in duodenum
portal vein rather than in the systemic circulation [8891]. This is an and proximal jejunum could account for the early phase of
important consideration when evaluating their physiological actions. postprandial GLP-1 in particular in response to carbohydrate meals.
In this regard, it has been suggested that gut peptides such as GLP- In contrast, direct contact of nutrients in the distal small intestine
1 or PYY3-36 exert their anorectic effects mainly via receptors close to may account for the second-phase of GLP-1 secretion and here
the site of secretion on receptors on vagal afferent bers innervating for the effects of lipids which are stronger, slower in onset but last
the lamina propria. It has been demonstrated that the anorectic effect longer. As mentioned earlier, GPR120, a fatty acid GPCR has been
of intraperitoneal administration of GLP-1 or PYY3-36 in rodents is implicated in the later phase of gut peptide secretion [68], however,
blocked by total abdominal vagotomy or subdiaphragmatic vagal the physiological relevance of these direct effects remains unclear
deafferentiation (SDA) [92,93]. In addition, there is solid evidence that because the majority of ingested food does not transit to distal
GLP-1 receptors in the hepatic portal region contribute to the effect of intestinal segments.
GLP-1 on peripheral glucose handling in rodents [94]. Very recently, Alternatively, the second phase secretion may depend on the
also intestinal CCK has been implicated in the regulation of glucose production of metabolites from non-absorbable food by the gut
homeostasis via a gut-brain-liver neuronal axis independently of microora, most importantly short chain free fatty acids such as
changes in circulating insulin levels [95]. The authors propose that in acetate, propionate or butyrate [80]. In addition, bile acids have
response to gut lipid sensing, intestinal CCK activates CCK-1 receptors recently been suggested to function as complex metabolic regulators
on visceral vagal afferents which relay a signal via the brain to the and not just simple lipid solubilizers [110]. Katsuma et al. [111]
liver to directly inhibit glucose production [95]. In line with these showed that bile acids promote GLP-1 secretion through TGR5 in
ndings, a recent study using functional magnetic resonance imaging entero-endocrine STC-1 cells implicating that bile acids may function
conducted in humans showed that C12 FFAs increase brainstem- and as mediators of lipid-stimulated secretion of GLP-1 within the
hypothalamic-activity within 24 min following infusion, and that enterohepatic recirculation.
this effect is dependent on CCK-1 receptors [96].
Finally, a role of the lymphatic transport of gut peptides has been 7. Summary and conclusions
suggested as mechanism to protect GI peptides such as GLP-1 from
degradation. Tso and coworkers [97,98] demonstrated that postpran- Nutrient sensing in the gut represents a complex interplay
dially GLP-1 concentrations in intestinal lymph are up to 10-fold between EEC and visceral afferents. Digestive processes such as
higher than in portal blood. In contrast to insulin that enters the pancreatic secretion, gastric emptying or even meal termination are
lymph mainly by ltration from capillaries, it appears that GLP-1 is regulated by chemical components of ingested food that either
partly secreted directly into visceral lacteals [97]. Also, the volume in (i) trigger the secretion of peripheral humoral factors such as CCK,
which GLP-1 is diluted is less in lymph and the concentration of GLP-1 or PYY or (ii) activate independently (or as downstream target
dipeptidylpeptidase IV (DPP-IV), a protease that leads to the rapid of EEC products) primary afferent neurons. The chemosensory signals
inactivation of GLP-1, is considerably lower in lymph. The physiolog- are initiated by a complex machinery that mediates different taste
ical relevance of the lymphatic transport of gut peptides, however, modalities with specic chemosensory receptors in the stomach,
awaits further investigation. duodenum and the small and large intestine. Recent examples of the
specic receptor systems include the sweet-taste receptor hetero-
6. Chemosensory mechanisms and secretion patterns of dimer T1R2/T1R3 or receptors for medium and long-chain FFA such as
gut peptides GPR120.
Despite the increasing scientic interest and endeavor in this
Gut hormone secretion by nutrients provide an example of research area, one of the major problems in studying nutrient sensing,
different mechanisms of luminal chemosensory mechanisms. however, remains that EEC are extremely difcult to study in
For example GLP-1 shows a biphasic secretion pattern, with an particular in humans. 1) EEC are sparse and irregular localized along
early-phase secretion within 1530 min after meal onset and a the gut and it is not possible to obtain direct access in humans.
prolonged second phase from 60 min up to 2 or 3 hours [89,99103]. 2) Measurements of hormone plasma level are only an indirect
The time course of glucose-stimulated GLP-1 secretion is more rapid measure of EEC function and their secretory responses to specic
and shorter lived, whereas the effects of lipids are stronger, slower in nutrients 3) Local paracrine interaction between EEC and vagal
onset but last longer [28,100,101,104106]. Different mechanisms afferents are not reected in hormone plasma level. Therefore, most
have been proposed to account for the different secretory responses. evidence for the mechanisms underlying GI nutrient sensing comes
A neurohumoral proximal to distal loop is proposed to account for from in vitro or animal studies using specic knockout strains. In
the early-phase secretion because nutrients do not reach the distal evaluating these ndings, it is, however, important to recall that these
intestine, where the majority of L-cells is present, within 30 min after data are obtained mainly from transformed cells or transgenic mice
ingestion [100,101]. CCK or GIP secreted from the proximal small and it is not clear whether all ndings represent normal in vivo
intestine are possible candidates for mediating the stimulation of functions of EEC. The recent discrepancies on the effects of articial
distal L-cells [64,100]. Other studies have also suggested the in- sweetener-stimulated GLP-1 secretion between humans and in vitro
volvement of vagal sensory neurons and neural pathways with M1 studies are only one example emphasizing this issue.
68 R.E. Steinert, C. Beglinger / Physiology & Behavior 105 (2011) 6270

Why is it important to have a particular focus on studies conducted [12] Raybould H. Does your gut taste? Sensory transduction in the gastrointestinal
tract. News Physiol Sci Dec. 1998;13:27580.
in humans? Considering the epidemic proportions of obesity and its [13] Iggo A. Gastric mucosal chemoreceptors with vagal afferent bres in the cat. Q J
co-morbidities such as diabetes and cardiovascular disease, the Exp Physiol Cogn Med Sci Oct. 1957;42(4):398409.
pharmacological features of some of the gut peptides including CCK, [14] Berthoud H, Kressel M, Raybould H, Neuhuber W. Vagal sensors in the rat
duodenal mucosa: distribution and structure as revealed by in vivo DiI-tracing.
GLP-1 or PYY3-36 are of considerable interest in the development of Anat Embryol (Berl) Mar. 1995;191(3):20312.
effective clinical treatment: 1) CCK, GLP-1 or PYY3-36 are anorexi- [15] Hofer D, Asan E, Drenckhahn D. Chemosensory perception in the gut. News
genic (satiating) and this effect is seen not only in healthy subjects, Physiol Sci Feb. 1999;14:1823.
[16] Smith G, Jerome C, Cushin B, Eterno R, Simansky K. Abdominal vagotomy blocks
but is preserved in obese or diabetic persons [28,87,112114]. 2) Data the satiety effect of cholecystokinin in the rat. Science Aug. 1981;213(4511):
show that fasting and postprandial plasma levels of these peptides are 10367.
much lower in obese than in lean subjects suggesting that a deciency [17] Koda S, Date Y, Murakami N, Shimbara T, Hanada T, Toshinai K, et al. The role of
the vagal nerve in peripheral PYY3-36-induced feeding reduction in rats.
in circulating levels is associated with the pathogenesis of obesity
Endocrinology May 2005;146(5):236975.
[114116]. Moreover, GLP-1 functions as a physiological incretin [18] Nakagawa A, Satake H, Nakabayashi H, Nishizawa M, Furuya K, Nakano S, et al.
hormone that enhances postprandial insulin release, suppresses Receptor gene expression of glucagon-like peptide-1, but not glucose-dependent
glucagon secretion, delays gastric emptying and thereby improves insulinotropic polypeptide, in rat nodose ganglion cells. Auton Neurosci Jan.
2004;110(1):3643.
the postprandial glucose prole [89]. [19] Zhang X, Shi T, Holmberg K, Landry M, Huang W, Xiao H, et al. Expression and
The question is whether it is possible to directly increase the regulation of the neuropeptide Y Y2 receptor in sensory and autonomic ganglia.
secretion of these peptides from endogenous stores using specic Proc Natl Acad Sci USA Jan. 1997;94(2):72934.
[20] Date Y, Murakami N, Toshinai K, Matsukura S, Niijima A, Matsuo H, et al. The role
stimuli. For example, the development of specic secretagogues to of the gastric afferent vagal nerve in ghrelin-induced feeding and growth
increase plasma levels of GLP-1, analogous to the use of sulfonylureas, hormone secretion in rats. Gastroenterology Oct. 2002;123(4):11208.
could induce an increase in insulin secretion for the treatment of type [21] Burdyga G, Lal S, Varro A, Dimaline R, Thompson D, Dockray G. Expression of
cannabinoid CB1 receptors by vagal afferent neurons is inhibited by cholecys-
2 diabetes. tokinin. J Neurosci Mar. 2004;24(11):270815.
In addition, the benecial effects of Roux-en-Y bypass surgery on [22] Page A, O'Donnell T, Blackshaw L. Inhibition of mechanosensitivity in visceral
postprandial satiation, weight loss and diabetes are thought to be due, primary afferents by GABAB receptors involves calcium and potassium channels.
Neuroscience 2006;137(2):62736.
at least in part, to the increased GLP-1, PYY and CCK secretions seen [23] Berthoud H. Vagal and hormonal gutbrain communication: from satiation to
after such surgery [112,116]. If endogenous secretion of these satisfaction. Neurogastroenterol Motil May 2008;20(Suppl 1):6472.
peptides could be modulated without serious anatomic and irrevers- [24] Raybould H. Gut chemosensing: interactions between gut endocrine cells and
visceral afferents. Auton Neurosci Aug. 2010;153(1-2):416.
ible interventions such as Roux-en-Y bypass, it would be a major
[25] Sbarbati A, Osculati F. A new fate for old cells: brush cells and related elements.
break-through in diabetes and obesity treatment. At present, J Anat Apr. 2005;206(4):34958.
however, the exact chemosensory mechanisms and their functional [26] Abello J, Ye F, Bosshard A, Bernard C, Cuber J, Chayvialle J. Stimulation of
involvements in the secretion of gut peptides remain poorly glucagon-like peptide-1 secretion by muscarinic agonist in a murine intestinal
endocrine cell line. Endocrinology May 1994;134(5):20117.
understood and more research will be required in humans. [27] Drucker D, Jin T, Asa S, Young T, Brubaker P. Activation of proglucagon gene
transcription by protein kinase-A in a novel mouse enteroendocrine cell line. Mol
Endocrinol Dec. 1994;8(12):164655.
8. Funding [28] Beglinger C, Degen L. Gastrointestinal satiety signals in humansphysiologic
roles for GLP-1 and PYY? Physiol Behav Nov. 2006;89(4):4604.
[29] Karhunen L, Juvonen K, Huotari A, Purhonen A, Herzig K. Effect of protein, fat,
The work was supported in part by a grant of the Swiss National carbohydrate and bre on gastrointestinal peptide release in humans. Regul Pept
Science Foundation (Grant No. 320000118330) and an unrestricted Aug. 2008;149(13):708.
grant of Hoffmann-La Roche. [30] Raben A, Agerholm-Larsen L, Flint A, Holst J, Astrup A. Meals with similar energy
densities but rich in protein, fat, carbohydrate, or alcohol have different effects on
energy expenditure and substrate metabolism but not on appetite and energy
intake. Am J Clin Nutr Jan. 2003;77(1):91100.
Acknowledgements
[31] Gribble F, Williams L, Simpson A, Reimann F. A novel glucose-sensing mechanism
contributing to glucagon-like peptide-1 secretion from the GLUTag cell line.
We thank the team of the Clinical Research Center and Department Diabetes May 2003;52(5):114754.
[32] Fushiki T, Kojima A, Imoto T, Inoue K, Sugimoto E. An extract of Gymnema sylvestre
of Biomedicine Mrs. Claudia Blsi, Mrs. Sylvia Ketterer and Mrs.
leaves and puried gymnemic acid inhibits glucose-stimulated gastric inhibitory
Gerdien Gamboni for expert technical assistance. peptide secretion in rats. J Nutr Dec. 1992;122(12):236773.
[33] Wachters-Hagedoorn R, Priebe M, Heimweg J, Heiner A, Englyst K, Holst J, et al.
The rate of intestinal glucose absorption is correlated with plasma glucose-
References dependent insulinotropic polypeptide concentrations in healthy men. J Nutr Jun.
2006;136(6):15116.
[1] McHugh PR, Moran TH. Calories and gastric emptying: a regulatory capacity with [34] Cani P, Holst J, Drucker D, Delzenne N, Thorens B, Burcelin R, et al. GLUT2 and the
implications for feeding. Am J Physiol May 1979;236(5):R25460. incretin receptors are involved in glucose-induced incretin secretion. Mol Cell
[2] Creutzfeldt W. Insulinotropic factors of the gutthe broadening incretin concept. Endocrinol Sep. 2007;276(12):1823.
Gastroenterology Jun. 1980;78(6):16312. [35] Burdakov D, Luckman SM, Verkhratsky A. Glucose-sensing neurons of the
[3] Uneyama H, Takeuchi K. New frontiers in gut nutrient sensor researchfrom hypothalamus. Philos Trans R Soc Lond B Biol Sci Dec. 29 2005;360(1464):222735.
taste physiology to gastroenterology: preface. J Pharmacol Sci Jan. 2010;112(1): [36] Miki T, Seino S. Roles of KATP channels as metabolic sensors in acute metabolic
67. changes. J Mol Cell Cardiol Jun. 2005;38(6):91725.
[4] Bayliss W, Starling E. The mechanism of pancreatic secretion. J Physiol Sep. [37] Rorsman P. The pancreatic beta-cell as a fuel sensor: an electrophysiologist's
1902;28(5):32553. viewpoint. Diabetologia May 1997;40(5):48795.
[5] Fujita T, Kobayashi S. Structure and function of gut endocrine cells. Int Rev Cytol [38] Tolhurst G, Reimann F, Gribble F. Nutritional regulation of glucagon-like peptide-
Suppl 1977;6:187233. 1 secretion. J Physiol Jan. 2009;587(Pt 1):2732.
[6] Blackshaw L, Grundy D, Scratcherd T. Involvement of gastrointestinal mechano- [39] Reimann F, Habib A, Tolhurst G, Parker H, Rogers G, Gribble F. Glucose sensing in
and intestinal chemoreceptors in vagal reexes: an electrophysiological study. J L cells: a primary cell study. Cell Metab Dec. 2008;8(6):5329.
Auton Nerv Syst Mar. 1987;18(3):22534. [40] Parker H, Habib A, Rogers G, Gribble F, Reimann F. Nutrient-dependent secretion
[7] Mei N. Intestinal chemosensitivity. Physiol Rev Apr. 1985;65(2):21137. of glucose-dependent insulinotropic polypeptide from primary murine K cells.
[8] Furness J, Kunze W, Clerc N. Nutrient tasting and signaling mechanisms in the Diabetologia Feb. 2009;52(2):28998.
gut. II. The intestine as a sensory organ: neural, endocrine, and immune [41] Ritzel U, Fromme A, Ottleben M, Leonhardt U, Ramadori G. Release of glucagon-
responses. Am J Physiol Nov. 1999;277(5 Pt 1):G922-8. like peptide-1 (GLP-1) by carbohydrates in the perfused rat ileum. Acta Diabetol
[9] Furness J, Jones C, Nurgali K, Clerc N. Intrinsic primary afferent neurons and nerve Mar. 1997;34(1):1821.
circuits within the intestine. Prog Neurobiol Feb. 2004;72(2):14364. [42] Chandrashekar J, Hoon M, Ryba N, Zuker C. The receptors and cells for
[10] Furness J, Kunze W, Bertrand P, Clerc N, Bornstein J. Intrinsic primary afferent mammalian taste. Nature Nov. 2006;444(7117):28894.
neurons of the intestine. Prog Neurobiol Jan. 1998;54(1):118. [43] Hofer D, Puschel B, Drenckhahn D. Taste receptor-like cells in the rat gut
[11] Berthoud H, Neuhuber W. Functional and chemical anatomy of the afferent vagal identied by expression of alpha-gustducin. Proc Natl Acad Sci USA Jun. 25
system. Auton Neurosci Dec. 2000;85(13):117. 1996;93(13):66314.
R.E. Steinert, C. Beglinger / Physiology & Behavior 105 (2011) 6270 69

[44] Wu S, Rozengurt N, Yang M, Young S, Sinnett-Smith J, Rozengurt E. Expression acid binding G protein-coupled receptor, Gpr41. Proc Natl Acad Sci USA Oct.
of bitter taste receptors of the T2R family in the gastrointestinal tract and 2008;105(43):1676772.
enteroendocrine STC-1 cells. Proc Natl Acad Sci USA Feb. 2002;99(4):23927. [73] Overton H, Babbs A, Doel S, Fyfe M, Gardner L, Grifn G, et al. Deorphanization of
[45] Rozengurt E, Sternini C. Taste receptor signaling in the mammalian gut. Curr Opin a G protein-coupled receptor for oleoylethanolamide and its use in the discovery
Pharmacol Dec. 2007;7(6):55762. of small-molecule hypophagic agents. Cell Metab Mar. 2006;3(3):16775.
[46] Dyer J, Salmon K, Zibrik L, Shirazi-Beechey S. Expression of sweet taste receptors [74] Chu Z, Carroll C, Alfonso J, Gutierrez V, He H, Lucman A, et al. A role for intestinal
of the T1R family in the intestinal tract and enteroendocrine cells. Biochem Soc endocrine cell-expressed g protein-coupled receptor 119 in glycemic control by
Trans Feb. 2005;33(Pt 1):3025. enhancing glucagon-like peptide-1 and glucose-dependent insulinotropic pep-
[47] Sternini C, Anselmi L, Rozengurt E. Enteroendocrine cells: a site of taste in tide release. Endocrinology May 2008;149(5):203847.
gastrointestinal chemosensing. Curr Opin Endocrinol Diab Obes Feb. 2008;15(1): [75] Lauffer L, Iakoubov R, Brubaker P. GPR119 is essential for oleoylethanolamide-
738. induced glucagon-like peptide-1 secretion from the intestinal enteroendocrine
[48] Rozengurt N, Wu S, Chen M, Huang C, Sternini C, Rozengurt E. Colocalization of L-cell. Diabetes May 2009;58(5):105866.
the alpha-subunit of gustducin with PYY and GLP-1 in L cells of human colon. Am [76] Adachi T, Tanaka T, Takemoto K, Koshimizu T, Hirasawa A, Tsujimoto G. Free fatty
J Physiol Gastrointest Liver Physiol Nov. 2006;291(5):G792802. acids administered into the colon promote the secretion of glucagon-like
[49] Sutherland K, Young R, Cooper N, Horowitz M, Blackshaw L. Phenotypic peptide-1 and insulin. Biochem Biophys Res Commun Feb. 2006;340(1):3327.
characterization of taste cells of the mouse small intestine. Am J Physiol [77] Tanaka T, Katsuma S, Adachi T, Koshimizu T, Hirasawa A, Tsujimoto G. Free fatty
Gastrointest Liver Physiol May 2007;292(5):G14208. acids induce cholecystokinin secretion through GPR120. Naunyn-Schmiedebergs
[50] Jang H, Kokrashvili Z, Theodorakis M, Carlson O, Kim B, Zhou J, et al. Gut- Arch Pharmacol Jun. 2008;377(46):5237.
expressed gustducin and taste receptors regulate secretion of glucagon-like [78] Abumrad N, Coburn C, Ibrahimi A. Membrane proteins implicated in long-chain
peptide-1. Proc Natl Acad Sci USA Sep. 2007;104(38):1506974. fatty acid uptake by mammalian cells: CD36, FATP and FABPm. Biochim Biophys
[51] Hofer D, Drenckhahn D. Identication of the taste cell G-protein, alpha- Acta Oct. 1999;1441(1):413.
gustducin, in brush cells of the rat pancreatic duct system. Histochem Cell Biol [79] Laugerette F, Passilly-Degrace P, Patris B, Niot I, Febbraio M, Montmayeur J, et al.
Sep. 1998;110(3):3039. CD36 involvement in orosensory detection of dietary lipids, spontaneous
[52] Hofer D, Drenckhahn D. Identication of brush cells in the alimentary and fat preference, and digestive secretions. J Clin Invest Nov. 2005;115(11):
respiratory system by antibodies to villin and mbrin. Histochemistry Nov. 317784.
1992;98(4):23742. [80] Delzenne N, Blundell J, Brouns F, Cunningham K, De Graaf K, Erkner A, et al.
[53] Gebhard A, Gebert A. Brush cells of the mouse intestine possess a specialized Gastrointestinal targets of appetite regulation in humans. Obes Rev Mar.
glycocalyx as revealed by quantitative lectin histochemistry. Further evidence for 2010;11(3):23450.
a sensory function. J Histochem Cytochem Jun. 1999;47(6):799808. [81] Little TJ, Feinle-Bisset C. Oral and gastrointestinal sensing of dietary fat and
[54] Margolskee R, Dyer J, Kokrashvili Z, Salmon K, Ilegems E, Daly K, et al. T1R3 and appetite regulation in humans: modication by diet and obesity. Front Neurosci
gustducin in gut sense sugars to regulate expression of Na+glucose 2010;4:178.
cotransporter 1. Proc Natl Acad Sci USA Sep. 2007;104(38):1507580. [82] Dockray G. Luminal sensing in the gut: an overview. J Physiol Pharmacol Dec.
[55] Chen M, Wu S, Reeve JJ, Rozengurt E. Bitter stimuli induce Ca2+ signaling and 2003;54(Suppl 4):917.
CCK release in enteroendocrine STC-1 cells: role of L-type voltage-sensitive Ca2+ [83] Choi S, Lee M, Shiu A, Yo S, Halldn G, Aponte G. GPR93 activation by protein
channels. Am J Physiol Cell Physiol Oct. 2006;291(4):C72639. hydrolysate induces CCK transcription and secretion in STC-1 cells. Am J Physiol
[56] Young R, Sutherland K, Pezos N, Brierley S, Horowitz M, Rayner C, et al. Gastrointest Liver Physiol May 2007;292(5):G136675.
Expression of taste molecules in the upper gastrointestinal tract in humans with [84] Reimann F, Williams L, da Silva Xavier G, Rutter G, Gribble F. Glutamine potently
and without type 2 diabetes. Gut Mar. 2009;58(3):33746. stimulates glucagon-like peptide-1 secretion from GLUTag cells. Diabetologia
[57] Steinert RE, Frey F, Topfer A, Drewe J, Beglinger C. Effects of carbohydrate sugars Sep. 2004;47(9):1592601.
and articial sweeteners on appetite and the secretion of gastrointestinal satiety [85] Matsumura K, Miki T, Jhomori T, Gonoi T, Seino S. Possible role of PEPT1 in gastro-
peptides. Br J Nutr Jan. 2011;24:19. intestinal hormone secretion. Biochem Biophys Res Commun Nov. 2005;336(4):
[58] Ma J, Bellon M, Wishart J, Young R, Blackshaw L, Jones K, et al. Effect of the 102832.
articial sweetener, sucralose, on gastric emptying and incretin hormone release [86] Hebert S, Cheng S, Geibel J. Functions and roles of the extracellular Ca2+-sensing
in healthy subjects. Am J Physiol Gastrointest Liver Physiol Apr. 2009;296(4): receptor in the gastrointestinal tract. Cell Calcium Mar. 2004;35(3):23947.
G7359. [87] Woods S. Gastrointestinal satiety signals I. An overview of gastrointestinal
[59] Fujita Y, Wideman R, Speck M, Asadi A, King D, Webber T, et al. Incretin release signals that inuence food intake. Am J Physiol Gastrointest Liver Physiol Jan.
from gut is acutely enhanced by sugar but not by sweeteners in vivo. Am J Physiol 2004;286(1):G7G13.
Endocrinol Metab Mar. 2009;296(3):E4739. [88] Deacon C, Pridal L, Klarskov L, Olesen M, Holst J. Glucagon-like peptide 1
[60] Little T, Gupta N, Case R, Thompson D, McLaughlin J. Sweetness and bitterness undergoes differential tissue-specic metabolism in the anesthetized pig. Am J
taste of meals per se does not mediate gastric emptying in humans. Am J Physiol Physiol Sep. 1996;271(3 Pt 1):E45864.
Regul Integr Comp Physiol Sep. 2009;297(3):R6329. [89] Holst JJ. The physiology of glucagon-like peptide 1. Physiol Rev Oct. 2007;87(4):
[61] Kong M, Chapman I, Goble E, Wishart J, Wittert G, Morris H, et al. Effects of oral 140939.
fructose and glucose on plasma GLP-1 and appetite in normal subjects. Peptides [90] Eysselein V, Eberlein G, Schaeffer M, Grandt D, Goebell H, Niebel W, et al.
1999;20(5):54551. Characterization of the major form of cholecystokinin in human intestine: CCK-
[62] Sirinek K, Levine B, O'Dorisio T, Cataland S. Gastric inhibitory polypeptide (GIP) 58. Am J Physiol Feb. 1990;258(2 Pt 1):G25360.
release by actively transported, structurally similar carbohydrates. Proc Soc Exp [91] Grandt D, Schimiczek M, Struk K, Shively J, Eysselein V, Goebell H, et al.
Biol Med Jul. 1983;173(3):37985. Characterization of two forms of peptide YY, PYY(136) and PYY(336), in the
[63] Shima K, Suda T, Nishimoto K, Yoshimoto S. Relationship between molecular rabbit. Peptides 1994;15(5):81520.
structures of sugars and their ability to stimulate the release of glucagon-like [92] Ruttimann EB, Arnold M, Hillebrand JJ, Geary N, Langhans W. Intrameal hepatic
peptide-1 from canine ileal loops. Acta Endocrinol (Copenh) Oct. 1990;123(4): portal and intraperitoneal infusions of glucagon-like peptide-1 reduce sponta-
46470. neous meal size in the rat via different mechanisms. Endocrinology Mar.
[64] Beglinger S, Drewe J, Schirra J, Gke B, D'Amato M, Beglinger C. Role of fat 2009;150(3):117481.
hydrolysis in regulating glucagon-like Peptide-1 secretion. J Clin Endocrinol [93] Abbott C, Monteiro M, Small C, Sajedi A, Smith K, Parkinson J, et al. The inhibitory
Metab Feb. 2010;95(2):87986. effects of peripheral administration of peptide YY(336) and glucagon-like
[65] McLaughlin J, Grazia Luc M, Jones M, D'Amato M, Dockray G, Thompson D. Fatty peptide-1 on food intake are attenuated by ablation of the vagalbrainstem
acid chain length determines cholecystokinin secretion and effect on human hypothalamic pathway. Brain Res May 2005;1044(1):12731.
gastric motility. Gastroenterology Jan. 1999;116(1):4653. [94] Vahl TP, Tauchi M, Durler TS, Elfers EE, Fernandes TM, Bitner RD, et al. Glucagon-
[66] Edfalk S, Steneberg P, Edlund H. Gpr40 is expressed in enteroendocrine cells like peptide-1 (GLP-1) receptors expressed on nerve terminals in the portal vein
and mediates free fatty acid stimulation of incretin secretion. Diabetes Sep. mediate the effects of endogenous GLP-1 on glucose tolerance in rats.
2008;57(9):22807. Endocrinology Oct. 2007;148(10):496573.
[67] Hirasawa A, Hara T, Katsuma S, Adachi T, Tsujimoto G. Free fatty acid receptors [95] Cheung GW, Kokorovic A, Lam CK, Chari M, Lam TK. Intestinal cholecystokinin
and drug discovery. Biol Pharm Bull Oct. 2008;31(10):184751. controls glucose production through a neuronal network. Cell Metab Aug.
[68] Hirasawa A, Tsumaya K, Awaji T, Katsuma S, Adachi T, Yamada M, et al. Free fatty 2009;10(2):99109.
acids regulate gut incretin glucagon-like peptide-1 secretion through GPR120. [96] Lassman DJ, McKie S, Gregory LJ, Lal S, D'Amato M, Steele I, et al. Dening the role
Nat Med Jan. 2005;11(1):904. of cholecystokinin in the lipid-induced human brain activation matrix.
[69] Miyauchi S, Hirasawa A, Ichimura A, Hara T, Tsujimoto G. New frontiers in gut Gastroenterology Apr. 2010;138(4):151424.
nutrient sensor research: free fatty acid sensing in the gastrointestinal tract. J [97] D'Alessio D, Lu W, Sun W, Zheng S, Yang Q, Seeley R, et al. Fasting and
Pharmacol Sci Jan. 2010;112(1):1924. postprandial concentrations of GLP-1 in intestinal lymph and portal plasma:
[70] Cherbut C, Ferrier L, Roz C, Anini Y, Blottire H, Lecannu G, et al. Short-chain evidence for selective release of GLP-1 in the lymph system. Am J Physiol Regul
fatty acids modify colonic motility through nerves and polypeptide YY release in Integr Comp Physiol Dec. 2007;293(6):R21639.
the rat. Am J Physiol Dec. 1998;275(6 Pt 1):G141522. [98] Kohan A, Yoder S, Tso P. Lymphatics in intestinal transport of nutrients and
[71] Fukumoto S, Tatewaki M, Yamada T, Fujimiya M, Mantyh C, Voss M, et al. Short- gastrointestinal hormones. Ann NY Acad Sci Oct. 2010;1207(Suppl 1):E4451.
chain fatty acids stimulate colonic transit via intraluminal 5-HT release in rats. [99] Drucker D. The biology of incretin hormones. Cell Metab Mar. 2006;3(3):15365.
Am J Physiol Regul Integr Comp Physiol May 2003;284(5):R126976. [100] Brubaker P, Anini Y. Direct and indirect mechanisms regulating secretion of
[72] Samuel B, Shaito A, Motoike T, Rey F, Backhed F, Manchester J, et al. Effects of glucagon-like peptide-1 and glucagon-like peptide-2. Can J Physiol Pharmacol
the gut microbiota on host adiposity are modulated by the short-chain fatty- Nov. 2003;81(11):100512.
70 R.E. Steinert, C. Beglinger / Physiology & Behavior 105 (2011) 6270

[101] Schirra J, Katschinski M, Weidmann C, Schfer T, Wank U, Arnold R, et al. Gastric [109] Kokrashvili Z, Mosinger B, Margolskee R. Taste signaling elements expressed in
emptying and release of incretin hormones after glucose ingestion in humans. gut enteroendocrine cells regulate nutrient-responsive secretion of gut
J Clin Invest Jan. 1996;97(1):92103. hormones. Am J Clin Nutr Sep. 2009;90(3):822 S-5 S.
[102] Herrmann C, Gke R, Richter G, Fehmann H, Arnold R, Gke B. Glucagon-like [110] Thomas C, Pellicciari R, Pruzanski M, Auwerx J, Schoonjans K. Targeting bile-
peptide-1 and glucose-dependent insulin-releasing polypeptide plasma levels in acid signalling for metabolic diseases. Nat Rev Drug Discov Aug. 2008;7(8):
response to nutrients. Digestion 1995;56(2):11726. 67893.
[103] Rask E, Olsson T, Soderberg S, Johnson O, Seckl J, Holst JJ, et al. Impaired incretin [111] Katsuma S, Hirasawa A, Tsujimoto G. Bile acids promote glucagon-like peptide-1
response after a mixed meal is associated with insulin resistance in nondiabetic secretion through TGR5 in a murine enteroendocrine cell line STC-1. Biochem
men. Diab Care Sep. 2001;24(9):16405. Biophys Res Commun Apr. 1 2005;329(1):38690.
[104] Elliott RM, Morgan LM, Tredger JA, Deacon S, Wright J, Marks V. Glucagon-like [112] Karra E, Chandarana K, Batterham RL. The role of peptide YY in appetite
peptide-1 (736)amide and glucose-dependent insulinotropic polypeptide regulation and obesity. J Physiol Jan. 15 2009;587(Pt 1):1925.
secretion in response to nutrient ingestion in man: acute post-prandial and [113] le Roux CW, Batterham RL, Aylwin SJ, Patterson M, Borg CM, Wynne KJ, et al.
24-h secretion patterns. J Endocrinol Jul. 1993;138(1):15966. Attenuated peptide YY release in obese subjects is associated with reduced
[105] Anini Y, Fu-Cheng X, Cuber JC, Kervran A, Chariot J, Roz C. Comparison of the satiety. Endocrinology Jan. 2006;147(1):38.
postprandial release of peptide YY and proglucagon-derived peptides in the rat. [114] Gutzwiller J, Drewe J, Gke B, Schmidt H, Rohrer B, Lareida J, et al. Glucagon-like
Pugers Arch Aug. 1999;438(3):299306. peptide-1 promotes satiety and reduces food intake in patients with diabetes
[106] Fu-Cheng X, Anini Y, Chariot J, Castex N, Galmiche J, Rosz C. Mechanisms of mellitus type. Am J Physiol May 1999;276(5 Pt 2):R1541-4.
peptide YY release induced by an intraduodenal meal in rats: neural regulation [115] Batterham R, Cohen M, Ellis S, Le Roux C, Withers D, Frost G, et al. Inhibition of food
by proximal gut. Pugers Arch Mar. 1997;433(5):5719. intake in obese subjects by peptide YY3-36. N Engl J Med Sep. 2003;349(10):
[107] Anini Y, Brubaker PL. Muscarinic receptors control glucagon-like peptide 1 9418.
secretion by human endocrine L cells. Endocrinology Jul. 2003;144(7):324450. [116] Peterli R, Wlnerhanssen B, Peters T, Devaux N, Kern B, Christoffel-Courtin C,
[108] Theodorakis M, Carlson O, Michopoulos S, Doyle M, Juhaszova M, Petraki K, et al. et al. Improvement in glucose metabolism after bariatric surgery: comparison of
Human duodenal enteroendocrine cells: source of both incretin peptides, GLP-1 laparoscopic Roux-en-Y gastric bypass and laparoscopic sleeve gastrectomy: a
and GIP. Am J Physiol Endocrinol Metab Mar. 2006;290(3):E5509. prospective randomized trial. Ann Surg Aug. 2009;250(2):23441.

Das könnte Ihnen auch gefallen