You are on page 1of 19

618177

research-article2015
TAE0010.1177/2042018815618177Therapeutic Advances in Endocrinology and MetabolismR Pais, FM Gribble

Therapeutic Advances in Endocrinology and Metabolism Review

Stimulation of incretin secreting cells


Ther Adv Endocrinol
Metab

2016, Vol. 7(1) 2442

DOI: 10.1177/
Ramona Pais, Fiona M. Gribble and Frank Reimann 2042018815618177

The Author(s), 2015.


Reprints and permissions:
Abstract: The incretin hormones glucose-dependent insulinotropic polypeptide (GIP) and http://www.sagepub.co.uk/
glucagon like peptide-1 (GLP-1) are secreted from enteroendocrine cells in the gut and journalsPermissions.nav

regulate physiological and homeostatic functions related to glucose control, metabolism


and food intake. This review provides a systematic summary of the molecular mechanisms
underlying secretion from incretin cells, and an understanding of how they sense and interact
with lumen and vascular factors and the enteric nervous system through transporters and
G-protein coupled receptors (GPCRs) present on their surface to ultimately culminate in
hormone release. Some of the molecules described below such as sodium coupled glucose
transporter 1 (SGLT1), G-protein coupled receptor (GPR) 119 and GPR40 are targets of novel
therapeutics designed to enhance endogenous gut hormone release. Synthetic ligands at these
receptors aimed at treating obesity and type 2 diabetes are currently under investigation.

Keywords: Incretin, Glucagon-like peptide-1 (GLP-1), Glucose-dependent insulinotropic


polypeptide (GIP)

The incretin effect in their totality, contribute ~1% of intestinal epi- Correspondence to:
Frank Reimann,
After the proposal of secretin as a blood-borne thelial cells. In recent years, transgenic technol- Doctorate rer. nat., Dipl.
factor coordinating the arrival of food in the upper Biochem. and
ogy has allowed the development of mice which Fiona M. Gribble, D.Phil,
small intestine with exocrine pancreatic secretion express fluorescent protein reporters under the BM, BCh
[Bayliss and Starling, 1902], a similar hormonal The Wellcome TrustMRC
control of enteroendocrine hormone promoters Institute of Metabolic
axis promoting insulin release from the endocrine [Reimann etal. 2008; Parker etal. 2009; Chandra Science, Metabolic
pancreas was proposed by La Barre in 1932, coin- Research Laboratories,
etal. 2010; Wang etal. 2011; Suzuki etal. 2013]. University of Cambridge,
ing the term incretin. The incretin effect describes This technique enabled the isolation, purification Addenbrookess Hospital,
the fact that, even when the insulin-secreting and Box 289, Hills Road,
and systematic characterization of these otherwise Cambridge, CB2 0QQ, UK
inherently glucose-sensitive pancreatic cell is elusive cells and led to rapid strides in our under- fr222@cam.ac.uk and
exposed to matching blood glucose levels, only fmg23@cam.ac.uk
standing of EEC biology. For one, the concept of
Ramona Pais, Doctorate
approximately half as much insulin is secreted these cells being uni/bi hormonal has been chal- rer. nat, MSc
when glucose is provided by an intravenous infu- lenged and recent evidence points towards them The Wellcome TrustMRC
Institute of Metabolic
sion compared with oral ingestion/intestinal being more plurihormonal than previously Science, Metabolic
absorption of glucose [Perley and Kipnis, 1967]. thought [Egerod et al. 2012; Habib et al. 2012; Research Laboratories,
University of Cambridge,
We now know of a number of gut-derived hor- Sykaras et al. 2014]. Flow cytometric (FACS) Cambridge, UK
mones that boost glucose-dependent insulin analysis and immunostaining have revealed that,
release, including for example, sulfated cholecys- while in the colon, most L cells contained GLP-1
tokinin (CCK8S) [Zawalich etal. 1986], but the and PYY, the picture is different in the upper
true incretins known to date are glucose-depend- small intestine. Most small intestinal L cells con-
ent insulinotropic polypeptide (GIP) and gluca- tained CCK, ~10% were GIP positive and ~20%
gon like peptide-1 (GLP-1) [Dupre et al. 1973; were PYY positive [Habib etal. 2012]. Recently,
Kreymann etal. 1987]. GIP and GLP-1 are pro- we identified insulin-like peptide-5 (INSL5) to be
duced and secreted from specialized endocrine a product of colonic but not small intestinal L
cells of the gut, enteroendocrine cells (EECs), cells and showed that its levels were increased in
namely K and L cells, respectively (Figure 1).
calorie-restricted mice and reduced after feeding.
INSL5 administration increased food intake in
Incretin secreting cells wildtype mice but not in mice lacking its receptor
K and L cells are subsets of enteroendocrine cells RXFP4, contrasting with the anorexic properties
found scattered in the intestinal epithelium that, of other L-cell hormones [Grosse etal. 2014].

24 http://tae.sagepub.com
R Pais, FM Gribble et al.

Figure 1. Incretin hormone secretion from K and L cells.


Glucose dependent insulinotropic polypeptide (GIP) is secreted from K cells, which are predominantly found in the
duodenum, whereas glucagon-like peptide-1 (GLP-1) is secreted from L cells, which increase in numbers in the distal
intestine. Both cell types are so-called open enteroendocrine cells with direct contact to the intestinal lumen, allowing
sampling of the chyme. Nutrient transporters expressed in K and L cells play an important part in this chemosensation
(see text for details). The difference in location, however, is likely to impact on the observed hormone responses to nutrient
ingestion. It is now thought that sufficient L cells are present in the proximal intestine to account for early rises of GLP-1
under normal conditions (left hand side). However, when absorption is blocked (right hand side), the increased delivery to
the distal intestine brings more L cells in contact with nutrients and/or their fermentation/putrification products, resulting
in elevated GLP-1 levels. This in turn slows gastric emptying, potentially thereby prolonging exposure of proximal cells to
chyme. While glucose stimulated GIP secretion is thus abolished in Sglt-1 knockout mice, emphasizing the importance of
this transporter in K cells, GIP (as well as GLP-1) levels in B0at1 knockout animals are elevated in comparison with wildtype
litter mates 60 min after food consumption.

Like other EECs, K and L cells are polarized and Another important aspect of the polarization of
exhibit an open-type morphology with an apical EECs is that their apical and basolateral surfaces
pole consisting of microvilli in direct contact with differ in their accessibility to luminal and vascu-
the lumen and a broad basolateral side from lar factors; whether sensory receptors are located
which dense core secretory vesicles exocytose on the apical or basolateral membrane can be
[Kieffer and Habener, 1999]. A unique feature of functionally critical, as for example the exclusive
these EECs was revealed using laser scanning expression of the sodium coupled glucose trans-
confocal microscopy of CCK-GFP and PYY- porter 1 (SGLT-1) on the apical membrane (see
GFP cells. Pseudopod-like processes were below), easily explains why incretin secreting
observed at the base of the cells, extending cells should be blind to elevation of vascular
towards adjacent cells and forming synapse-like glucose concentrations. However, basolateral
structures, thereby presumably exerting a parac- localization of receptors might be important to
rine effect on neighbouring enterocytes [Chandra shield them from saturating ligand concentra-
etal. 2010; Bohrquez etal. 2011]. However, the tions in the intestinal lumen, a scenario likely for
specific function of these pseudopod structures is the bile acid sensing receptor, G-protein cou-
yet to be established. pled bile acid receptor 1 (GPBAR1) and the

http://tae.sagepub.com 25
Therapeutic Advances in Endocrinology and Metabolism 7(1)

short chain fatty acid sensing receptors FFAR2/3 Depletion of sodium chloride in the lumen
(see below). impaired glucose-mediated GLP-1 secretion in
isolated perfused rat small intestine [Kuhre etal.
2015], suggesting that secretion requires
Enteroendocrine cell sensing Na-coupled uptake by the L cells. Furthermore,
For decades it has been known that the presence support for the involvement of SGLT1-mediated
in the lumen of food or its macronutrient compo- glucose influx come from studies where co-
nents (carbohydrates, fats and proteins) regulates administration of the SGLT inhibitor, phloridzin,
incretin hormone secretion. Elevation of circulat- with glucose in the upper small intestine blocked
ing GLP-1 levels can be detected within 1015 glucose-induced GLP-1 and GIP secretion in
minutes of eating and persists for several hours, mice [Moriya etal. 2009] and rats [Kuhre etal.
depending on the nutritional composition of the 2015]. Phloridzin also blocked hormone secre-
meal. The molecular mechanisms behind this tion triggered by nonmetabolizable substrates of
nutrient-dependent secretion have become clear SGLT: -methyl-d-glucopyranoside (-MDG)
from studies conducted by our group and others, and 3-O-methyl-d-glucose (3-OMG) [Moriya
aided by the engineering of transgenic mice etal. 2009].
wherein the EECs can be identified, isolated and
studied. Several studies have shown that EECs Sglt1 deficient mice displayed impaired GIP and
directly sense nutrients in the lumen, and this has GLP-1 release early after oral glucose administra-
been attributed to the activity of nutrient trans- tion [Gorboulev etal. 2012], but surprisingly had
porters, receptors and metabolism. In addition, higher GLP-1 levels at later time points following
hormone secretion has been linked to non-nutri- glucose gavage [Powell etal. 2013a]. One hypoth-
ent stimulation by factors such as cytokines, bile esis to explain these divergent effects on early and
acids and even gut microbial metabolites. late GLP-1 secretion would be that impaired glu-
cose absorption in the small intestine causes
This review focuses on the nutrient and non- increased glucose delivery and microbial fermen-
nutrient sensing mechanisms employed by GLP-1 tation in the distal intestine to produce short
and GIP-secreting L and K cells, respectively. chain fatty acids which in turn stimulate secretion
While most of the mechanisms described here from L cells via alternative pathways [Powell etal.
hold true for both cell types, some differences 2013a, 2013b]. This exemplifies an important
exist [Parker etal. 2009]. difference between K and L cells as K cells are
mostly found in the duodenum, no late glucose
stimulated GIP secretion was seen in Sglt1 knock-
Nutrient regulation out mice [Powell etal. 2013b] whereas, consist-
ent with the increased L-cell number in the distal
Carbohydrate sensing intestine, interventions that shift nutrients down
Glucose is a well-recognized stimulus of GLP-1 the gut tend to increase GLP-1 secretion.
secretion and has consistently been demonstrated
to cause GLP-1 release in in vitro enteroendo- Whether cellular metabolism contributes to glu-
crine cell lines, primary murine intestinal culture cose-induced incretin secretion is debatable. In
systems, and in vivo mouse models and humans. GLUTag cells, a murine enteroendocrine cell line
The concentrations of glucose in the lumen that expresses the proglucagon gene and secretes
required to trigger GLP-1 release in vivo are not GLP-1, glucose metabolism raises intracellular
clear and concentrations ranging from 5 to 1000 adenosine triphosphate (ATP) levels, causing clo-
mM have been reported by various groups to trig- sure of ATP-sensitive potassium (KATP) channels,
ger release from perfused ileum [Shima et al. membrane depolarization, intracellular Ca2+ ele-
1990; Sugiyama et al. 1994; Ritzel et al. 1997]. vation and GLP-1 secretion [Parker etal. 2012].
The pathway underlying glucose-dependent L and K cells express glucokinase and KATP chan-
secretion has been well characterized and requires nel subunits [Reimann et al. 2008; Parker et al.
the cotransport of sodium ions by the electrogenic 2009] and, while this machinery was shown to be
sodium/glucose cotransporter 1 (SGLT1). The required for glucose-stimulated GLP-1 secretion
influx of Na+ ions depolarizes the plasma mem- in GLUTag cells, in humans treated with the
brane, opening voltage sensitive calcium channels KATP channel inhibitors glibenclamide and repa-
and exocytosis of GLP-1-containing secretory glinide, GLP-1 secretion after an oral glucose tol-
vesicles [Gribble etal. 2003]. erance test was not affected [Stephens et al.

26 http://tae.sagepub.com
R Pais, FM Gribble et al.

2011]. A recent study investigated the role of Sweet taste receptors have also been proposed as
KATP channels in GIP secretion and concluded glucose sensors in incretin secreting cells. On the
that, in healthy mice, the channels are mostly one hand, expression of members of the T1R
closed and are therefore unlikely to play a major sweet taste receptors and the -subunit of the
role in glucose-stimulated hormone secretion G-protein gustducin have been reported in the
[Ogata et al. 2014]. Other sugars like fructose, small intestine of mice and the enteroendocrine
which is not a substrate for SGLT1, trigger cell line, STC-1 [Dyer etal. 2005] and colocaliza-
GLP-1 release in GLUTag cells by closure of tion of -gustducin with PYY and GLP-1 was
KATP channels in response to increased metabo- demonstrated in L cells of the human colon
lism [Gribble etal. 2003]. Fructose also induced [Rozengurt etal. 2006] and a subset of duodenal
GLP-1, CCK, PYY and neutrotensin (NTS) L and K cells [Young et al. 2013]. -Gustducin
secretion but not GIP secretion in healthy young null mice receiving glucose gavage showed defi-
humans and GLP-1 but not GIP secretion in cient release of GLP-1 [Jang etal. 2007] in com-
mice and rats [Kuhre et al. 2014]. The authors parison with wildtype mice. Similar disruption of
speculated that K cells might be less excitable glucose-stimulated GLP-1 secretion was observed
than L cells and so while, the weak fructose stim- in T1r3 null mice [Kokrashvili etal. 2009] and a
ulus might activate L cells, it is not sufficient to sweet taste receptor antagonist interfered with
activate K cells. However, reduced excitability of glucose-stimulated GLP-1 and PYY elevation in
K cells has not been experimentally demonstrated human volunteers [Steinert et al. 2011]. On the
and a recent report demonstrated fructose- other hand, ingestion of artificial sweeteners did
stimulated GIP secretion in mice with streptozo- not trigger GLP-1 in mice [Fujita etal. 2009] or
tocin-induced diabetes which seemed, however, humans [Ma etal. 2009], and their intraluminal
also not dependent on KATP-channel closure administration in the upper small intestine of
[Seino etal. 2015]. mice did not trigger hormone release [Moriya
etal. 2009].
Glucose transporter 2 (GLUT2), a facilitative
glucose transporter expressed in K and L cells, The finding that SGLT-1 inhibition or knockout
has been proposed to play a role in incretin release completely abolished GIP secretion in vivo
upstream of metabolism. GLUT2 is thought to [Powell et al. 2013b] argues against an apical
translocate to the apical membrane in response to location of a glucose sensing (taste) receptor in
elevated luminal glucose [Kellett and Helliwell, K cells, as the resulting elevated (unabsorbed)
2000], although the importance of this phenom- luminal sugar should then result in increased
enon is controversial [Gorboulev et al. 2012]. secretion. It could be argued that SGLT-1 might
Phloretin, an inhibitor of facilitative glucose but transport glucose across the epithelium where it is
not sodium coupled transport, abolished glucose then sensed by basolaterally located sweet taste
uptake into L cells in primary murine intestinal receptor, a mechanism described later for bile
cultures, although in contrast to phloridizin it had acid detection by TGR5 (see below). However,
little effect on glucose stimulated secretion this seems unlikely as incretin secretion from
[Parker et al. 2012]. Nonetheless, phloretin mixed primary epithelial cultures, which give
impaired GLP-1 secretion in the GLUTag cell unrestricted access to both sides of an enteroen-
line model [Parker etal. 2012] and perfused rat docrine cell, was also abolished by SGLT-1
small intestine [Kuhre etal. 2015], and reduced knockout/inhibition and was not responsive to
GIP and GLP-1 secretion in isolated rat small saturating concentrations of artificial sweeteners
intestine loops [Mace et al. 2012]. GLUT2 [Parker et al. 2012; Reimann et al. 2008]. It
knockout mice have been reported to have remains, however, possible that sweet taste recep-
impaired GLP-1, but not GIP responses, possibly tor expression is especially sensitive to culture
due to reduced GLP-1 content in the intestine conditions, although a recent study using a per-
[Cani et al. 2007a]. However, as a more recent fused intestinal preparation failed to observe
study found no significant differences in GIP and effects of artificial sweeteners [Kuhre etal. 2014].
GLP-1 secretion in GLUT2 knockout mice after An alternative explanation for the impaired incre-
an oral glucose gavage [Rder et al. 2014], it tin secretion observed in mice with altered taste
seems that GLUT2 and facilitative glucose trans- receptor pathway activity could be that the well
port in general is of less importance in K and L documented taste receptor dependent increase in
cells compared with its well established role in the SGLT-1 expression and glucose absorption
stimulus secretion coupling of pancreatic cells. [Zietek and Daniel,2015] should be impaired in

http://tae.sagepub.com 27
Therapeutic Advances in Endocrinology and Metabolism 7(1)

these mice, possibly reducing SGLT-1 expression peptones revealed the involvement of the calcium
in enteroendocrine cells. More work is needed to sensing receptor (CASR) and voltage gated cal-
substantiate a role of sweet taste receptors in cium channels (VGCC) in mediating peptone-
incretin hormone secretion. stimulated GLP-1 release [Pais et al. 2015].
CASRs primary role as a calcium sensor regulat-
ing parathyroid hormone release [Brown and
Protein sensing Herbert, 1995] is well recognized and established,
Dietary proteins are known to promote satiety but increasing evidence supports the idea that
and weight loss, and the satiating effects of pro- CASR also acts as an amino acid sensor in the
teins were found to be associated with the release gut, mediating macronutrient-dependent secre-
of the anorectic hormone, PYY, and suppression tion of gut hormones such as gastrin [Feng etal.
of the orexigenic hormone, ghrelin. In age- 2010], CCK [Liou etal. 2011; Wang etal. 2011],
matched normal weight and obese volunteers, a GIP and GLP-1 [Mace etal. 2012; Diakogiannaki
high protein diet caused a greater reduction in etal. 2013; Pais etal. 2015]. Meat hydrolysate has
hunger and a larger increment in PYY compared been linked to the activation of mitogen-activated
with isocaloric high-carbohydrate or high-fat protein kinases (MAPK) and stimulation of
meals [Batterham et al. 2006]. Similar effects GLP-1 secretion from murine-derived STC-1
were seen in rodents, in which increasing the die- and human-derived NCI-H716 cell lines [Reimer,
tary protein content reduced ad libitum calorie 2006]. In GLUTag cells, albumin egg hydro-
intake and increased circulating PYY levels, and lysate also increased transcription of the pro-
long-term high-protein feeding resulted in signifi- glucagon gene [Cordier-Bussat etal. 1998]. The
cantly less weight gain, less white adipose tissue, extent to which these effects are downstream of
and lower plasma leptin levels compared with the CASR activation remains to be established.
corresponding isocaloric normal-protein diets.
Additionally, Pyy null mice were found to be Among amino acids, l- glutamine (l-Gln) was
resistant to the satiating and weight-reducing consistently shown to trigger GLP-1 secretory
effects of high protein diets [Batterham et al. responses in human volunteers and patients with
2006]. Oral whey protein in combination with type 2 diabetes [Greenfield etal. 2009; Samocha-
glucose augmented GLP-1 responses in mice but Bonet etal. 2011] and also in in vitro studies per-
had no effect on GIP secretion [Gunnarsson etal. formed in primary mouse cultures and GLUTag
2006]. Interestingly, the same study showed that cells. The effect of l-Gln has been attributed to
whereas total GIP secretion was unaffected, the its ability both to trigger membrane depolariza-
active form of GIP, measured with N-terminally tion via electrogenic Na+ dependent amino acid
directed antibodies, was increased by whey pro- uptake leading to opening of L- and Q-type volt-
tein. The authors suggested that ingested protein age gated calcium channels, and to elevate cyto-
resulted in slower GIP inactivation, leading to an plasmic cyclic adenosine monophosphate (cAMP)
increase in the active form, but no change in cir- concentrations [Reimann et al. 2004; Tolhurst
culating total GIP levels. Several other studies etal. 2011]. In primary small intestinal cultures,
reported differential effects of oral protein inges- glutamine-triggered GLP-1 secretion was inhib-
tion on the two incretins, with greater effects ited by antagonists of CASR [Pais et al. 2015].
observed on GLP-1 than GIP secretion [Elliott Other amino acids, such as glycine and alanine,
etal. 1993]. are strong stimulants of GLP-1 secretion from
GLUTag cells through activation of the iono-
In the perfused rat intestine, GLP-1 release was tropic glycine receptor [Gameiro etal. 2005], but
triggered by luminal perfusion of a meat protein this mechanism was not observed in colonic L
hydrolysate known as peptone, and the response cells in primary culture. l-Phenylalanine (l-Phe),
magnitude was similar in the upper and lower half however, was a highly effective stimulus of GLP-1
of the small intestine [Svendsen etal. 2015]. Meat release in mouse small intestinal cultures [Pais
peptones strongly stimulated GLP-1 secretion etal. 2015], but was not an exceptional stimulus
from primary murine colonic cultures in colonic cultures, and was ineffective in
[Diakogiannaki etal. 2013] and we recently dem- GLUTag cells [Reimann et al. 2004; Tolhurst
onstrated GLP-1 release from primary murine et al. 2011]. The identity of the L-cell sensor
duodenal cultures after treatment with meat, veg- underlying l-Phe triggered secretion remains to
etable and casein-derived peptones [Pais et al. be established, and although some studies have
2015]. Mechanistic studies performed with meat proposed the involvement of CASR [Mace etal.

28 http://tae.sagepub.com
R Pais, FM Gribble et al.

2012], we were unable to block l-Phe triggered Several GPCRs have been implicated in the sens-
GLP-1 secretion from primary intestinal cultures ing of fatty acids and the concomitant release of
using CASR antagonists [Pais etal. 2015]. Jiang gut hormones by EECs. Those identified until
and colleagues reported higher meal-dependent now include GPR40 (FFAR1), GPR120
GLP-1 levels in mice lacking the neutral amino (FFAR4) and GPR119. GPR40 is activated by
acid transporter B0AT1 than in controls and sug- medium to long chain free fatty acids (FFAs)
gested that reduced neutral amino acid absorp- [Briscoe etal. 2003] and found to be present in
tion in the null mice causes an overload of amino GIP and GLP-1 secreting cells in mice. Following
acids in the lower gut and stimulation of the large an oral high-fat diet (HFD), Gpr40 deficient mice
reservoir of distally located GLP-1 secreting cells exhibited diminished secretion of GLP-1 and
[Jiang etal. 2015]. These results mirror the eleva- GIP with concomitant reduction in insulin levels
tion of glucose-triggered GLP-1 release in mice [Edfalk etal. 2008]. GPR120 is also activated by
lacking SGLT-1, which are similarly postulated unsaturated long chain FFAs and was found co-
to reflect increased glucose delivery to the distal localized with GLP-1 in human colon. In HEK
gut, as discussed above. cells stably expressing GPR120, long chain FFAs
evoked an increase in intracellular calcium [Ca2+]i
Several other candidate sensory pathways for pro- in a dose-dependent fashion and, in STC-1 cells,
tein digestion products have been described. -linoleic acid strongly stimulated GLP-1 release
Activation of the G-protein coupled receptor [Hirasawa etal. 2005]. The increase in [Ca2+]i in
GPRC6A in GLUTag cells is triggered by ornith- STC-1 cells induced by -linolenic acid was elim-
ine [Oya et al. 2013]. The proton-coupled elec- inated by siRNA specific for mouse Gpr120 con-
trogenic peptide transporter PEPT1 [Daniel, firming the role of this receptor in long chain
2004] was shown to be critical for dipeptide trig- FFA-mediated GLP-1 release [Hirasawa et al.
gered GLP-1 secretion in studies using agonists 2005].
and antagonists of the transporter and pept1 defi-
cient mice [Diakogiannaki et al. 2013]. Another A recent report emphasized the greater impor-
receptor activated by polypeptides, lysophospha- tance of GPR120 for GIP compared with GLP-1
tidic acid receptor 5 (LPAR5, GPR92/93) was secretion [Iwasaki etal. 2015] and the same group
shown to mediate CCK [Choi etal. 2007] but not reported specific enrichment of the fatty acid
GLP-1 secretion [Diakogiannaki et al. 2013]. binding protein FABP5 in K cells, suggesting a
Further, the umami taste receptor dimer Tas1R1/ role in fatty acid detection [Shibue et al. 2015].
Tas1R3 was shown to be involved in amino acid Ligands of GPR119 include the fatty acid deriva-
triggered CCK secretion [Daly etal. 2013]. tives oleoylethanolamide (OEA) [Overton et al.
2006] and lysophophatidylcholine (LPC) [Soga
et al. 2005], as well as monoacylglycerides
Fat sensing [Hansen etal. 2011; Mande etal. 2015; Lauffer
It was observed in the 1980s that infusion of lipid etal. 2009]. OEA administration in rats reduced
emulsion into the ileum of humans reduced food food intake and weight gain [Rodrguez De
intake and promoted satiety, and it was later Fonseca etal. 2001] and stimulated GLP-1 secre-
found that these effects were accompanied by an tion from perfused rat ileum [Lauffer etal. 2009],
increase in plasma CCK levels [Welch etal. 1985; GLUTag cells [Lauffer etal. 2009] and primary
Drewe et al. 1992]. Indeed, fats also potently cultured murine L cells [Moss et al. 2015].
stimulate GLP-1 and PYY release [Feltrin etal. GPR119 is Gs-coupled and increases intracellu-
2004; Little et al. 2005]. The main products of lar cAMP levels upon activation. GLUTag cells
lipid digestion are monoglycerides (MGs) and transfected with specific Gpr119 siRNA failed to
fatty acids, and inhibition of fat digestion by lipase increase cAMP levels in response to OEA [Lauffer
inhibitors attenuated the rise in plasma levels of et al. 2009]. Gpr119 knockout mice showed
CCK, PYY and GLP-1 after a fat-meal [Feinle diminished nutrient-stimulated GLP-1 secretion
etal. 2003; Feinle-Bisset etal. 2005; Ellrichmann [Lan etal. 2009]. Also, oral administration of the
etal. 2008; Beglinger etal. 2010]. CCK, GLP-1 GPR119 agonist AR231453 increased plasma
and PYY release were found to be dependent on concentrations of GLP-1 and GIP in wildtype but
fatty acid chain length with only fatty acids greater not Gpr119 knockout animals [Chu etal. 2008].
than C10 being effective in stimulating hormone 2-Oleoyl glycerol, a product of triglyceride diges-
secretion [Feltrin etal. 2004; Feinle-Bisset etal. tion, also acts on GPR119 and its administration
2005; Little etal. 2005]. to humans significantly increased plasma GLP-1

http://tae.sagepub.com 29
Therapeutic Advances in Endocrinology and Metabolism 7(1)

and GIP levels [Hansen etal. 2011; Mande etal. Whereas fats acutely stimulate GLP-1 secretion
2015]. Pharmacological GPR119 agonists devel- as evidenced from above, chronic high-fat feeding
oped for human studies and tested in patients in mice reduced numbers of GLP-1 and GIP pos-
with type 2 diabetes have not, however, shown itive cells and significantly decreased the expres-
significant metabolic benefits [Katz etal. 2012]. sion of enteroendocrine hormones, nutrient
The reason behind this is uncertain and the thera- sensing machinery and enteroendocrine-specific
peutic potential of GPR119 is still under transcription factors [Richards et al. 2015].
investigation. Hayashi and colleagues demonstrated that endo-
plasmic reticulum stress, induced with excessive
A range of non-GPCR machinery has also been doses of palmitate, increased the expression of
implicated in free-fatty-acid-triggered incretin endoplasmic reticulum stress markers, Chop and
secretion. Protein kinase C was implicated in BiP, reduced protein levels and function of pro-
oleic acid triggered GLP-1 secretion in GLUTag hormone convertase 1/3, and decreased GLP-1
cells [Iakoubov et al. 2007] and rodents secretion from GLUTag cells [Hayashi et al.
[Iakoubov etal. 2011], and fatty acid transport 2014]. Aranias and colleagues, however, observed
protein was found to contribute to oleic acid an increase in L-cell density in the jejunum of
induced GLP-1 secretion in vitro and in vivo morbidly obese subjects and elevated GLP-1
[Poreba et al. 2012]. Triglycerides (TGs) are secretion in HFD fed mice after an oral glucose
resynthesized in the enterocytes of small intes- challenge. They hypothesized that increased
tine from absorbed fatty acids and MGs by the GLP-1 secretion would favour insulin secretion
sequential action of monoacylglycerol acyltrans- and counter balance diet-induced insulin resist-
ferase (MGAT) and diacylglycerol acyltransferase ance [Aranias etal. 2015]. The influence of diet
(DGAT). The newly formed TGs are then on L cells and gut hormone secretion is clearly
incorporated into chylomicrons (CM) by micro- not completely understood, and more work needs
somal triglyceride transfer protein (MTP). to be done in this field.
MGAT2 and DGAT1 deficient mice displayed
impaired GIP secretion after oral triglyceride
gavage, but increased GLP-1 and PYY levels Non nutrient regulation
which remained elevated even 2 hours after gav-
age in comparison with wildtype controls [Okawa Gut microbiota
et al. 2009]. In addition, administration of a Gut bacteria have a profound impact on the host
pharmacological inhibitor of DGAT1 prior to oil and influence, in many ways, host physiology and
gavage reduced postprandial TG levels and led metabolism [Tremaroli and Bckhed, 2012].
to prolonged elevated levels of GLP-1 in mice EECs lie in close proximity to microbes and/or
[Ables etal. 2012] Furthermore, an inhibitor of their metabolic products in the gut and the result-
intestinal specific MTP was shown to elevate ing crosstalk regulates hormone secretion.
GLP-1 and PYY in rats fed a high fat diet [Hata Bacterial fermentation of undigested prebiotics
et al. 2011]. CM formation and secretion were and complex carbohydrates produces short chain
shown to be critical for GLP-1 [Lu etal. 2012] fatty acids (SCFAs) such as acetate, butyrate and
and GIP [Shimotoyodome et al. 2009] release, propionate. SCFAs induced GLP-1 secretion
as revealed in rodent studies using surfactants from primary colonic cultures and this was shown
that inhibited CM formation. Recently, Wang to be mediated by GPR43 (FFAR2) and GPR41
and colleagues showed that Apolipoprotein A-IV (FFAR3). GPR43 is Gi- and Gq-coupled and
(ApoA-IV) knockout mice have increased lym- triggered an increase in [Ca2+]i in colonic L cells
phatic and plasma GLP-1 levels 30 minutes after following activation by SCFAs. Mice deficient in
intraduodenal administration of Ensure, a com- Ffar2 and Ffar3 displayed reduced SCFA-
mercially available liquid meal, in comparison triggered GLP-1 secretion [Tolhurst etal. 2012].
with wildtype controls. Interestingly, in another Additionally, butyrate, acetate and propionate
experiment conducted by the same group, acute administration protected mice from developing
administration of ApoA-IV prior to intraduode- diet-induced obesity and insulin resistance and
nal Ensure infusion did not alter GLP-1 levels in stimulated gut hormone release [Lin etal. 2012].
wildtype mice or the ApoA-IV-/- mice. They A recent report, however, observed improved glu-
attributed the increased GLP-1 levels in the cose tolerance in Ffar2/3 double knockout mice,
ApoA-IV knockout mice to increased expression which was explained by the direct inhibitory effect
of Gpr119 in the ileum [Wang etal. 2015]. of these receptors in pancreatic cells, and found

30 http://tae.sagepub.com
R Pais, FM Gribble et al.

no significant effect of Villin-Cre mediated condi- Bile acids


tional knockout of these receptors in the intestine Bile acids play an important role in the digestion
[Tang etal. 2015]. In contrast, impaired glucose and absorption of dietary fats by facilitating the
tolerance of Ffar3-/- mice was recapitulated, but a formation of micelles. Primary as well as second-
selective FFAR3 agonist also impaired glucose ary bile acids (formed as a result of bacterial mod-
tolerance, presumably by direct action in the islet ification) act as signalling molecules and strongly
[Forbes et al. 2015], and it was concluded that stimulate GLP-1 and PYY secretion [Ullmer
the observed anorectic effect of this compound etal. 2013]. They are known to act at two specific
was mostly mediated through elevation of PYY. receptors: the nuclear farnesoid X receptor
(FXR); and the transmembrane G-protein cou-
It is now well established that obesity and type 2 pled receptor TGR5 (GPBAR1). Bile acid trig-
diabetes affect gut microbiome diversity and gered GLP-1 secretion was found to be TGR5
composition [Ley et al. 2005; Qin et al. 2012]. dependent in STC-1 cells [Katsuma etal. 2005],
Prebiotic feeding in ob/ob mice increased L-cell GLUTag cells and primary murine intestinal cul-
number, proglucagon mRNA levels and portal tures [Parker et al. 2012] and in vivo [Thomas
plasma GLP-1 levels, and this was mediated by etal. 2009]. Downstream signalling involved ele-
the increased production of acetate and propion- vation of cAMP and [Ca2+]i [Parker etal. 2012].
ate [Everard etal. 2011]. Characterization of the Deoxycholic acid infusion in the colon increased
microbiome revealed that the abundance of enteroglucagon (GLP-1) and PYY secretion in
Akkermansia muciniphila was reduced in obese humans undergoing colonoscopy [Adrian et al.
and diabetic mice [Everard etal. 2013] and feed- 1993] and rectal taurocholate infusion reduced
ing of this bacteria was positively associated with food intake, lowered glucose and increased GLP-
increased L-cell number and secretion [Everard 1, PYY and insulin in obese type-2 diabetic vol-
etal. 2011]. Several other studies showed benefi- unteers [Adrian etal. 2012].
cial effects of prebiotics treatment on GLP-1,
GLP-2 and PYY secretion in rodents and humans However, there has been debate about whether
[Cani etal. 2007b, 2009a, 2009b]. bile acids target their receptors on L cells from the
apical or basolateral compartment. Brighton and
Although a large body of evidence suggests that colleagues measured GLP-1 release from murine
SCFAs produced by gut bacterial fermentation ileum mounted in small volume Ussing chambers;
stimulate GLP-1 secretion, Wichmann and col- they showed that basolateral taurodeoxycholate
leagues demonstrated that in germ-free (GF) (TDCA) or a TGR5 agonist stimulated GLP-1
mice, having severely reduced SCFAs, basal secretion but that the effects of apically-applied
plasma GLP-1 levels were 3-fold higher than con- TDCA were abolished by inhibition of bile acid
ventional raised mice (CONV-R). They attrib- uptake across the epithelium. Preferential stimula-
uted this observation to increased proglucagon tion of GLP-1 release by basolateral versus apical
(Gcg) expression in the caecum and colon. In bile acids was confirmed in the perfused rat gut,
addition, GF mice had more GLP-1 positive cells leading to the conclusion that bile acid triggered
in the caecum and colon than the CONV-R mice. GLP-1 secretion is predominantly mediated
Colonization of GF mice with SCFA-producing by basolaterally located TGR5 on the L cell
bacteria increased acetate and propionate and [Brighton etal. 2015]. These findings dampen the
correspondingly decreased Gcg expression and hope of developing TGR5 agonists as GLP-1
GLP-1 positive cells. Conversely, depletion of secretagogues, as TGR5 ligands entering the sys-
microbiota in CONV-R with antibiotics treat- temic circulation have been found to trigger gall
ment over 3 days decreased SCFA levels and bladder overfilling [Li etal. 2011]. It is interesting
increased Gcg expression [Wichmann etal. 2013]. to note that a recent report suggests that FFAR1
Other bacterial metabolites known to affect (GPR40) is also located on the basolateral rather
GLP-1 secretion include indole. Indole is pro- than the apical membrane of enteroendocrine
duced from tryptophan by gut bacteria and, while cells [Christensen etal. 2015] and one might spec-
acutely stimulated GLP-1 secretion by inhibiting ulate that this will also extend to the related short-
voltage gated K+ channels, after prolonged expo- chain fatty acid receptors FFAR2 (GPR43) and
sure it slowed ATP production by blocking FFAR3 (GPR41), as physiological observed lumi-
NADH dehydrogenase leading to a delayed nal concentrations of short chain fatty acids
reduction of GLP-1 secretion [Chimerel et al. are well above their reported potencies for these
2014]. receptors.

http://tae.sagepub.com 31
Therapeutic Advances in Endocrinology and Metabolism 7(1)

A number of studies have shown that some of the A number of studies have identified a stimulatory
beneficial effects of gastric bypass surgery on glu- effect of GIP on GLP-1 secretion in rodents.
cose metabolism in diabetic subjects can be attrib- Intravenous infusion of GIP, for example, elevated
uted to increased bile acid delivery to the distal gut plasma levels of proglucagon-derived peptide
and enhanced release of gut hormones such as (GLP-1) in rats [Roberge et al. 1993] and GIP
GLP-1 and PYY [Patti et al. 2009]. Bile acid treatment triggered GLP-1 release from primary
sequestrants (BAS) are anion exchange resins that canine L cells [Damholt et al. 1998]. It is not,
trap bile acids in the lumen and have cholesterol however, currently believed that this plays an
lowering and antidiabetic properties [Prawitt etal. important role in humans, as even pharmacologi-
2014]. Additionally, they were shown to enhance cally raised GIP levels had little effect on plasma
GLP-1 secretion in diet-induced obese mice and GLP-1 [Nauck etal. 1993]. Ghrelin, an orexigenic
increased proglucagon gene expression down- hormone released from the stomach, enhanced
stream of TGR5 [Harach etal. 2012]. Recently, glucose-induced GLP-1 release in vivo in mice
Fxr was shown to be expressed in mouse ileal and and in vitro in rodent and human L cells through
colonic L cells and colocalized with GLP-1 in the an extracellular-signal-regulated kinase ERK
human jejunum. Surprisingly, treatment with the 1/2-dependent pathway [Gagnon etal. 2015a].
FXR agonist GW406 decreased gcg expression
and FXR activation was demonstrated to inhibit Obesity is a pro-inflammatory condition associ-
glycolysis, leading to reduced intracellular ATP ated with elevated levels of circulating pro-
levels and impaired glucose-dependent GLP-1 inflammatory cytokines which are released from
secretion [Trabelsi etal. 2015]. the adipose tissue and infiltrating immune cells.
The endocrine and secretory behaviour of adipose
tissue was revealed with the discovery that adipo-
Hormones/cytokines cytes synthesize and secrete the cytokine tumour
An emerging body of evidence shows that there is necrosis factor (TNF) [Hotamisligil et al.
a crosstalk between the enteroendocrine system 1993] and the hormone leptin [Zhang etal. 1994].
and other organs in the body and that gut hor- Since then, several adipose-derived proteins have
mone secretion is influenced by factors such as been discovered that have profound physiological
hormones and cytokines which act on the EECs effects; they have collectively been given the term
to influence secretion either positively or nega- adipokines. Leptin has well established effects on
tively. Somatostatin (SST) is produced from D food intake and elevated circulating levels in obe-
cells in the oxyntic and pyloric regions of the sity are proportional to fat mass [Considine etal.
stomach and its secretion is stimulated by GIP, 1996]. The long form of the leptin receptor,
GLP-1 and CCK but not gastrin [Adriaenssens Ob-Rb, was found on human and rodent L cells
etal. 2015]. SST in turn exerts a suppressive tone and leptin was observed to stimulate GLP-1 secre-
over neighbouring EECs in the stomach as well as tion in vitro and in vivo in rats and mice [Anini and
in the gut. Acting at least in part via the Gi- Brubaker, 2003]. Among other adipokines, inter-
coupled SSTR5 receptor, SST inhibited GIP and leukin (IL) 6 is regarded as a key player contribut-
GLP-1 secretion in primary murine intestinal cul- ing to obesity linked insulin resistance [Senn etal.
tures and acts by suppressing cAMP levels in 2002; Eder etal. 2009]. IL-6 is also secreted from
EECs [Moss etal. 2012]. exercising skeletal muscle and was shown to stim-
ulate GLP-1 secretion not only from intestinal L
Intestinal endocannabinoids exert orexigenic cells but also pancreatic cells [Ellingsgaard etal.
effects by signalling through vagal afferent neu- 2011]. Exercise has been shown to be associated
rons to satiety centres in the central nervous sys- with elevated levels of GLP-1, PYY and pancre-
tem [Izzo and Sharkey, 2010]. Levels of the atic polypeptide [Martins etal. 2007] and IL-6 is
endocannabinoid anandamide have been reported a candidate mediator of this effect.
to increase in the small intestine of rats upon food
deprivation and levels returned to baseline upon Recently, TNF was demonstrated to affect
refeeding [Gmez etal. 2002]. The cannabinoid GLP-1 secretion. Whereas acute application of
receptor CB1 was found to be highly expressed in TNF increased GLP-1 secretion from a human
GIP secreting K cells and, consistent with this GLP-1 secreting cell line, TNF pretreatment
finding, GIP but not GLP-1 secretion was of both human and murine cell lines reduced
reduced by the CB1 synthetic agonist methanan- gcg expression and impaired GIP-triggered
damide (mAEA) [Moss etal. 2012]. GLP-1 secretion. Co-administration of a nuclear

32 http://tae.sagepub.com
R Pais, FM Gribble et al.

factor-B (NF) inhibitor abolished the inhibi- GLP-1 concentrations [Theodorakis et al. 2006;
tory effect of TNF pretreatment on GLP-1 Habib etal. 2012] and that they directly respond
responses and treatment with the TNF inhibitor to nutrient stimulation [Pais etal. 2015]. L cells lie
etanercept improved GLP-1 secretion in HFD- in close proximity to neurons and microvascula-
induced obese mice [Gagnon et al. 2015b]. ture in the gut and studies performed on isolated
Another adipokines, RANTES (CCL5) which is vascularly perfused rat colon and ileum demon-
upregulated in obesity [Wu et al. 2007; strated stimulatory roles for neurotransmitters,
Keophiphath et al. 2010] was shown to reduce calcitonin-gene related peptide (CGRP) and gas-
glucose-stimulated GLP-1 release in vitro in NCI- trin releasing peptide (GRP) on GLP-1 secretion
H716 cells and plasma levels of GLP-1 and GLP-2 [Plaisancie et al. 1994; Dumoulin et al. 1995].
after an oral glucose load in mice [Pais etal. 2014]. GRP is released from GRPergic neurons and its
role in GLP-1 secretion was confirmed in studies
Progesterone increased plasma levels of GIP and using GRP agonists [Roberge and Brubaker,
GLP-1 in mice, and was shown to be mediated by 1996] and GRP deficient mice [Persson et al.
the G-protein coupled receptors PAQR5 and 2000]. Cholinergic regulation of GLP-1 secretion
PAQR7 by receptor knockdown studies conducted was shown in the murine STC-1 cell line, medi-
in GLUTag cells [Flock etal. 2013]. Insulin recep- ated by the muscarinic receptor M3 [Abello etal.
tor expression was also found on murine and 1994]. Other studies have shown roles for M1 and
human L cells, and insulin was found to stimulate M2 in stimulating GLP-1 release from L cells
GLP-1 secretion in vitro from murine L cells by in vitro, and M1 in mediating GLP-1 release from
activating the phosphatidylinositol 3 kinase-Akt rat in vivo [Anini etal. 2002]. Pituitary adenylate
and MAPK kinase (MEK)-ERK1/2 pathways. cyclase-activating polypeptide (PACAP) increased
Pretreatment with high concentrations of insulin GLP-1 secretion from GLUTag cells by increas-
for 24 hours caused insulin resistance and reduced ing intracellular cAMP levels [Simpson et al.
basal and insulin induced GLP-1 secretion [Lim 2007]. -Aminobutyric acid (GABA) receptors
et al. 2009]. Nesfatin-1, an anorexigenic peptide were detected on the STC-1 and GLUTag cell
found in the hypothalamus [Oh-I et al. 2006], lines: GABA triggered CCK and GLP-1 secretion
colocalized with GIP and GLP-1 positive cells in mediated by the GABAA receptor subtype in
mouse small intestine and stimulated GLP-1 STC-1 cells [Glassmeier etal. 1998] and by both
secretion from STC-1 cells [Ramesh etal. 2015]. GABAA and GABAC receptors in GLUTag cells
[Gameiro etal. 2005].

Neural regulation
The enteric nervous system is sometimes referred Cell number and sensitivity
to as a second brain because of its degree of An alternative or supplementary approach to the
autonomous function. It exerts several actions in acute stimulation of incretin secretion would be
the gastrointestinal tract such as control of gut the promotion of increased incretin expression in
motility, fluid exchange between the gut and the the intestine. FXR-mediated effects of bile acids
lumen, regulation of gastric and pancreatic secre- on pro-glucagon expression have already been
tion and gut defence, and it interacts with EECs mentioned. Alternatively, SCFA has been shown
in the gut epithelium to regulate hormone release. to increase the number of gcg-expressing cells in
Previous immunohistochemistry studies per- organotypic intestinal organoid cultures [Petersen
formed on rat and human tissue detected GLP-1- et al. 2014], which might underlie the reported
positive cells predominantly in the lower small increase in L-cell density in response to prebiotic
intestine and colon [Eissele etal. 1992] and led to fibre supplementation in ob/ob mice [Everard
the proposal that the initial rapid rise in GLP-1 etal. 2011] and the reduced gcg mRNA content
following a meal was indirectly mediated by neu- of Ffar2-/- derived intestines [Tolhurst et al.
ral/endocrine pathways arising from nutrient 2012]. More recently Petersen and colleagues
detection in the proximal gut, rather than by demonstrated that inhibition of NOTCH signal-
direct nutrient stimulation of L cells. ling pathways with the -secretase inhibitor diben-
zazepine (DBZ), increased L-cell numbers in in
More recent studies, however, have concluded vitro models of intestinal organoids cultures from
that enough GLP-1 cells are present in the proxi- mouse and human and augmented glucose-stim-
mal small intestine of a number of species to ulated GLP-1 secretion. DBZ also increased
account for the rapid postprandial rise of plasma L-cell numbers in the intestines of HFD fed mice

http://tae.sagepub.com 33
Therapeutic Advances in Endocrinology and Metabolism 7(1)

and type 2 diabetes mouse models and improved and gut function. The incretins, in particular,
glucose-stimulated insulin release and glucose have garnered a great deal of interest owing to
tolerance [Petersen etal. 2015]. their antidiabetic and satiating effects. GLP-1
based drugs (GLP-1 mimetics and inhibitors of
Another potentially interesting aspect of incretin GLP-1 degradation) and bariatric surgery have
secretion investigated only recently is its link to shown immense beneficial effects on glycaemia
circadian rhythm and the possibility that it plays a and weight loss in humans. Fuelled by our under-
role in entraining these rhythms to food intake. standing of the intricate pathways by which EEC
Gil-Lozano and colleagues demonstrated circa- cells sense nutrients and other factors in their
dian rhythmicity in the secretion of GLP-1 in milieu, strategies to harness the tremendous
response to different secretagogues in vitro and potential of the enteroendocrine system by target-
well as in vivo in rats fasted for 4 hours before ing endogenous gut hormone production offer
being administered a glucose load. They also great hope for the future treatment of diabetes
showed rhythmic expression of clock genes in and obesity.
GLUTag cells and demonstrated that knockdown
of two rhythmically expressed target genes (thyro- Funding
troph embryonic factor and protein tyrosine The author(s) disclosed receipt of the following
phosphatase 4a1) resulted in altered GLP-1 financial support for the research, authorship,
release [Gil-Lozano etal. 2014]. and/or publication of this article: Research in the
Reimann and Gribble laboratories is currently
funded by the Wellcome Trust (grants 106262/
Physiological relevance Z/14/Z and 106263/Z/14/Z), Full4Health (grants
Therapies targeting the incretin axis have proved FP7/2011-2015 no: 266408) and the Medical
highly effective in treating type 2 diabetes and Research Council (MRC) (grant MRC_ MC_
provide the additional benefits of weight loss and UU_12012/3).
cardioprotection [Drucker and Nauck, 2006;
Gejl et al. 2012]. Roux-en-Y gastric bypass Conflict of interest statement
(RYGB) is currently the most widely used bariat- The author(s) declared no potential conflicts of
ric procedure for treating severe obesity. interest with respect to the research, authorship,
Anastomosis of the jejunum to a small gastric and/or publication of this article.
pouch causes rapid postprandial delivery of nutri-
ents to the distal gut with its large population of L
cells. The consequent exaggerated release of
GLP-1 is believed to underlie some of the remark- References
able improvements in glucose tolerance found in Abello, J., Ye, F., Bosshard, A., Bernard, C., Cuber,
people undergoing surgery, in many cases result- J. and Chayvialle, J. (1994) Stimulation of glucagon-
ing in remission of type 2 diabetes [Korner etal. like peptide-1 secretion by muscarinic agonist in a
2007; Dirksen et al. 2010; Falkn et al. 2011; murine intestinal endocrine cell line. Endocrinology
Jimnez et al. 2012]. High accompanying PYY 134: 20112017.
levels after surgery likely contribute to the Ables, G., Yang, K., Vogel, S., Hernandez-Ono, A.,
observed reductions in appetite [Korner et al. Yu, S., Yuen, J. etal. (2012) Intestinal DGAT1
2005; Mornigo etal. 2008]. Interestingly, nutri- deficiency reduces postprandial triglyceride and retinyl
ent malabsorption in mice deficient in Sglt1 ester excursions by inhibiting chylomicron secretion and
[Powell etal. 2013a], B0at1 [Jiang etal. 2015] and delaying gastric emptying. J Lipid Res 53: 23642379.
Dgat1 [Okawa etal. 2009] seems to present a sce- Adriaenssens, A., Yee Hong Lam, B., Billing, L.,
nario that mimics some aspects of bariatric sur- Skeffington, K., Sewing, S., Reimann, F. etal.
gery. In each case elevated postprandial GLP-1 (2015) A transcriptome-led exploration of molecular
levels have been reported, probably reflecting the mechanisms regulating somatostatin-producing
increased delivery of nonabsorbed nutrients to D-cells in the gastric epithelium. Endocrinology:
the hindgut (Figure 1), although not reproducing en20151301.
the rapid peaks found after surgery. Adrian, T., Ballantyne, G., Longo, W., Bilchik, A.,
Graham, S., Basson, M. etal. (1993) Deoxycholate
Conclusion is an important releaser of peptide YY and
The enteroendocrine system secretes a myriad of enteroglucagon from the human colon. Gut 34:
hormones that are pivotal to glucose homeostasis 12191224.

34 http://tae.sagepub.com
R Pais, FM Gribble et al.

Adrian, T., Gariballa, S., Parekh, K., Thomas, promote L-cell differentiation in the proximal colon of
S., Saadi, H., Al Kaabi, J. etal. (2012) Rectal rats. Br J Nutr 98: 3237.
Taurocholate increases L cell and insulin secretion,
Cani, P., Lecourt, E., Dewulf, E., Sohet, F.,
and decreases blood glucose and food intake in obese
Pachikian, B., Naslain, D. etal. (2009a) Gut
type 2 diabetic volunteers. Diabetologia 55: 23432347.
microbiota fermentation of prebiotics increases
Anini, Y. and Brubaker, P. (2003) Role of leptin in satietogenic and incretin gut peptide production with
the regulation of glucagon-like peptide-1 secretion. consequences for appetite sensation and glucose
Diabetes 52: 252259. response after a meal. Am J Clin Nutr 90: 12361243.
Anini, Y., Hansotia, T. and Brubaker, P. (2002) Cani, P., Possemiers, S., Van De Wiele, T., Guiot,
Muscarinic receptors control postprandial release of Y., Everard, A., Rottier, O. etal. (2009b) Changes
glucagon-like peptide-1: in vivo and in vitro studies in in gut microbiota control inflammation in obese
rats. Endocrinology 143: 24202426. mice through a mechanism involving GLP-2-driven
improvement of gut permeability. Gut 58: 10911103.
Aranias, T., Grosfeld, A., Poitou, C., Omar, A., Le
Gall, M., Miquel, S. etal. (2015) Lipid-rich diet Chandra, R., Samsa, L., Vigna, S. and Liddle,
enhances L-Cell density in obese subjects and in mice R. (2010) Pseudopod-Like basal cell processes in
through improved L-cell differentiation. J Nutr Sci 4: intestinal cholecystokinin cells. Cell Tissue Res 341:
e22. 289297.
Batterham, R., Heffron, H., Kapoor, S., Chivers, J., Chimerel, C., Emery, E., Summers, D., Keyser,
Chandarana, K., Herzog, H. etal. (2006) Critical U., Gribble, F. and Reimann, F. (2014) Bacterial
role for peptide YY in protein-mediated satiation and metabolite indole modulates incretin secretion from
body-weight regulation. Cell Metab 4: 223233. intestinal enteroendocrine L cells. Cell Rep 9:
12021208.
Bayliss, W. and Starling, E. (1902) The mechanism of
pancreatic secretion. J Physiol 28: 325353. Choi, S., Lee, M., Shiu, A., Yo, S., Halldn, G. and
Aponte, G. (2007) GPR93 activation by protein
Beglinger, S., Drewe, J., Schirra, J., Gke, B.,
hydrolysate induces CCK transcription and secretion
Damato, M. and Beglinger, C. (2010) Role of fat
in STC-1 cells. Am J Physiol Gastrointest Liver Physiol
hydrolysis in regulating glucagon-like peptide-1
292: G13661375.
secretion. J Clin Endocrinol Metab 95: 879886.
Christensen, L., Kuhre, R., Janus, C., Svendsen,
Bohrquez, D., Chandra, R., Samsa, L., Vigna,
B. and Holst, J. (2015) Vascular, but not luminal,
S. and Liddle, R. (2011) Characterization of basal
activation of FFAR1 (GPR40) stimulates GLP-1
pseudopod-like processes in ileal and colonic PYY
Secretion from isolated perfused rat small intestine.
cells. J Mol Histol 42: 313.
Physiol Rep 3: 312551.
Brighton, C., Rievaj, J., Kuhre, R., Glass, L.,
Chu, Z., Carroll, C., Alfonso, J., Gutierrez, V., He,
Schoonjans, K., Holst, J. etal. (2015) Bile Acids
H., Lucman, A. etal. (2008) A role for intestinal
trigger GLP-1 release predominantly by accessing
endocrine cell-expressed G Protein-coupled receptor
basolaterally-located G-protein coupled bile acid
119 in glycemic control by enhancing glucagon-like
receptors. Endocrinology: en20151321.
peptide-1 and glucose-dependent insulinotropic
Briscoe, C., Tadayyon, M., Andrews, J., Benson, W., peptide release. Endocrinology 149: 20382047.
Chambers, J., Eilert, M. etal. (2003) The orphan
Considine, R., Sinha, M., Heiman, M., Kriauciunas,
G protein-coupled receptor GPR40 Is activated by
A., Stephens, T., Nyce, M. etal. (1996) Serum
medium and long chain fatty acids. J Biol Chem 278:
immunoreactive-leptin concentrations in normal-
1130311311.
weight and obese humans. N Engl J Med 334:
Brown, E. and Hebert, S. (1995) Hot Papers. Signal 292295.
transduction. Cloning and characterization of an
Cordier-Bussat, M., Bernard, C., Levenez, F., Klages,
extracellular Ca2+-sensing receptor from bovine
N., Laser-Ritz, B., Philippe, J. etal. (1998) Peptones
parathyroid, Nature (1995), 366: 575580 (by
stimulate both the secretion of the incretin hormone
Brown, E. etal. Comments. Scientist 9: 1515.
glucagon-like peptide 1 and the transcription of the
Cani, P., Holst, J., Drucker, D., Delzenne, N., proglucagon gene. Diabetes 47: 10381045.
Thorens, B., Burcelin, R. etal. (2007a) GLUT2 and
the incretin receptors are involved in glucose-induced Daly, K., Al-Rammahi, M., Moran, A., Marcello, M.,
incretin secretion. Mol Cell Endocrinol 276: 1823. Ninomiya, Y. and Shirazi-Beechey, S. (2013) Sensing
of Amino acids by the gut-expressed taste receptor
Cani, P., Hoste, S., Guiot, Y. and Delzenne, N. T1R1-T1R3 Stimulates cck secretion. Am J Physiol
(2007b) Dietary Non-digestible carbohydrates Gastrointest Liver Physiol 304: G271282.

http://tae.sagepub.com 35
Therapeutic Advances in Endocrinology and Metabolism 7(1)

Damholt, A., Buchan, A. and Kofod, H. (1998) Eissele, R., Gke, R., Willemer, S., Harthus, H.,
Glucagon-like-peptide-1 secretion from canine L-cells Vermeer, H., Arnold, R. etal. (1992) Glucagon-
is increased by glucose-dependent-insulinotropic like peptide-1 cells in the gastrointestinal tract and
peptide but unaffected by glucose. Endocrinology 139: pancreas of rat, pig and man. Eur J Clin Invest 22:
20852091. 283291.
Daniel, H. (2004) Molecular and integrative Ellingsgaard, H., Hauselmann, I., Schuler, B., Habib,
physiology of intestinal peptide transport. Annu Rev A., Baggio, L., Meier, D. etal. (2011) Interleukin-6
Physiol 66: 361384. enhances insulin secretion by increasing glucagon-like
peptide-1 secretion from L cells and alpha cells. Nat
Diakogiannaki, E., Pais, R., Tolhurst, G., Parker, H., Med 17: 14811489.
Horscroft, J., Rauscher, B. etal. (2013) Oligopeptides
Stimulate glucagon-like peptide-1 secretion in mice Elliott, R., Morgan, L., Tredger, J., Deacon, S.,
through proton-coupled uptake and the calcium- Wright, J. and Marks, V. (1993) Glucagon-like
sensing receptor. Diabetologia 56: 26882696. peptide-1 (736)amide and glucose-dependent
insulinotropic polypeptide secretion in response
Dirksen, C., Hansen, D., Madsbad, S., Hvolris, L., to nutrient ingestion in man: acute post-prandial
Naver, L., Holst, J. etal. (2010) Postprandial diabetic and 24-h secretion patterns. J Endocrinol 138:
glucose tolerance is normalized by gastric bypass 159166.
feeding as opposed to gastric feeding and is associated
with exaggerated GLP-1 secretion: a case report. Ellrichmann, M., Kapelle, M., Ritter, P., Holst,
Diabetes Care 33: 375377. J., Herzig, K., Schmidt, W. etal. (2008) Orlistat
inhibition of intestinal lipase acutely increases
Drewe, J., Gadient, A., Rovati, L. and Beglinger, C. appetite and attenuates postprandial glucagon-like
(1992) Role of circulating cholecystokinin in control peptide-1-(736)-amide-1, cholecystokinin, and
of fat-induced inhibition of food intake in humans. peptide YY concentrations. J Clin Endocrinol Metab
Gastroenterology 102: 16541659. 93: 39953998.
Drucker, D. and Nauck, M. (2006) The incretin Everard, A., Belzer, C., Geurts, L., Ouwerkerk,
system: glucagon-like peptide-1 receptor agonists and J., Druart, C., Bindels, L. etal. (2013) Cross-talk
dipeptidyl peptidase-4 inhibitors in type 2 diabetes. between Akkermansia muciniphila and intestinal
Lancet 368: 16961705. epithelium controls diet-induced obesity. Proc Natl
Acad Sci U S A 110: 90669071.
Dumoulin, V., Dakka, T., Plaisancie, P., Chayvialle,
J. and Cuber, J. (1995) Regulation of glucagon- Everard, A., Lazarevic, V., Derrien, M., Girard, M.,
like peptide-1-(736) amide, peptide YY, and Muccioli, G., Muccioli, G. etal. (2011) Responses of
neurotensin secretion by neurotransmitters and gut gut microbiota and glucose and lipid metabolism to
hormones in the isolated vascularly perfused rat ileum. prebiotics in genetic obese and diet-induced leptin-
Endocrinology 136: 51825188. resistant mice. Diabetes 60: 27752786.

Dupre, J., Ross, S., Watson, D. and Brown, J. (1973) Falkn, Y., Hellstrm, P., Holst, J. and Nslund, E.
Stimulation of insulin secretion by gastric inhibitory (2011) Changes in glucose homeostasis after Roux-
polypeptide in man. J Clin Endocrinol Metab 37: en-Y gastric bypass surgery for obesity at day three,
826828. two months, and one year after surgery: role of gut
peptides. J Clin Endocrinol Metab 96: 22272235.
Dyer, J., Salmon, K., Zibrik, L. and Shirazi-Beechey,
S. (2005) Expression of sweet taste receptors of the Feinle-Bisset, C., Patterson, M., Ghatei, M.,
T1R family in the intestinal tract and enteroendocrine Bloom, S. and Horowitz, M. (2005) Fat digestion is
cells. Biochem Soc Trans 33: 302305. required for suppression of ghrelin and stimulation
of peptide YY and pancreatic polypeptide secretion
Eder, K., Baffy, N., Falus, A. and Fulop, A. (2009) by intraduodenal lipid. Am J Physiol Endocrinol Metab
The major inflammatory mediator interleukin-6 and 289: E948E953.
obesity. Inflamm Res 58: 727736.
Feinle, C., ODonovan, D., Doran, S., Andrews,
Edfalk, S., Steneberg, P. and Edlund, H. (2008) J., Wishart, J., Chapman, I. etal. (2003) Effects of
GPR40 is expressed in enteroendocrine cells and fat digestion on appetite, APD motility, and gut
mediates free fatty acid stimulation of incretin hormones in response to duodenal fat infusion in
secretion. Diabetes 57: 22802287. humans. Am J Physiol Gastrointest Liver Physiol 284:
G798G807.
Egerod, K., Engelstoft, M., Grunddal, K., Nhr, M.,
Secher, A., Sakata, I. etal. (2012) A major lineage of Feltrin, K., Little, T., Meyer, J., Horowitz,
enteroendocrine cells coexpress CCK, secretin, GIP, M., Smout, A., Wishart, J. etal. (2004) Effects
GLP-1, PYY, and neurotensin but not somatostatin. of intraduodenal fatty acids on appetite,
Endocrinology 153: 57825795. antropyloroduodenal motility, and plasma CCK and

36 http://tae.sagepub.com
R Pais, FM Gribble et al.

GLP-1 in humans vary with their chain length. Am J Gorboulev, V., Schrmann, A., Vallon, V., Kipp,
Physiol Regul Integr Comp Physiol 287: R524533. H., Jaschke, A., Klessen, D. etal. (2012) Na(+)-D-
SGLT1 is pivotal for intestinal glucose absorption
Feng, J., Petersen, C., Coy, D., Jiang, J., Thomas,
and glucose-dependent incretin secretion. Diabetes 61:
C., Pollak, M. etal. (2010) Calcium-Sensing receptor
187196.
is a physiologic multimodal chemosensor regulating
gastric G-cell growth and gastrin secretion. Proc Natl Greenfield, J., Farooqi, I., Keogh, J., Henning,
Acad Sci U S A 107: 1779117796. E., Habib, A., Blackwood, A. etal. (2009) Oral
glutamine increases circulating glucagon-like peptide
Flock, G., Cao, X., Maziarz, M. and Drucker, D. 1, glucagon, and insulin concentrations in lean, obese,
(2013) Activation of enteroendocrine membrane and type 2 diabetic subjects. Am J Clin Nutr 89:
progesterone receptors promotes incretin secretion 106113.
and improves glucose tolerance in mice. Diabetes 62:
283290. Gribble, F., Williams, L., Simpson, A. and Reimann,
F. (2003) A novel glucose-sensing mechanism
Forbes, S., Stafford, S., Coope, G., Heffron, H., Real, contributing to glucagon-like peptide-1 secretion from
K., Newman, R. etal. (2015) Selective FFA2 agonism the GLUTag cell line. Diabetes 52: 11471154.
appears to act via intestinal PYY to reduce transit and
food intake, but does not improve glucose tolerance in Grosse, J., Heffron, H., Burling, K., Akhter Hossain,
mouse models. Diabetes 64: 37633771. M., Habib, A., Rogers, G. etal. (2014) Insulin-like
peptide 5 is an orexigenic gastrointestinal hormone.
Fujita, Y., Wideman, R., Speck, M., Asadi, A., King,
Proc Natl Acad Sci U S A 111: 1113311138.
D., Webber, T. etal. (2009) Incretin release from gut
is acutely enhanced by sugar but not by sweeteners Gunnarsson, P., Winzell, M., Deacon, C., Larsen,
in vivo. Am J Physiol Endocrinol Metab 296: M., Jelic, K., Carr, R. etal. (2006) Glucose-Induced
E473E479. incretin hormone release and inactivation are
differently modulated by oral fat and protein in mice.
Gagnon, J., Baggio, L., Drucker, D. and Brubaker,
Endocrinology 147: 31733180.
P. (2015a) Ghrelin is a novel regulator of GLP-1
secretion. Diabetes 64: 15131521. Habib, A., Richards, P., Cairns, L., Rogers, G.,
Gagnon, J., Sauv, M., Zhao, W., Stacey, H., Wiber, Bannon, C., Parker, H. etal. (2012) Overlap of
S., Bolz, S. etal. (2015b) Chronic exposure to tumor endocrine hormone expression in the mouse intestine
necrosis factor impairs secretion of glucagon-like revealed by transcriptional profiling and flow
peptide-1. Endocrinology: en20151361. cytometry. Endocrinology 153: 30543065.

Gameiro, A., Reimann, F., Habib, A., OMalley, Hansen, K., Rosenkilde, M., Knop, F., Wellner, N.,
D., Williams, L., Simpson, A. etal. (2005) The Diep, T., Rehfeld, J. etal. (2011) 2-Oleoyl glycerol
neurotransmitters glycine and GABA stimulate is a GPR119 agonist and signals GLP-1 release in
glucagon-like peptide-1 release from the GLUTag cell humans. J Clin Endocrinol Metab 96: E1409E1417.
line. J Physiol 569: 761772. Harach, T., Pols, T., Nomura, M., Maida, A.,
Gejl, M., Sndergaard, H., Stecher, C., Bibby, B., Watanabe, M., Auwerx, J. etal. (2012) TGR5
Mller, N., Btker, H. etal. (2012) Exenatide alters potentiates GLP-1 secretion in response to anionic
myocardial glucose transport and uptake depending exchange resins. Sci Rep 2: 430.
on insulin resistance and increases myocardial blood
Hata, T., Mera, Y., Ishii, Y., Tadaki, H., Tomimoto,
flow in patients with type 2 diabetes. J Clin Endocrinol
D., Kuroki, Y. etal. (2011) JTT-130, a novel
Metab 97: E11651169.
intestine-specific inhibitor of microsomal triglyceride
Gil-Lozano, M., Mingomataj, E., Wu, W., Ridout, transfer protein, suppresses food intake and gastric
S. and Brubaker, P. (2014) Circadian Secretion of emptying with the elevation of plasma peptide YY and
the intestinal hormone GLP-1 by the rodent L cell. glucagon-like peptide-1 in a dietary fat-dependent
Diabetes 63: 36743685. manner. J Pharmacol Exp Ther 336: 850856.

Glassmeier, G., Herzig, K., Hpfner, M., Lemmer, Hayashi, H., Yamada, R., Das, S., Sato, T.,
K., Jansen, A. and Scherubl, H. (1998) Expression Takahashi, A., Hiratsuka, M. etal. (2014) Glucagon-
of functional GABAA receptors in cholecystokinin- Like peptide-1 production in the GLUTag cell line is
secreting gut neuroendocrine murine STC-1 cells. impaired by free fatty acids via endoplasmic reticulum
J Physiol 510: 805814. stress. Metabolism 63: 800811.

Gmez, R., Navarro, M., Ferrer, B., Trigo, J., Bilbao, Hirasawa, A., Tsumaya, K., Awaji, T., Katsuma, S.,
A., Del Arco, I. etal. (2002) A peripheral mechanism Adachi, T., Yamada, M. etal. (2005) Free fatty acids
for CB1 cannabinoid receptor-dependent modulation regulate gut incretin glucagon-like peptide-1 secretion
of feeding. J Neurosci 22: 96129617. through GPR120. Nat Med 11: 9094.

http://tae.sagepub.com 37
Therapeutic Advances in Endocrinology and Metabolism 7(1)

Hotamisligil, G., Shargill, N. and Spiegelman, B. Keophiphath, M., Rouault, C., Divoux, A.,
(1993) Adipose expression of tumor necrosis factor- Clment, K. and Lacasa, D. (2010) CCl5 promotes
alpha: direct role in obesity-linked insulin resistance. macrophage recruitment and survival in human
Science 259: 8791. adipose tissue. Arterioscler Thromb Vasc Biol 30: 3945.

Iakoubov, R., Ahmed, A., Lauffer, L., Bazinet, R. and Kieffer, T. and Habener, J. (1999) The glucagon-like
Brubaker, P. (2011) Essential role for protein kinase peptides. Endocrine Reviews 20: 876913.
C in oleic acid-induced glucagon-like peptide-1
Kokrashvili, Z., Mosinger, B. and Margolskee, R.
secretion in vivo in the rat. Endocrinology 152:
(2009) T1R3 and alpha-gustducin in gut regulate
12441252.
secretion of glucagon-like peptide-1. Ann N Y Acad
Iakoubov, R., Izzo, A., Yeung, A., Whiteside, C. and Sci 1170: 9194.
Brubaker, P. (2007) Protein kinase Czeta is required Korner, J., Bessler, M., Cirilo, L., Conwell, I., Daud,
for oleic acid-induced secretion of glucagon-like A., Restuccia, N. etal. (2005) Effects of Roux-en-Y
peptide-1 by intestinal endocrine L cells. Endocrinology gastric bypass surgery on fasting and postprandial
148: 10891098. concentrations of plasma ghrelin, peptide YY, and
Iwasaki, K., Harada, N., Sasaki, K., Yamane, S., Iida, insulin. J Clin Endocrinol Metab 90: 359365.
K., Suzuki, K. etal. (2015) Free fatty acid receptor Korner, J., Bessler, M., Inabnet, W., Taveras, C. and
GPR120 is highly expressed in enteroendocrine K Holst, J. (2007) Exaggerated glucagon-like peptide-1
cells of the upper small intestine and has a critical role and blunted glucose-dependent insulinotropic peptide
in GIP secretion after fat ingestion. Endocrinology 156: secretion are associated with Roux-en-Y gastric bypass
837846. but not adjustable gastric banding. Surg Obes Relat Dis
Izzo, A. and Sharkey, K. (2010) Cannabinoids and 3: 597601.
the gut: new developments and emerging concepts. Kreymann, B., Williams, G., Ghatei, M. and
Pharmacol Ther 126: 2138. Bloom, S. (1987) Glucagon-like peptide-1 736: a
Jang, H., Kokrashvili, Z., Theodorakis, M., Carlson, physiological incretin in man. Lancet 2: 13001304.
O., Kim, B., Zhou, J. etal. (2007) Gut-expressed
Kuhre, R., Frost, C., Svendsen, B. and Holst, J.
gustducin and taste receptors regulate secretion of
(2015) Molecular mechanisms of glucose-stimulated
glucagon-like peptide-1. Proc Natl Acad Sci U S A
GLP-1 secretion from perfused rat small intestine.
104: 1506915074.
Diabetes 64: 370382.
Jiang, Y., Rose, A., Sijmonsma, T., Brer, A.,
Kuhre, R., Gribble, F., Hartmann, B., Reimann,
Pfenninger, A., Herzig, S. etal. (2015) Mice lacking
F., Windelv, J., Rehfeld, J. etal. (2014) Fructose
neutral amino acid transporter B(0)AT1 (SlC6A19)
stimulates GLP-1 but not GIP secretion in mice, rats,
have elevated levels of FGF21 and GLP-1 and
and humans. Am J Physiol Gastrointest Liver Physiol
improved glycaemic control. Mol Metab 4:
306: G622G630.
406417.
Lan, H., Vassileva, G., Corona, A., Liu, L., Baker,
Jimnez, A., Casamitjana, R., Flores, L., Viaplana, J.,
H., Golovko, A. etal. (2009) GPR119 Is required for
Corcelles, R., Lacy, A. etal. (2012) Long-term effects
physiological regulation of glucagon-like peptide-1
of sleeve gastrectomy and Roux-en-Y gastric bypass
secretion but not for metabolic homeostasis. J
surgery on type 2 diabetes mellitus in morbidly obese
Endocrinol 201: 219230.
subjects. Ann Surg 256: 10231029.
Lauffer, L., Iakoubov, R. and Brubaker, P. (2009)
Katsuma, S., Hirasawa, A. and Tsujimoto, G. (2005)
GPR119 Is essential for oleoylethanolamide-induced
Bile acids promote glucagon-like peptide-1 secretion
glucagon-like peptide-1 secretion from the intestinal
through TGR5 in a murine enteroendocrine cell line
enteroendocrine L-cell. Diabetes 58: 10581066.
STC-1. Biochem Biophys Res Commun 329: 386390.
Ley, R., Bckhed, F., Turnbaugh, P., Lozupone,
Katz, L., Gambale, J., Rothenberg, P., Vanapalli,
C., Knight, R. and Gordon, J. (2005) Obesity alters
S., Vaccaro, N., Xi, L. etal. (2012) Effects of JNJ-
gut microbial ecology. Proc Natl Acad Sci U S A 102:
38431055, a novel GPR119 receptor agonist, in
1107011075.
randomized, double-blind, placebo-controlled studies
in subjects with type 2 diabetes. Diabetes Obes Metab Li, T., Holmstrom, S.R., Kir, S., Umetani, M.,
14: 709716. Schmidt, D.R., Kliewer, S.A. and Mangelsdorf, D.J.
(2011) The G protein-coupled bile acid receptor,
Kellett, G. and Helliwell, P. (2000) The diffusive TGR5, stimulates gallbladder filling. Mol Endocrinol
component of intestinal glucose absorption is 25: 10661071.
mediated by the glucose-induced recruitment of
GLUT2 to the brush-border membrane. Biochem J Lim, G., Huang, G., Flora, N., Leroith, D.,
350: 155162. Rhodes, C. and Brubaker, P. (2009) Insulin

38 http://tae.sagepub.com
R Pais, FM Gribble et al.

regulates glucagon-like peptide-1 secretion from the secretion in primary murine L-cells. Peptides. DOI:
enteroendocrine L cell. Endocrinology 150: 580591. 10.1016/j.peptides.2015.06.012.
Lin, H., Frassetto, A., Kowalik, E., Nawrocki, A., Lu, Moss, C., Marsh, W., Parker, H., Ogunnowo-Bada,
M., Kosinski, J. etal. (2012) Butyrate and propionate E., Riches, C., Habib, A. etal. (2012) Somatostatin
protect against diet-induced obesity and regulate gut receptor 5 and cannabinoid receptor 1 activation
hormones via free fatty acid receptor 3-independent inhibit secretion of glucose-dependent insulinotropic
mechanisms. PLoS One 7: e35240. polypeptide from intestinal K cells in rodents.
Diabetologia 55: 30943103.
Liou, A., Sei, Y., Zhao, X., Feng, J., Lu, X., Thomas,
C. etal. (2011) The extracellular calcium-sensing Nauck, M., Heimesaat, M., Orskov, C., Holst, J.,
receptor is required for cholecystokinin secretion Ebert, R. and Creutzfeldt, W. (1993) Preserved
in response to L-phenylalanine in acutely isolated incretin activity of glucagon-like peptide 1 [736
intestinal I cells. Am J Physiol Gastrointest Liver Physiol amide] but not of synthetic human gastric inhibitory
300: G538G546. polypeptide in patients with type-2 diabetes mellitus.
Little, T., Feltrin, K., Horowitz, M., Smout, A., J Clin Invest 91: 301307.
Rades, T., Meyer, J. etal. (2005) Dose-related effects Ogata, H., Seino, Y., Harada, N., Iida, A., Suzuki,
of lauric acid on antropyloroduodenal motility, K., Izumoto, T. etal. (2014) KATP channel as well as
gastrointestinal hormone release, appetite, and energy SGLT1 participates in GIP secretion in the diabetic
intake in healthy men. Am J Physiol Regul Integr Comp state. J Endocrinol 222: 191200.
Physiol 289: R1090R1098.
Oh-I, S., Shimizu, H., Satoh, T., Okada, S., Adachi,
Lu, W., Yang, Q., Yang, L., Lee, D., Dalessio, D. S., Inoue, K. etal. (2006) Identification of nesfatin-1
and Tso, P. (2012) Chylomicron formation and as a satiety molecule in the hypothalamus. Nature 443:
secretion is required for lipid-stimulated release of 709712.
incretins GLP-1 and GIP. Lipids 47: 571580.
Okawa, M., Fujii, K., Ohbuchi, K., Okumoto, M.,
Ma, J., Bellon, M., Wishart, J., Young, R., Blackshaw,
Aragane, K., Sato, H. etal. (2009) Role of MGAT2
L., Jones, K. etal. (2009) Effect of the artificial
and DGAT1 in the release of gut peptides after
sweetener, sucralose, on gastric emptying and incretin
triglyceride ingestion. Biochem Biophys Res Commun
hormone release in healthy subjects. Am J Physiol
390: 377381.
Gastrointest Liver Physiol 296: G735G739.
Overton, H., Babbs, A., Doel, S., Fyfe, M., Gardner,
Mace, O., Schindler, M. and Patel, S. (2012) The
L., Griffin, G. etal. (2006) Deorphanization of a G
regulation of K- and L-cell activity by GLUT2 and
protein-coupled receptor for oleoylethanolamide and
the calcium-sensing receptor CASR in rat small
its use in the discovery of small-molecule hypophagic
intestine. J Physiol 590: 29172936.
agents. Cell Metab 3: 167175.
Mande, M., Hansen, K., Hartmann, B.,
Oya, M., Kitaguchi, T., Pais, R., Reimann, F.,
Rehfeld, J., Holst, J. and Hansen, H. (2015) The
Gribble, F. and Tsuboi, T. (2013) The G Protein-
2-monoacylglycerol moiety of dietary fat appears
coupled receptor family C group 6 subtype a
to be responsible for the fat-induced release of
(GPRC6a) receptor is involved in amino acid-induced
GLP-1 in humans. Am J Clin Nutr. DOI: 10.3945/
glucagon-like peptide-1 secretion from GLUTag cells.
ajcn.115.106799.
J Biol Chem 288: 45134521.
Martins, C., Morgan, L., Bloom, S. and Robertson,
Pais, R., Gribble, F. and Reimann, F. (2015)
M. (2007) Effects of exercise on gut peptides, energy
Signalling pathways involved in the detection
intake and appetite. J Endocrinol 193: 251258.
of peptones by murine small intestinal
Mornigo, R., Vidal, J., Lacy, A., Delgado, S., enteroendocrine L-cells. Peptides. DOI: 10.1016/j.
Casamitjana, R. and Gomis, R. (2008) Circulating peptides.2015.07.019.
peptide YY, weight loss, and glucose homeostasis after
Pais, R., Zietek, T., Hauner, H., Daniel, H. and
gastric bypass surgery in morbidly obese subjects. Ann
Skurk, T. (2014) Rantes (CCL5) reduces glucose-
Surg 247: 270275.
dependent secretion of glucagon-like peptides 1 and
Moriya, R., Shirakura, T., Ito, J., Mashiko, S. 2 and impairs glucose-induced insulin secretion in
and Seo, T. (2009) Activation of sodium-glucose mice. Am J Physiol Gastrointest Liver Physiol 307:
cotransporter 1 ameliorates hyperglycemia by G330G337.
mediating incretin secretion in mice. Am J Physiol
Parker, H., Adriaenssens, A., Rogers, G., Richards,
Endocrinol Metab 297: E1358E1365.
P., Koepsell, H., Reimann, F. etal. (2012)
Moss, C., Glass, L., Diakogiannaki, E., Pais, Predominant role of active versus facilitative glucose
R., Lenaghan, C., Smith, D. etal. (2015) Lipid transport for glucagon-like peptide-1 secretion.
derivatives activate GPR119 and trigger GLP-1 Diabetologia 55: 24452455.

http://tae.sagepub.com 39
Therapeutic Advances in Endocrinology and Metabolism 7(1)

Parker, H., Habib, A., Rogers, G., Gribble, F. and through enhancement of splanchnic glucose
Reimann, F. (2009) Nutrient-dependent secretion of utilization. Trends Endocrinol Metab 25: 235244.
glucose-dependent insulinotropic polypeptide from
primary murine K cells. Diabetologia 52: 289298. Qin, J., Li, Y., Cai, Z., Li, S., Zhu, J., Zhang, F. etal.
(2012) A metagenome-wide association study of gut
Parker, H., Wallis, K., Le Roux, C., Wong, K., microbiota in type 2 diabetes. Nature 490: 5560.
Reimann, F. and Gribble, F. (2012) Molecular
mechanisms underlying bile acid-stimulated glucagon- Ramesh, N., Mortazavi, S. and Unniappan, S.
like peptide-1 secretion. Br J Pharmacol 165: (2015) Nesfatin-1 stimulates glucagon-like peptide-1
414423. and glucose-dependent insulinotropic polypeptide
secretion from STC-1 cells in vitro. Biochem Biophys
Patti, M., Houten, S., Bianco, A., Bernier, R., Res Commun 462: 124130.
Larsen, P., Holst, J. etal. (2009) Serum bile acids
are higher in humans with prior gastric bypass: Reimann, F., Habib, A., Tolhurst, G., Parker, H.,
potential contribution to improved glucose and lipid Rogers, G. and Gribble, F. (2008) Glucose sensing
metabolism. Obesity 17: 16711677. in L cells: a primary cell study. Cell Metabolism 8:
532539.
Perley, M. and Kipnis, D. (1967) Plasma insulin
responses to oral and intravenous glucose: studies Reimann, F., Williams, L., Da Silva Xavier, G.,
in normal and diabetic subjects. J Clin Invest 46: Rutter, G. and Gribble, F. (2004) Glutamine potently
19541962. stimulates glucagon-like peptide-1 secretion from
Persson, K., Gingerich, R., Nayak, S., Wada, K., GLUTag cells. Diabetologia 47: 15921601.
Wada, E. and Ahrn, B. (2000) Reduced GLP-1 and Reimer, R. (2006) Meat hydrolysate and essential
insulin responses and glucose intolerance after gastric amino acid-induced glucagon-like peptide-1 secretion,
glucose in GRP receptor-deleted mice. Am J Physiol in the human NCI-H716 enteroendocrine cell line, is
Endocrinol Metab 279: E956E962. regulated by extracellular signal-regulated kinase1/2
Petersen, N., Reimann, F., Bartfeld, S., Farin, H., and P38 mitogen-activated protein kinases. J
Ringnalda, F., Vries, R. etal. (2014) Generation of L Endocrinol 191: 159170.
cells in mouse and human small intestine organoids.
Richards, P., Pais, R., Habib, A., Brighton, C.,
Diabetes 63: 410420.
Yeo, G., Reimann, F. etal. (2015) High Fat diet
Petersen, N., Reimann, F., Van Es, J., Van Den impairs the function of glucagon-like peptide-1
Berg, B., Kroone, C., Pais, R. etal. (2015) Targeting producing L-cells. Peptides. DOI: 10.1016/j.
development of incretin-producing cells increases peptides.2015.06.006.
insulin secretion. J Clin Invest 125: 379385.
Ritzel, U., Fromme, A., Ottleben, M., Leonhardt, U.
Plaisancie, P., Bernard, C., Chayvialle, J. and and Ramadori, G. (1997) Release of glucagon-like
Cuber, J. (1994) Regulation of Glucagon-like peptide-1 (GLP-1) by carbohydrates in the perfused
peptide-1-(736) amide secretion by intestinal rat ileum. Acta Diabetol 34: 1821.
neurotransmitters and hormones in the isolated
vascularly perfused rat colon. Endocrinology 135: Roberge, J. and Brubaker, P. (1993) Regulation of
23982403. intestinal proglucagon-derived peptide secretion by
glucose-dependent insulinotropic peptide in a novel
Poreba, M., Dong, C., Li, S., Stahl, A., Miner, J. enteroendocrine loop. Endocrinology 133: 233240.
and Brubaker, P. (2012) Role of fatty acid transport
protein 4 in oleic acid-induced glucagon-like Roberge, J., Gronau, K. and Brubaker, P. (1996)
peptide-1 secretion from murine intestinal L cells. Am Gastrin-releasing peptide is a novel mediator of
J Physiol Endocrinol Metab 303: E899907. proximal nutrient-induced proglucagon-derived
peptide secretion from the distal gut. Endocrinology
Powell, D., Dacosta, C., Gay, J., Ding, Z., Smith, 137: 23832388.
M., Greer, J. etal. (2013a) Improved glycemic control
in mice lacking SGLT1 and SGLT2. Am J Physiol Rder, P., Geillinger, K., Zietek, T., Thorens, B.,
Endocrinol Metab 304: E117E130. Koepsell, H. and Daniel, H. (2014) The role of
SGLT1 and GLUT2 in intestinal glucose transport
Powell, D., Smith, M., Greer, J., Harris, A., Zhao, S., and sensing. PLoS One 9: e89977.
Dacosta, C. etal. (2013b) Lx4211 increases serum
glucagon-like peptide 1 and peptide YY levels by Rodrguez De Fonseca, F., Navarro, M., Gmez,
reducing sodium/glucose cotransporter 1 (SGLT1)- R., Escuredo, L., Nava, F., Fu, J. etal. (2001) An
mediated absorption of intestinal glucose. J Pharmacol anorexic lipid mediator regulated by feeding. Nature
Exp Ther 345: 250259. 414: 209212.
Prawitt, J., Caron, S. and Staels, B. (2014) Glucose- Rozengurt, N., Wu, S., Chen, M., Huang, C.,
Lowering effects of intestinal bile acid sequestration Sternini, C. and Rozengurt, E. (2006) Colocalization

40 http://tae.sagepub.com
R Pais, FM Gribble et al.

of the alpha-subunit of gustducin with PYY and short-term therapy with glibenclamide and repaglinide
GLP-1 in L cells of human colon. Am J Physiol on incretin hormones and oxidative damage associated
Gastrointest Liver Physiol 291: G792G802. with postprandial hyperglycaemia in people with
type 2 diabetes mellitus. Diabetes Res Clin Pract 94:
Samocha-Bonet, D., Wong, O., Synnott, E.,
199206.
Piyaratna, N., Douglas, A., Gribble, F. etal. (2011)
Glutamine reduces postprandial glycemia and Sugiyama, K., Manaka, H., Kato, T., Yamatani, K.,
augments the glucagon-like peptide-1 response in type Tominaga, M. and Sasaki, H. (1994) Stimulation of
2 diabetes patients. J Nutr 141: 12331238. truncated glucagon-like peptide-1 release from the
isolated perfused canine ileum by glucose absorption.
Senn, J., Klover, P., Nowak, I. and Mooney, R.
Digestion 55: 2428.
(2002) Interleukin-6 induces cellular insulin resistance
in hepatocytes. Diabetes 51: 33913399. Suzuki, K., Harada, N., Yamane, S., Nakamura, Y.,
Sasaki, K., Nasteska, D. etal. (2013) Transcriptional
Seino, Y., Ogata, H., Maekawa, R., Izumoto, T.,
regulatory factor X6 (RFX6) increases gastric
Iida, A., Harada, N., Miki, T., Seino, S., Inagaki,
inhibitory polypeptide (GIP) expression in
N., Tsunekawa, S., Oiso, Y. and Hamada, Y. (2015)
enteroendocrine K-cells and is involved in GIP
Fructose induces glucose-dependent insulinotropic
hypersecretion in high fat diet-induced obesity. J Biol
polypeptide, glucagon-like peptide-1 and insulin
Chem 288: 19291938.
secretion: Role of adenosine triphosphate-sensitive
K(+) channels. J Diabetes Investig 6: 522526. Svendsen, B., Pedersen, J., Albrechtsen, N.,
Hartmann, B., Torng, S., Rehfeld, J. etal. (2015)
Shibue, K., Yamane, S., Harada, N., Hamasaki, A.,
An analysis of cosecretion and coexpression of gut
Suzuki, K., Joo, E. etal. (2015) Fatty acid-binding
hormones from male rat proximal and distal small
protein 5 regulates diet-induced obesity via GIP
intestine. Endocrinology 156: 847857.
secretion from enteroendocrine K cells in response
to fat ingestion. Am J Physiol Endocrinol Metab 308: Sykaras, A., Demenis, C., Cheng, L., Pisitkun, T.,
E583E591. McLaughlin, J., Fenton, R. etal. (2014) Duodenal
CCK cells from male mice express multiple hormones
Shima, K., Suda, T., Nishimoto, K. and Yoshimoto,
including ghrelin. Endocrinology 155: 33393351.
S. (1990) Relationship between molecular structures
of sugars and their ability to stimulate the release of Tang, C., Ahmed, K., Gille, A., Lu, S., Grne, H.,
glucagon-like peptide-1 from canine ileal loops. Acta Tunaru, S. etal. (2015) Loss of FFA2 and FFA3
Endocrinol 123: 464470. Increases insulin secretion and improves glucose
tolerance in type 2 diabetes. Nat Med 21: 173177.
Shimotoyodome, A., Fukuoka, D., Suzuki, J., Fujii,
Y., Mizuno, T., Meguro, S. etal. (2009) Coingestion Theodorakis, M., Carlson, O., Michopoulos, S.,
of acylglycerols differentially affects glucose-induced Doyle, M., Juhaszova, M., Petraki, K. etal. (2006)
insulin secretion via glucose-dependent insulinotropic Human duodenal enteroendocrine cells: source of
polypeptide in C57BL/6J mice. Endocrinology 150: both incretin peptides, GLP-1 and GIP. Am J Physiol
21182126. Endocrinol Metab 290: E550E559.
Simpson, A.K., Ward, P.S., Wong, K.Y., Collord, Thomas, C., Gioiello, A., Noriega, L., Strehle, A.,
G.J., Habib, A.M., Reimann, F. and Gribble, F.M. Oury, J., Rizzo, G. etal. (2009) TGR5-mediated bile
(2007) Cyclic AMP triggers glucagon-like peptide-1 acid sensing controls glucose homeostasis. Cell Metab
secretion from the GLUTag enteroendocrine cell line. 10: 167177.
Diabetologia 50: 21812189.
Tolhurst, G., Heffron, H., Lam, Y., Parker, H.,
Soga, T., Ohishi, T., Matsui, T., Saito, T., Habib, A., Diakogiannaki, E. etal. (2012) Short-chain
Matsumoto, M., Takasaki, J. etal. (2005) fatty acids stimulate glucagon-like peptide-1 secretion
Lysophosphatidylcholine enhances glucose-dependent via the g-protein-coupled receptor FFAR2. Diabetes
insulin secretion via an orphan G-protein-coupled 61: 364371.
receptor. Biochem Biophys Res Commun 326: 744751.
Tolhurst, G., Zheng, Y., Parker, H., Habib, A.,
Steinert, R., Gerspach, A., Gutmann, H., Asarian, L., Reimann, F. and Gribble, F. (2011) Glutamine
Drewe, J. and Beglinger, C. (2011) The functional triggers and potentiates glucagon-like peptide-1
involvement of gut-expressed sweet taste receptors secretion by raising cytosolic Ca2+ and camp.
in glucose-stimulated secretion of glucagon-like Endocrinology 152: 405413.
peptide-1 (GLP-1) and peptide YY (Pyy). Clin Nutr
Trabelsi, M., Daoudi, M., Prawitt, J., Ducastel,
30: 524532.
S., Touche, V., Sayin, S. etal. (2015) Farnesoid X
Stephens, J., Bodvarsdottir, T., Wareham, K., Prior, receptor inhibits glucagon-like peptide-1 production
S., Bracken, R., Lowe, G. etal. (2011) Effects of by enteroendocrine L cells. Nat Commun 6: 7629.

http://tae.sagepub.com 41
Therapeutic Advances in Endocrinology and Metabolism 7(1)

Tremaroli, V. and Bckhed, F. (2012) Functional modulation of energy availability in the colon
interactions between the gut microbiota and host regulates intestinal transit. Cell Host Microbe 14:
metabolism. Nature 489: 242249. 582590.
Ullmer, C., Alvarez Sanchez, R., Sprecher, U., Raab, Wu, H., Ghosh, S., Perrard, X., Feng, L., Garcia,
S., Mattei, P., Dehmlow, H. etal. (2013) Systemic G., Perrard, J. etal. (2007) T-cell accumulation and
bile acid sensing by G protein-coupled bile acid regulated on activation, normal T cell expressed and
receptor 1 (GPBAR1) promotes PYY and GLP-1 secreted upregulation in adipose tissue in obesity.
release. Br J Pharmacol 169: 671684. Circulation 115: 10291038.
Wang, F., Yang, Q., Huesman, S., Xu, M., Li, X., Young, R., Chia, B., Isaacs, N., Ma, J., Khoo, J., Wu,
Lou, D. etal. (2015) The role of apolipoprotein a-IV T. etal. (2013) Disordered Control of intestinal sweet
in regulating glucagon-like peptide-1 secretion. Am J taste receptor expression and glucose absorption in
Physiol Gastrointest Liver Physiol 309: G680G687. type 2 diabetes. Diabetes 62: 35323541.
Wang, Y., Chandra, R., Samsa, L., Gooch, B., Fee, Zawalich, W., Cote, S. and Diaz, V. (1986) Influence
B., Cook, J. etal. (2011) Amino Acids stimulate of cholecystokinin on insulin output from isolated
cholecystokinin release through the Ca2+-sensing perifused pancreatic islets. Endocrinology 119:
receptor. Am J Physiol Gastrointest Liver Physiol 300: 616621.
G528G537.
Zhang, Y., Proenca, R., Maffei, M., Barone, M.,
Welch, I., Saunders, K. and Read, N. (1985) Effect Leopold, L. and Friedman, J.M. (1994) Positional
of ileal and intravenous infusions of fat emulsions cloning of the mouse obese gene and its human
on feeding and satiety in human volunteers. homologue. Nature 372: 425432.
Visit SAGE journals online
Gastroenterology 89: 12931297.
Zietek, T. and Daniel, H. (2015) Intestinal nutrient
http://tae.sagepub.com
Wichmann, A., Allahyar, A., Greiner, T., Plovier, sensing and blood glucose control. Curr Opin Clin
SAGE journals H., Lundn, G., Larsson, T. etal. (2013) Microbial Nutr Metab Care 18: 381388.

42 http://tae.sagepub.com

You might also like