SlideShare a Scribd company logo
1 of 11
Download to read offline
REVIEWS
Ghrelin, the proglucagon-derived peptides
and peptide YY in nutrient homeostasis
Charlotte X. Dong and Patricia L. Brubaker
Abstract | Dysregulation of nutrient homeostasis is implicated in the current epidemics of obesity and type 2
diabetes mellitus. The maintenance of homeostasis in the setting of repeated cycles of feeding and fasting
occurs through complex interactions between metabolic, hormonal and neural factors. Although pancreatic
islets, the liver, muscle, adipocytes and the central nervous system are all key players in this network, the
gastrointestinal tract is the first tissue exposed to ingested nutrients and thus has an important role. This
Review focuses on several of the endocrine hormones released by the gastrointestinal tract prior to or during
nutrient ingestion that have key roles in maintaining energy balance. These hormones include the gastric
orexigenic hormone, ghrelin, and the distal L cell anorexigenic and metabolic hormones, glucagon-like peptide
(GLP)‑1, GLP‑2, oxyntomodulin and peptide YY. Each of these hormones exerts a distinct set of biological
actions to maintain nutrient homeostasis, the properties of which are currently, or might soon be, exploited in
the clinic for the treatment of obesity and type 2 diabetes mellitus.
Dong, C. X. & Brubaker, P Nat. Rev. Gastroenterol. Hepatol. 9, 705–715 (2012); published online 2 October 2012;
. L.
doi:10.1038/nrgastro.2012.185

Introduction
A complex interplay of metabolic, hormonal and neural
factors is required to maintain nutrient homeostasis
under a wide variety of environmental conditions includ­
ing, most notably, the daily cycle of feeding and fasting.
Although pancreatic islets, the liver, muscle, adipocytes
and the central nervous system (CNS) are all key players
in this network, the gastrointestinal tract is the first tissue
affected by nutrient ingestion. As dysregulation of nutri­
ent homeostasis might be linked to the current epi­demics
of obesity and type 2 diabetes mellitus, this Review
focuses on several of the hormones released by the
gastro­intestinal tract prior to or during nutrient ingestion
that have key roles in maintaining energy balance.
Many gastrointestinal hormones have now been iden­
tified,1 making the gut the largest endocrine organ of the
body; most of these hormones are produced by entero­
endocrine cells scattered throughout the gastro­intestinal
epithelium. 2 These cells were originally classified as
being either ‘open’ or ‘closed’ to the intestinal lumen on
the basis of the presence or absence of microvilli on the
luminal surface.3 Some enteroendocrine cells are also
directly innervated and/or extend long ‘axonal-like’ pro­
cesses into the lamina propria, both of which probably
permit direct interactions with the nervous system.4,5
These findings are consistent with the expression of
multiple entero­ ndocrine hormone receptors by the
e
Competing interests
C. X. Dong declares no competing interests. P Brubaker
. L.
declares associations with the following companies: Eli Lilly;
Merck, Sharp & Dome; NPS Pharmaceuticals. See the article
online for full details of the relationships.

vagus nerve.6 Open-type enteroendocrine cells seem to
‘sense’ luminal nutrients through multiple mechanisms,
including both classic transporters and membranebound nutrient receptors, through which intracellular
signalling pathways are activated.7 Collectively, these
findings indicate the existence of a highly inter-related
network between luminal nutrients, gut hormones and
the nervous system.
Two enteroendocrine cells have been selected for this
Review on the basis of the demonstrated importance
of their constituent hormones to whole body energy
homeo­ tasis, as well as on the basis of their actual or
s
potential use in the treatment of the prevalent syndromes
of nutrient dysregulation, obesity and type 2 diabetes
mellitus. These two cells are the proximal X/A-like cell,
which releases ghrelin, and the distal L cell, which pro­
duces the proglucagon-­ erived peptides, glucagon-like
d
peptide (GLP)‑1, GLP‑2 and oxynto­ odulin, as well as
m
peptide YY (PYY). Of note, although all of these peptides
are also synthesized in the brain, the focus of this Review
is on the gastro­ ntestinal tract as the major source of
i
these hormones in the peripheral circulation. Finally, the
focus of this Review on these specific peptides does not
discount the key contributions of other gut hormones
to nutrient homeostasis, including glucose-dependent
insulino­ ropic peptide (GIP) and cholecystokinin.8,9
t
Indeed, develop­ ental, functional and compensatory
m
relationships between ghrelin, GLP‑1, GLP‑2, PYY, GIP
and cholecystokinin have been reported,8,10,11 indicat­
ing that the gastrointestinal hormones function in a
highly integrated fashion to optimize nutrient digestion,
absorption and assimilation.

NATURE REVIEWS | GASTROENTEROLOGY & HEPATOLOGY 	
© 2012 Macmillan Publishers Limited. All rights reserved

Department of
Physiology, Medical
Sciences Building,
University of Toronto,
1 King’s College Circle,
Toronto, ON M5S 1A8,
Canada (C. X. Dong,
P. L. Brubaker).
Correspondence to:
P Brubaker
. L.
p.brubaker@utoronto.ca

VOLUME 9  |  DECEMBER 2012  |  705
REVIEWS
Key points
■■ Enteroendocrine cells in the gastrointestinal tract produce various hormones
that have essential roles in the maintenance of nutrient homeostasis in both
the fasting and fed states
■■ Ghrelin is an orexigenic peptide released by the proximal X/A-like cells prior to
nutrient ingestion; the enteroendocrine L cells release glucagon-like peptide
(GLP)‑1, GLP‑2, oxyntomodulin and peptide YY (PYY) in response to nutrient
ingestion
■■ GLP‑1 is a key incretin hormone, which also inhibits glucagon release, gastric
emptying and appetite; agents that increase GLP‑1 action are used to treat
type 2 diabetes mellitus
■■ GLP‑2 regulates nutrient absorption by the gastrointestinal tract through
enhancement of intestinal growth, nutrient digestion and transport, as well as
blood flow
■■ The physiologic role of oxyntomodulin remains to be established, but
exogenous administration suppresses food intake; PYY3–36NH2 has an important
role as an anorexigenic gut hormone
■■ Surgically induced alterations in the levels of ghrelin and/or GLP‑1,
oxyntomodulin and PYY improve glycaemic control and induce weight loss;
pharmacological manipulation of these gut hormones might prove beneficial

The X/A-like cell
Ghrelin was first discovered in 1999 as a growthhormone-­releasing peptide.12 The following character­
istics make ghrelin unique amongst the gut hormones:
first, it is located primarily in the proximal gut; second,
it is produced by both closed-type and open-type entero­
endocrine cells; third, it is acylated; fourth, levels are
increased in the fasting state; and fifth, it is orexigenic.
As a consequence, the regulation of ghrelin synthesis,
secretion and biological activities differs markedly from
that of GLP‑1, GLP‑2, oxyntomodulin and PYY.
The rat X/A-like cell (known as the P/D 1 cell in
humans) was identified as the ghrelin-producing cell in
2000, having originally been named ‘X’ for its unknown
hormone product and ‘A-like’ for the morphologic simi­
larity of its secretory granules with those of the pancre­
atic A cell.13,14 X/A-like cells represent ~20% of the total
number of endocrine cells of the gastric corpus mucosa
in rats, with profoundly reduced cell numbers along the
aboral axis.15 Interestingly, the proximal X/A-like cells
are almost exclusively closed-type enteroendocrine cells,
whereas >60% of the distal X/A-like cells (that is, ileal,
cecal and colonic) are open type.15 Although the func­
tional importance of these different cell types remains
unclear, their morphologic characteristics suggest that
these cells might be differentially regulated, such that the
distal, but not the proximal, cells might be responsive to
the presence of nutrients in the intestinal lumen.
Synthesized by post-translational processing of
proghrelin through the actions of prohormone con­
vertase (PC) 1/3,16 ghrelin is modified on Ser3 through
covalent linkage to a medium-chain (C8–C10) fatty
acid by ghrelin O‑acyltransferase (GOAT).17 This acyla­
tion is required for the activity of ghrelin on the growth
hormone secretagogue receptor, now also known as
the ghrelin receptor (GRLN‑R),12 and is a unique posttranslational modification amongst all of the known
peptide hormones. Hence, GOAT levels determine the
proportion of bioactive ghrelin that is released by the
X/A-like cell.
706  |  DECEMBER 2012  |  VOLUME 9

Circulating levels of ghrelin increase before a meal in
both rodents and humans, and are suppressed by nutri­
ent ingestion (Figure 1).18,19 Given the closed nature of
the proximal X/A-like cell, it has proven difficult to elu­
cidate the exact mechanisms regulating ghrelin release in
response to fasting and nutrient ingestion. Nonetheless,
in vivo studies have implicated both sympathetic and
parasympathetic neurons as stimulatory effectors of
the X/A-like cell, as well as hyperglycaemia, insulin and
somato­ tatin from gastric D cells as inhibitory regulators
s
of ghrelin release.20–24 Studies on primary rodent gastric
X/A-like cell cultures have confirmed the stimulatory
effects of epinephrine and norepinephrine on ghrelin
release.25,26 Furthermore, 1 mM glucose (comparable
to hypo­ lycaemia) enhanced ghrelin release, whereas
g
5 mM ‘normoglycaemic’ glucose had no effect and 10 mM
glucose inhibited ghrelin release; inhibitory effects of
insulin were also observed but only in the setting of low
or normal glucose levels.25,26 These findings are consistent
with the demonstration that these cells express glucosesensing machinery, including transporters, hexokinases
and the KATP channel, as well as a functional insulin recep­
tor/phosphatidylinositol 3 (PI3)-kinase/Akt signalling
pathway.25,26 Hence, the available data suggest that ghrelin
release before a meal is coupled to activation of the sym­
pathetic nervous system. Conversely, the prandial suppres­
sion of ghrelin secretion is probably mediated, at least in
part, by increasing levels of glucose and/or insulin. Finally,
given the original identification of ghrelin-producing cells
as ‘A-like’ on the basis of their morphological characteris­
tics,13,14 it is interesting that the regulation of ghrelin secre­
tion by X/A-like cells bears a striking resemblance to that
of pancreatic glucagon by the A cell, which is also stimu­
lated by the sympathetic nervous system and inhibited by
high levels of both glucose and insulin.27,28 The finding of
a lineage relationship between pancreatic islet ghrelin and
A cells further suggests that the gastric X/A-like and islet
A cells might also be related,29 although transgenic expres­
sion studies have not identified any such link to date.29–31
Nonetheless, these findings suggest that, under both
fasting and fed conditions, ghrelin and glucagon secretion
are coordinated to maintain nutrient homeostasis.
The orexigenic action of ghrelin and the require­
ment for the GRLN‑R in nutrient homeostasis has been
demon­strated in rodents and humans using a wide variety
of gain-of-function and loss-of-function approaches, and
is mediated not only by increased food intake, but also
via decreased energy expenditure.32–36 As ghrelin crosses
the blood–brain barrier, related in part to its acylation
status,37 the orexigenic effects of this gut hormone are
thought to be mediated through direct effects on neurons
in the central feeding centre, including those expressing
the orexins.32,38 However, the GRLN‑R is also expressed
by the vagus nerve and, although controversial, one study
has demonstrated that both truncal vagotomy and vagal
afferent ablation can prevent the orexigenic actions of
peripherally administered ghrelin on food intake.39
Studies have also demonstrated that ghrelin has a
role in glucose homeostasis through expression of the
GRLN‑R in pancreatic islets, the liver and, to a much



www.nature.com/nrgastro
© 2012 Macmillan Publishers Limited. All rights reserved
REVIEWS
lesser extent, muscle. 40 Consistent with these find­
ings, ghrelin has been shown to both inhibit glucose-­
stimulated insulin secretion and decrease insulin
sensitivity in normal mice, with improved glucose toler­
ance found in knockout animals.34,36 Administration of a
GOAT inhibitor also enhances glucose tolerance in mice,
in a ghrelin-­ ependent manner.35 However, the glycae­
d
mic actions of ghrelin might also be mediated through
its effects as a growth hormone secretagogue, as GOAT
knockout mice subjected to severe caloric restriction
experience lethal hypoglycaemia, even in the setting of
profoundly reduced insulin levels, and ghrelin replace­
ment prevents the mortality through increased growth
hormone levels.41 Together, these findings suggest the
existence of a metabolic negative feedback loop that
regulates circulating ghrelin levels, probably through
modulation of blood glucose levels.
Given the important role of ghrelin in stimulat­
ing appetite, it is not surprising that ghrelin levels are
decreased in obese individuals, consistent with negative
feedback regulation under conditions of positive energy
balance. 42–44 However, concentrations of circulating
ghrelin are reduced even further by insulin resistance
that is associated with either obesity or type 2 diabe­
tes mellitus.43,45 These in vivo data are in keeping with
reports that ghrelin release by primary X/A-like cells
is inhibited by physiological concentrations of insulin
in the setting of low glucose levels,25,26 but not with the
finding that insulin resistance in these cells prevents
insulin-induced suppression of ghrelin.26 Furthermore,
the suppression of endogenous ghrelin levels in all
of these conditions raises the question as to whether
antagonism of the ghrelin system will provide additional
benefit to body weight regulation in any of these condi­
tions. Collectively, therefore, it remains to be established
whether manipulation of ghrelin signalling will be of
clinical benefit in conditions characterized by obesity.
No strong link has been found between body weight
dysregulation and polymorphisms in the ghrelin–
GRLN‑R axis.46 However, increased ghrelin levels have
been reported in individuals with anorexia, which sug­
gests a role for ghrelin resistance in this condition.47 A
number of clinical trials have now examined the effects
of ghrelin administration in a variety of conditions char­
acterized by reduced food intake due to cachexia; to date,
results from these trials have been generally positive.48
Conversely, ghrelin receptor antagonists have been pro­
posed for use in obesity,49 with a reduction in body weight
associated with decreased food intake demonstrated in
diet-induced obese mice; enhanced glycaemic control
during an intraperitoneal glucose tolerance test was
also noted in these animals.32 Similar findings have been
reported for a GOAT inhibitor.35 However, results from
clinical trials using either approach are still pending.

The L cell
In contrast to ghrelin, peptides synthesized by the intes­
tinal L cell have the following characteristics: first, they
are produced predominantly in the distal small intestine
(that is, the ileum) and colon; second, they are found

Norepinephrine

Glucose

Insulin

IR

β1R

GLUT1/4/5

PI3K

cAMP

Glucokinase or
hexokinase

AKT

??

Low
concentrations
of glucose

High
concentrations
of glucose

PKA
Ghrelin

Figure 1 | The X/A-like cell. Under fasting conditions (for
example, low levels of insulin and glucose), the X/A-like cell
is stimulated by the sympathetic nervous system through
release of norepinephrine and activation of the β1R, which is
known to be coupled to cAMP and PKA signalling. Low
concentrations of glucose also directly stimulate ghrelin
release, at least in vitro. Under fed conditions (for example,
high levels of insulin and glucose), insulin inhibits ghrelin
release through the canonical PI3K/Akt pathway, whereas
glucose inhibits secretion through a pathway that might
involve glucose transporters and glucokinase or hexokinase.
Abbreviations: β1R, β1 adrenergic receptor; GLUT1/4/5,
glucose transporter 1, 4 or 5; IR, insulin receptor; PI3K,
phosphatidylinositol‑3 kinase‑1; PKA, protein kinase A.

only in open-type L cells; third, their levels are increased
by feeding; and fourth, they are anorexigenic.
GLP‑1, GLP‑2 and oxyntomodulin are synthesized by
PC1/3-mediated cleavage of proglucagon, and are cosecreted by the L cell.50–52 Although glicentin and two
intervening peptides are also liberated from proglucagon
in the L cell, no physiological actions have been delin­
eated for these hormones and we do not discuss them
further in this Review. The highest density of L cells is
in the distal gut, but they are also localized in the upper
gastro­ ntestinal tract of rats and humans, although
i
in markedly lower numbers than in the distal gut.53,54
Interestingly, findings have indicated that ~20% percent
of proximal L cells co-express GIP, and 40% express
cholecystokinin.11,55 By contrast, GIP and cholecysto­
kinin expression by distal L cells is fairly low, but ~45%
of the ileal and ~70% of the colonic L cells co-express
PYY, the functions of which are described below. 11,55
Furthermore, a study has indicated that the phenotype
of the colonic L cell is altered by abnormal carbohydrate
flux to this tissue, such that the cell increases its capacity
to respond to the presence of these luminal nutrients.56
Compared with proximal L cells, colonic L cells might

NATURE REVIEWS | GASTROENTEROLOGY  HEPATOLOGY 	
© 2012 Macmillan Publishers Limited. All rights reserved

VOLUME 9  |  DECEMBER 2012  |  707
REVIEWS

Insulin

Acetylcholine

Leptin

IR

M1R

LR

PI3K

MEK

AKT

ERK1/2

STAT3

GLP1/2
Oxyntomodulin
PYY

Ca2+

PKCζ

PKA/PKC

GPRs

Fatty acids
Oleoylethanoloamide
2-Oleoylglycerol

∆ψ

FATP4

Oleic acid

SGLT1

Glucose Na+

Figure 2 | The L cell. In the fed state, secretion by the L cell
is rapidly stimulated by the vagus, through release of
acetylcholine and activation of M1R. After passage down the
gastrointestinal tract, luminal nutrients then enhance
secretion through a variety of pathways: GPRs for long and
short-chain fatty acids, oleoylethanoloamide and
2‑oleoylglycerol (leading to activation of PKA or PKC); FATP4
for oleic acid (leading to PKCζ signalling); and SGLT1 for
sodium and glucose (resulting in depolarization (Ψ)-induced
calcium signalling). Insulin and leptin also enhance secretion
by the intestinal L cell in a positive-feedback fashion, through
canonical signalling pathways. Abbreviations: ERK1/2,
extracellular signal-regulated kinase 1/2; FATP4, fatty acid
transport protein 4; GLP‑1/2, glucagon-like peptide 1 or 2;
GPRs, G protein-coupled receptors; IR, insulin receptor; LR;
leptin receptor isoform b; M1R, the M1 muscarinic receptor;
MEK, MAPK/ERK kinase; PI3K, phosphatidylinositol‑3
kinase‑1; PKA, protein kinase A; PKC, protein kinase C; PYY,
peptide YY; SGLT1, sodium-glucose transporter 1; STAT3,
signal transducer activator of transcription 3.

also respond preferentially to short-chain fatty acids,57
which is consistent with the high density of gut bacteria
in this organ. The physiological importance of the differ­
ences between the proximal and distal L cell populations,
as well as their developmental aetiology, clearly requires
further examination.
Hormone secretion by the L cell is stimulated by nutri­
ent ingestion; plasma levels of these hormones exhibit an
early rise followed by a prolonged plateau phase.58 Studies
708  |  DECEMBER 2012  |  VOLUME 9

in rodents have demonstrated that the early L cell response
is mediated indirectly by proximal activation of the vagus
nerve, leading to muscarinic receptor signalling in the
distal L cell; some evidence exists for a similar pathway
in humans (Figure 2).4,59,60 By contrast, the prolonged
secretory phase is triggered by nutrients in the gut lumen
that act directly on ileal and/or colonic L cells through
a variety of mechanisms, including the following: sig­
nalling coupled through transporters (for example, for
glucose: sodium–glucose transporter‑1 and, to a lesser
extent, KATP channels, leading to Na+ and K+-induced
depolarization, respectively; and for oleic acid: fatty acid
transport protein 4 and atypical protein kinase C‑ζ);7,61–65
or G‑protein-coupled receptor signalling (for example, for
short-chain fatty acids: GRP40 and GRP43; for long-chain
fatty acids: GPR40 and GPR120; for oleoylethanol­ mide
a
and 2‑oleoylglycerol: GPR119; and for glucose: sweet taste
receptors). The bile acid receptor, TGR5, is also expressed
by the distal L cell.56,57,66–70 Thus, luminal nutrient compo­
sition has a key role in the stimulation of L cell secretion,
with diets enriched in oleic acid, for example, increasing
hormone release to a greater extent than diets high in
saturated fatty acids.71,72 Although L cell secretion is also
known to be modulated by a number of hormones and
neuropeptides (for example, cholecysto­ inin, gastrink
releasing peptide, GIP, insulin, leptin and somatostatin),73
the physiological role of many of these factors and how
their actions are coupled to nutrient ingestion remain to
be fully elucidated. Finally, just as the X/A-like cell shares
many regulatory features with the pancreatic A cell, so too
is hormone secretion by the L cell similar to that of islet
β cells, particularly with respect to the pathways regulating
glucose and fatty acid responses.74 Not only is hormone
secretion by the gut and pancreatic islets thereby coor­
dinated in the fed state, but many of these pathways are
currently under investigation as coordinated L and β‑cell
secretagogues, as discussed in more detail below.
Both insulin and leptin also serve as GLP‑1 secreta­
gogues, with the effects of insulin seeming to form
a positive feedback loop between the L cell and islet
β cell.75,76 The concomitant findings that conditions of
leptin and insulin resistance impair the L cell response to
oral glucose are consistent with reports of reduced GLP‑1
levels in individuals with obesity and insulin resistance
and/or type 2 diabetes mellitus, respectively,77–79 and indi­
cate that, like the X/A-like cell, the L cell is dysregulated
by perturbations in nutrient homeostasis.
Once released into the circulation, GLP‑1 and GLP‑2
are cleaved by dipeptidylpeptidase 4 (DPP‑4), render­
ing short bioactive half-lives of ~1–2 min and ~7 min,
respectively. 53,80,81 Oxyntomodulin has intermediate
susceptibility to degradation by DPP‑4, at least in vitro,
and a DPP‑4-resistant analogue has been reported to
demon­ trate enhanced bioactivity compared with the
s
native peptide in vivo.82,83 In the case of both GLP‑1 and
GLP‑2, the cleavage product serves as a partial agonist or
antagonist at the respective receptor,84,85 although trun­
cated GLP‑1 also seems to mediate some of the cardio­
protective actions of GLP‑1 through a yet-to-be-identified
receptor.86 Finally, PYY is also degraded by DPP‑4,87,88



www.nature.com/nrgastro
© 2012 Macmillan Publishers Limited. All rights reserved
REVIEWS
with the half-lives of both PYY1–36NH2 and PYY3–36NH2 being
~13 min,89 but, in contrast to the inactivation of GLP‑1,
GLP‑2 and oxynto­ odulin, DPP‑4-mediated cleavage of
m
PYY is actually required for its biological activity at the
Y2 receptor.90 Thus, the truncated peptide, PYY3–36NH2,
mediates the effects of PYY as a satiety factor.87
Together, therefore, GLP‑1, GLP‑2, oxyntomodulin
and PYY are all released by the intestinal L cell. However,
these L cell hormones have distinct profiles of biological
actions, which ultimately integrate at the level of whole
body nutrient homeostasis.

GLP‑1
As a result of the widespread distribution of the GLP‑1
receptor, in both peripheral tissues and the CNS,91–93 cir­
culating GLP‑1 exerts a diverse array of effects on nutrient
homeostasis. The major biological actions of GLP‑1 are
antidiabetic, and are most notably exerted through the
pancreatic β cell. Hence, GLP‑1 is an incretin that enhances
glucose-dependent insulin secretion, as well as stimulat­
ing insulin biosynthesis and, in rodents, promoting β‑cell
proliferation while inhibiting apoptosis.94,95 GLP‑1 also
inhibits release of glucagon by pancreatic α cells, delays
nutrient absorption through inhibition of gastric emptying
and intestinal motility, and enhances insulin sensitivity at
the level of both the liver and peripheral tissues—although
the majority of its effects on glycaemia seem to be medi­
ated through changes in islet hormone secretion.96 The
beneficial effects of circulating endogenous and exogenous
GLP‑1 or its analogues on regulation of glucose have also
been confirmed in studies using both antagonist and
receptor knockout approaches.97,98 However, peripherally
administered GLP‑1 and its analogues also have anorexi­
genic actions, inducing satiety and inhibiting food intake
in normal and obese rodents and humans, as well as in
patients with type 2 diabetes mellitus.99–102
As a consequence of the reported biological actions
of GLP‑1, several long-acting, DPP‑4-resistant GLP‑1based drugs have now been licensed for clinical use in the
treatment of type 2 diabetes mellitus, the major effects
of which are a reduction in haemoglobin A1c (HbA1c, a
glycated haemoglobin) levels in association with weight
loss.103 Of note, although GLP‑1R agonists do induce
weight loss in overweight and obese individuals with
and without type 2 diabetes mellitus, the overall effect
is fairly modest, averaging 2.9 kg.104 However, a GLP‑1–
glucagon co-agonist that takes advantage of the inhibi­
tory effects of GLP‑1 on food intake and the stimulatory
effects of glucagon on energy expenditure, while at the
same time balancing the hypoglycaemic and hyper­
glycaemic effects of the two peptides, respectively, indi­
cates that novel approaches to the use of GLP‑1 could
optimize its use in obesity. 105 Conversely, although
DPP‑4 inhibitors are used clinically to increase circulat­
ing levels of endo­ enous GLP‑1, as well as of the other
g
major incretin, GIP, in patients with type 2 diabetes mel­
litus,103 the use of these agents is associated with reduced
levels of HbA1c but not weight loss, the latter possibly
owing to concomitantly reduced production of bioactive
PYY3–36NH2, as discussed above. An alternative approach

to the use of GLP‑1 in the treatment of patients with
type 2 diabetes mellitus and/or obesity is enhancement
of endogenous GLP‑1 secretion by activation of novel
L cell secretory pathways and, in particular, those medi­
ating the responses to fatty acids, including GPR40 and
GPR119.74,106 Such agents would have the advantage of
concomitantly increasing the release of all of the L cell
hormones, of which oxynto­ odulin and PYY3–36NH2 are
m
also anorexigenic (discussed in more detail below).
Although GLP‑1 is released into the circulation as a
hormone, its short half-life suggests that at least some of its
physiological actions might be mediated indirectly, prob­
ably through the GLP‑1R expressed on afferent neurons.
Hence, despite the well-established direct effects of GLP‑1
on β cells in vitro,107,108 the in vivo actions of GLP‑1 to
stimulate insulin release have been reported to require
afferent neurons.92 Indeed, GLP‑1 increases vagal afferent
firing in rats.109 However, in direct comparative studies,
the requirement for sensory afferents in the actions of
GLP‑1 on both insulin release and food intake in mice
was observed at low, but not high, doses of GLP‑1.110,111
Whether some of the anorexigenic actions of GLP‑1 are
also mediated via suppression of gastric emptying remains
unclear.112 Nonetheless, these findings collectively suggest
that the physiological effects of endogenously secreted
GLP‑1, which includes the actions of GLP‑1 in patients
treated with DPP‑4 inhibitors, are mediated indirectly by
local effects of GLP‑1 on its receptor expressed by gastro­
intestinal and/or vagal afferent neurons.113 By contrast, the
pharmacologic actions of GLP‑1 and long-acting GLP‑1R
agonists seem to be exerted, at least in part, through direct
effects on the GLP‑1R in β cells and the brain.
Genetic studies have demonstrated that poly­
morphisms in the voltage-gated K + channel gene,
KCNQ1, are associated with reduced secretion of GLP‑1,
whereas variants in the genes for the GLP‑1R, the β‑cell
transcription factor, TCF7L2, and the neuroendocrine
transmembrane protein, Wolfram syndrome 1, lead to
reduced GLP‑1-stimulated insulin release.114–117 Hence,
inter­ ndividual differences in GLP‑1 release or func­
i
tion might be linked to alterations in the effectiveness of
DPP‑4 inhibitors and GLP‑1R agonists, respectively, in
patients with type 2 diabetes mellitus, a notion that has
yet to be rigorously assessed.

GLP‑2
In contrast to GLP‑1, the actions of GLP‑2 are more
restricted owing to the highly concentrated expression of
the GLP‑2R in the gastrointestinal tract; these receptors
are expressed in only a few other tissues, including the
vagus nerve and brain.118,119 GLP‑2 is, therefore, known
foremost for its effects in the gastrointestinal tract. Both
GLP‑2 and its degradation-resistant analogues increase
small and large intestinal weight, crypt-villus height
and mucosal surface area through stimulation of crypt
cell proliferation and inhibition of epithelial cell apop­
tosis.120–122 GLP‑2 also enhances nutrient absorption
through various actions, including increasing expression
of digestive enzymes and nutrient transporters, as well as
stimulating intestinal blood flow.120–122 Of note, although

NATURE REVIEWS | GASTROENTEROLOGY  HEPATOLOGY 	
© 2012 Macmillan Publishers Limited. All rights reserved

VOLUME 9  |  DECEMBER 2012  |  709
REVIEWS
GLP‑2 also inhibits gastric emptying, these effects are
minor compared with the actions of GLP‑1 on gastric
emptying.123 Furthermore, in a wide variety of animal
models of intestinal damage and insufficiency, GLP‑2
exerts protective effects on the gastrointestinal epithelium
by reducing inflammation and increasing barrier func­
tion, as well as by preventing and repairing damage.124–127
However, in contrast to the profound pharmacological
actions of GLP‑2, the role of the endogenous hormone
seems to be more limited, with only a small or no reduc­
tion in bowel size noted upon administration of a GLP‑2R
antagonist and in GLP‑2R null mice.128,129 Nonetheless,
endogenous GLP‑2 is required for the intestinal adaptive
responses to nutrient ingestion after a prolonged fast, as
well as for protection from chemically induced enteri­
tis.85,130 Importantly, the majority of the major findings
on GLP‑2 in rodent models have also been confirmed in
humans, most notably in patients with intestinal insuffi­
ciency owing to short bowel syndrome or damage associ­
ated with Crohn’s disease.131,132 Clinical trials are ongoing,
and a long-acting analogue of GLP‑2 has been approved in
Europe for use in patients with short bowel syndrome.133
Despite extensive characterization of the effects of
GLP‑2, the mechanistic basis of the action of GLP‑2 is only
poorly understood. The GLP‑2R is not localized to the
major site of action of GLP‑2 (that is, intestinal epithelial
cells) but rather it is localized to the intestinal subepithelial
myofibroblasts, enteric nervous system and diffuse entero­
endocrine cells;122 the actions of GLP‑2 must therefore be
exerted indirectly by the release of paracrine factors from
these cells. Downstream players that have been identified
to date include: insulin-like growth factor (IGF)‑1 and
IGF‑2, and the ErbB network for crypt cell proliferation in
the small and large intestine;130,134–136 keratinocyte growth
factor, for large bowel growth;137 vasoactive intestinal poly­
peptide, for anti-inflammatory actions in the small and
large intestine;138 and endothelial nitric oxide synthase, for
intestinal blood flow.139 Finally, and again in contrast to
the actions of GLP‑1, the known effects of GLP‑2 do not
seem to be mediated through afferent pathways, as activa­
tion of this receptor is not linked to any change in vagal
firing, and centrally administered but not peripherally
administered GLP‑2 inhibits food intake.109,140,141

Oxyntomodulin
Although oxyntomodulin was originally named for its
inhibitory effects on gastric acid secretion,142 these actions
are relatively weak compared with those exerted by other
gut peptides, such as somatostatin.143 Hence, the biologi­
cal role of oxyntomodulin remained somewhat uncer­
tain until ~10 years ago, when it was reported to inhibit
food intake in both rats and humans when administered
exogenously.144,145 Indeed, a number of studies have now
demonstrated that repeated peripheral administration
of either oxyntomodulin or its long-acting analogues
induces weight loss in obese rats, mice and humans, with
this effect being mediated through both suppression of
food intake and increased energy expenditure.146–148
Interestingly, early studies on oxyntomodulin and
gastric acid secretion suggested the existence of a receptor
710  |  DECEMBER 2012  |  VOLUME 9

for this hormone through which glucagon might also
act, although with less potency.142 Although no specific
oxyntomodulin receptor has been identified to date, this
hormone has been established to be an agonist for both
the GLP‑1R and the glucagon receptor. Hence, oxynto­
modulin has EC50s (half maximal effective concentra­
tions) for the GLP‑1R and glucagon receptor that are
approximately 100-fold and 10-fold greater than those
of GLP‑1 and glucagon, respectively.144,149,150 Consistent
with these findings, the actions of oxyntomodulin on the
gastric glands are now known to be mediated through
GLP‑1R, as are the effects of peripheral oxyntomodulin
to suppress food intake.151,152 However, the stimulatory
effect of oxyntomodulin on energy expenditure might
also be dependent upon the glucagon receptor, as this
is also a well-established effect of glucagon.153,154 Hence,
not only DPP‑4-resistant analogues of oxyntomodulin,
but also dual GLP‑1R/glucagon receptor oxynto­modulin
analogues have been developed as possible thera­ eutic
p
agents for the treatment of obesity. 83,148,155 Notably,
oxynto­modulin also improves glycaemic control, through
GLP‑1R-dependent increase of glucose-stimulated insulin
secretion.156 Although this effect might be expected to be
counter-balanced by oxyntomodulin-induced activation
of the glucagon receptor, the available data suggest that
the anti-hyperglycaemic effects of oxyntomodulin prevail
in the setting of dual receptor activation.153 Furthermore,
the mechanism of action of oxyntomodulin also implies
that polymorphisms in either the GLP-1R, the glucagon
receptor and/or their signalling pathways might influence
the effectiveness of these drugs.115–117,157
Finally, the cellular pathway(s) by which oxynto­
modulin inhibits food intake are thought to include
both the CNS and the vagus nerve. Peripheral admin­
istration of oxynto­ odulin activates a variety of central
m
nuclei involved in feeding, including the arcuate nucleus
that abuts areas of the hypothalamus characterized by
a leaky blood–brain barrier.150,152 Oxyntomodulin also
stimulates release of the anorexigenic neuropeptide,
α‑melanocyte-stimulating hormone, from hypothalamic
explants in vitro, which is consistent with possible direct
effects on central feeding centers.152 However, activation
of the nucleus of the solitary tract by peripheral oxynto­
modulin is also in keeping with a role for vagal sensing
of this hormone.150 Although studies of oxyntomodulin
actions on food intake following vagal disruption have
yet to be conducted, the inhibitory actions of oxynto­
modulin on pancreatic exocrine secretion are abrogated
in vagotomised rats, which indicates that signalling can
occur through this peripheral neural pathway.158

PYY
Which prohormone convertase cleaves proPYY to
PYY1–36NH2 is not known, although it is presumed to be
PC1/3 owing to the known actions of this enzyme on pro­
glucagon in the intestinal L cell.50 PYY1–36NH2 was first iden­
tified as a gut peptide with tyrosine (‘Y’) at its N‑terminal
and C‑terminal ends, and was initially characterized as
an inhibitor of both pancreatic exocrine secretion and
intestinal motility.159,160 However, although PYY1–36NH2



www.nature.com/nrgastro
© 2012 Macmillan Publishers Limited. All rights reserved
REVIEWS
binds to multiple receptors of the neuropeptide Y recep­
tor (YR) family, its cleavage by DPP‑4 in the circula­
tion converts it to a Y2-receptor-selective agonist.87,90
Thus, compared with both full-length PYY 1–36NH2
and neuropeptide Y, which stimulate food intake through
the Y1R and possibly the Y5R, the Y2R-selectivity of
PYY3–36NH2 and its agonists enables suppression of food
intake through inhibition of neuropeptide Y release in the
arcuate nucleus, while Y2R specific antagonists prevent
the anorexigenic actions of PYY3–36NH2.161–164 Consistent
with these findings, initial studies in mice have suggested
that the actions of PYY3–36NH2 are not mediated by periph­
eral sensory afferent nerves such as the vagus.111 However,
in the rat, the Y2R is expressed by the vagus nerve, and
peripherally administered PYY3–36NH2 stimulates gastric
vagal afferent firing, while vagotomy prevents the anorexi­
genic actions of PYY3–36NH2.165 Thus, the mechanism of
action of PYY3–36NH2 might exhibit species-­ pecific dif­
s
ferences. Alternatively, as the Y2R expressed by rat vagal
afferent neurons is regulated by cholecystokinin in a nutri­
ent-dependent manner,166 experimental differences could
also account for these apparent discrepancies.
Although great interest was generated by the first
reports of the anorexigenic actions of PYY3–36NH2 in
rodents as well as normal-weight and obese humans,163,167
these findings have been the subject of some controversy
in the literature, with both confirmatory and negative
findings reported.111,168,169 The reason for these discrepant
findings remains elusive. Nonetheless, both a rare PYY
mutation and Y2R variants have been linked to altera­
tions in body weight in humans.170–172 Hence, long-acting
PYY3–36NH2 analogues, as well as Y2R agonists, are cur­
rently under development for the treatment of obesity,
and many clinical trials are ongoing.

Combined effects
Although the L cell hormones are co-secreted, few studies
have examined the combined effects of simultaneous
increased levels of these hormones. With respect to feed­
back suppression of food intake, additive and/or syner­
gistic effects of PYY3–36NH2 with oxyntomodulin, as well
as of PYY3–36NH2 with GLP‑1 or its analogues have been
reported.111,173–175 Furthermore, PYY3–36NH2 and GLP‑1 also
have additive effects to delay gastric emptying.176 Finally,
GLP‑1 and GLP‑2 do not demonstrate additive effects
to suppress food intake,177 but do exert opposing effects
on glucagon release, with the inhibitory effect of GLP‑1
predominating.178 No studies to date have examined the
physiological consequences of increasing the levels of all
four of these hormones, or of their effects when given in
combination with all of the other gut hormones released
by nutrient intake. Such investigations will clearly be nec­
essary to fully understand the integrated contribution of
the L cell to nutrient homeostasis, particularly if L cell
secretagogues are to be used for the treatment of type 2
diabetes mellitus and/or obesity, as discussed above.

Clinical relevance
Two successful, surgical treatments for obesity—Rouxen‑Y gastric bypass (RYGB) and sleeve gastrectomy—also

improve or reverse the symptoms of type 2 diabetes mel­
litus. RYGB markedly reduces the size of the stomach
and diverts ingested nutrients and pancreato-biliary flow
directly to the distal small intestine, whereas sleeve gas­
trectomy introduces gastric restriction only.179 Despite
the differences between these surgical interventions,
the results of clinical trials indicate that both proce­
dures result in equivalent improvements in glycaemic
control and identical weight loss 1 year after surgery.180,181
Remarkably, the reductions in glucose levels occur
rapidly after surgery, often prior to the changes in body
weight.182 The success of RYGB for both resolution of
type 2 diabetes mellitus and weight loss has largely been
attributed to an increase in the release of peptides from
the L cells, with the effects to reduce ghrelin release being
controversial.181,183–187 The concomitant diversion of bile
flow to the distal small intestine might also contribute to
the increase in L cell secretion following RYGB through
activation of the L cell bile acid receptor, TGR5.69 Indeed,
a study has demonstrated that enhanced release of GLP‑1
following gastric bypass in obese rats is responsible for
the weight-loss-independent improvement in glucose
tolerance associated with this procedure.188 By contrast,
sleeve gastrectomy has been reported to cause greater
decreases in ghrelin levels and increased cholecystokinin
release than found with RYGB, in addition to increasing
L cell secretion.181 Thus, although differing somewhat in
their mechanisms of action, RYGB and sleeve gastrec­
tomy are unique approaches that manipulate the physio­
logical actions of the gut hormones to regulate nutrient
homeostasis through their combined effects on gut
nutrient handling, glucose tolerance and food intake.

Conclusion
This Review has focused on the hormones produced
by the enteroendocrine X/A-like cell (ghrelin) and the
L cell (GLP‑1, GLP‑2, oxyntomodulin and PYY). These
peptides have key roles in the integration of food intake
with metabolism, as well as providing feedback signals to
the brain to induce satiety during feeding. However, it is
recognized that the gastrointestinal tract releases many
other hormones that also have important roles in nutri­
ent homeostasis including, most notably, GIP and chol­
ecystokinin. Elucidation of the combined actions of all of
these hormones will be essential to develop a comprehen­
sive understanding of the integrative role that the gastro­
intestinal tract has in maintaining nutrient homeostasis
during fasting and in response to feeding.

Review criteria
PubMed was searched for full-length articles using the
keywords “ghrelin”, “GLP‑1”, “GLP‑2”, “oxyntomodulin”,
“enteroglucagon” and “PYY”, alone and in combination
with key author names and/or the keywords “secretion”,
“food intake”, “obesity”, “insulin resistance”, “type 2
diabetes” and/or “review”; review articles were used to
further search for original papers. All years were included
in the search. Articles published in languages other than
English were excluded from the analysis.

NATURE REVIEWS | GASTROENTEROLOGY  HEPATOLOGY 	
© 2012 Macmillan Publishers Limited. All rights reserved

VOLUME 9  |  DECEMBER 2012  |  711
REVIEWS
1.	

2.	

3.	

4.	

5.	

6.	
7.	

8.	
9.	

10.	

11.	

12.	

13.	

14.	

15.	

16.	

17.	

18.	

19.	

20.	

21.	

Rehfeld, J. F. A centenary of gastrointestinal
endocrinology. Horm. Metab. Res. 36, 735–741
(2004).
Ferri, G.‑L. et al. Tissue localization and relative
distribution of regulatory peptides in separated
layers from the human bowel. Gastroenterology
84, 777–786 (1983).
Larsson, L. I. Peptide secretory pathways in GI
tract: cytochemical contributions to regulatory
physiology of the gut. Am. J. Physiol. 239,
G237–G246 (1980).
Anini, Y., Hansotia, T.  Brubaker, P Muscarinic
. L.
receptors control postprandial release of
glucagon-like peptide‑1: in vivo and in vitro
studies in rats. Endocrinology 143, 2420–2426
(2002).
Bohorquez, D. V.  Liddle, R. A. Axon-like basal
processes in enteroendocrine cells:
characteristics and potential targets. Clin. Transl.
Sci. 4, 387–391 (2011).
Dockray, G. J. The versatility of the vagus.
Physiol. Behav. 97, 531–536 (2009).
Reimann, F., Tolhurst, G.  Gribble, F. M.
G‑protein‑coupled receptors in intestinal
chemosensation. Cell Metab. 15, 421–431
(2012).
Dockray, G. J. Cholecystokinin. Curr. Opin.
Endocrinol. Diabetes Obes. 19, 8–12 (2012).
Cho, Y. M.  Kieffer, T. J. K‑cells and glucosedependent insulinotropic polypeptide in health
and disease. Vitam. Horm. 84, 111–150 (2010).
Ali, S., Lamont, B. J., Charron, M. J. 
Drucker, D. J. Dual elimination of the glucagon
and GLP‑1 receptors in mice reveals plasticity in
the incretin axis. J. Clin. Invest. 121, 1917–1929
(2011).
Habib, A. M. et al. Overlap of endocrine hormone
expression in the mouse intestine revealed by
transcriptional profiling and flow cytometry.
Endocrinology 153, 3054–3065 (2012).
Kojima, M. et al. Ghrelin is a
growth‑hormone‑releasing acylated peptide from
stomach. Nature 402, 656–660 (1999).
Date, Y. et al. Ghrelin, a novel growth hormonereleasing acylated peptide, is synthesized in a
distinct endocrine cell type in the
gastrointestinal tracts of rats and humans.
Endocrinology 141, 4255–4261 (2000).
Rindi, G. et al. Characterisation of gastric ghrelin
cells in man and other mammals: studies in
adult and fetal tissues. Histochem. Cell Biol.
117, 511–519 (2002).
Sakata, I. et al. Ghrelin-producing cells exist as
two types of cells, closed- and opened-type cells,
in the rat gastrointestinal tract. Peptides 23,
531–536 (2002).
Zhu, X., Cao, Y., Voogd, K.  Steiner, D. F. On the
processing of proghrelin to ghrelin. J. Biol. Chem.
281, 38867–38870 (2006).
Kirchner, H. et al. GOAT links dietary lipids with
the endocrine control of energy balance. Nat.
Med. 15, 741–745 (2009).
Tschop, M., Smiley, D. L.  Heiman, M. L. Ghrelin
induces adiposity in rodents. Nature 407,
908–913 (2000).
Cummings, D. E. et al. A preprandial rise in
plasma ghrelin levels suggests a role in meal
initiation in humans. Diabetes 50, 1714–1719
(2001).
Mundinger, T. O., Cummings, D. E. 
Taborsky, G. J. Jr. Direct stimulation of ghrelin
secretion by sympathetic nerves. Endocrinology
147, 2893–2901 (2006).
Ao, Y. et al. Brainstem thyrotropin-releasing
hormone regulates food intake through vagaldependent cholinergic stimulation of ghrelin
secretion. Endocrinology 147, 6004–6010
(2006).

712  |  DECEMBER 2012  |  VOLUME 9

22.	 Shimada, M. et al. Somatostatin suppresses
ghrelin secretion from the rat stomach. Biochem.
Biophys. Res. Commun. 302, 520–525 (2003).
23.	 McCowen, K. C., Maykel, J. A., Bistrian, B. R. 
Ling, P
. R. Circulating ghrelin concentrations are
lowered by intravenous glucose or
hyperinsulinemic euglycemic conditions in
rodents. J. Endocrinol. 175, R7–R11 (2002).
24.	 Saad, M. F. et al. Insulin regulates plasma
ghrelin concentration. J. Clin. Endocrinol. Metab.
87, 3997–4000 (2002).
25.	 Sakata, I. et al. Glucose-mediated control of
ghrelin release from primary cultures of gastric
mucosal cells. Am. J. Physiol. Endocrinol. Metab.
302, E1300–E1310 (2012).
26.	 Gagnon, J.  Anini, Y. Insulin and norepinephrine
regulate ghrelin secretion from a rat primary
stomach cell culture. Endocrinology 153,
3646–3656 (2012).
27.	 Taborsky, G. J. Jr. The physiology of glucagon.
J. Diabetes Sci. Technol. 4, 1338–1344 (2010).
28.	 Walker, J. N. et al. Regulation of glucagon
secretion by glucose: paracrine, intrinsic or
both? Diabetes Obes. Metab. 13 (Suppl. 1),
95–105 (2011).
29.	 Kordowich, S., Collombat, P Mansouri, A. 
.,
Serup, P Arx and Nkx2.2 compound deficiency
.
redirects pancreatic alpha- and beta-cell
differentiation to a somatostatin/ghrelin coexpressing cell lineage. BMC Dev. Biol. 11,
52–67 (2011).
30.	 Ariyasu, H. et al. A postweaning reduction in
circulating ghrelin temporarily alters growth
hormone (GH) responsiveness to GH‑releasing
hormone in male mice but does not affect
somatic growth. Endocrinology 151, 1743–1750
(2010).
31.	 Sakata, I. et al. Characterization of a novel
ghrelin cell reporter mouse. Regul. Pept. 155,
91–98 (2009).
32.	 Esler, W. P et al. Small-molecule ghrelin receptor
.
antagonists improve glucose tolerance,
suppress appetite, and promote weight loss.
Endocrinology 148, 5175–5185 (2007).
33.	 Pfluger, P et al. Simultaneous deletion of
. T.
ghrelin and its receptor increases motor activity
and energy expenditure. Am. J. Physiol.
Gastrointest. Liver Physiol. 294, G610–G618
(2008).
34.	 Kirchner, H., Tong, J., Tschop, M. H. 
Pfluger, P Ghrelin and PYY in the regulation of
. T.
energy balance and metabolism: lessons from
mouse mutants. Am. J. Physiol. Endocrinol.
Metab. 298, E909–E919 (2010).
35.	 Barnett, B. P et al. Glucose and weight control in
.
mice with a designed ghrelin O‑acyltransferase
inhibitor. Science 330, 1689–1692 (2010).
36.	 Stengel, A.  Tache, Y. Ghrelin—a pleiotropic
hormone secreted from endocrine x/a-like cells
of the stomach. Front. Neurosci. 6, 24 (2012).
37.	 Banks, W. A., Tschop, M., Robinson, S. M. 
Heiman, M. L. Extent and direction of ghrelin
transport across the blood–brain barrier is
determined by its unique primary structure.
J. Pharmacol. Exp. Ther. 302, 822–827 (2002).
38.	 Toshinai, K. et al. Ghrelin-induced food intake is
mediated via the orexin pathway. Endocrinology
144, 1506–1512 (2003).
39.	 Date, Y. et al. The role of the gastric afferent
vagal nerve in ghrelin-induced feeding and
growth hormone secretion in rats.
Gastroenterology 123, 1120–1128 (2002).
40.	 Gnanapavan, S. et al. The tissue distribution of
the mRNA of ghrelin and subtypes of its receptor,
GHS‑R, in humans. J. Clin. Endocrinol. Metab. 87,
2988–2991 (2002).
41.	 Zhao, T. J. et al. Ghrelin O‑acyltransferase (GOAT)
is essential for growth hormone-mediated



42.	

43.	

44.	

45.	

46.	

47.	

48.	

49.	

50.	

51.	

52.	

53.	

54.	

55.	

56.	

57.	

58.	

59.	

survival of calorie-restricted mice. Proc. Natl
Acad. Sci. USA 107, 7467–7472 (2010).
Tschop, M. et al. Circulating ghrelin levels are
decreased in human obesity. Diabetes 50,
707–709 (2001).
Shiiya, T. et al. Plasma ghrelin levels in lean and
obese humans and the effect of glucose on
ghrelin secretion. J. Clin. Endocrinol. Metab. 87,
240–244 (2002).
Marzullo, P. et al. The relationship between
active ghrelin levels and human obesity involves
alterations in resting energy expenditure. J. Clin.
Endocrinol. Metab. 89, 936–939 (2004).
McLaughlin, T., Abbasi, F., Lamendola, C.,
Frayo, R. S.  Cummings, D. E. Plasma ghrelin
concentrations are decreased in insulinresistant obese adults relative to equally obese
insulin-sensitive controls. J. Clin. Endocrinol.
Metab. 89, 1630–1635 (2004).
Ukkola, O. et al. Role of ghrelin polymorphisms
in obesity based on three different studies.
Obes. Res. 10, 782–791 (2002).
Otto, B. et al. Weight gain decreases elevated
plasma ghrelin concentrations of patients with
anorexia nervosa. Eur. J. Endocrinol. 145,
669–673 (2001).
Akamizu, T.  Kangawa, K. The physiological
significance and potential clinical applications
of ghrelin. Eur. J. Intern. Med. 23, 197–202
(2012).
Patterson, M., Bloom, S. R.  Gardiner, J. V.
Ghrelin and appetite control in humans-potential application in the treatment of obesity.
Peptides 32, 2290–2294 (2011).
Dhanvantari, S., Seidah, N. G.  Brubaker, P. L.
Role of prohormone convertases in the tissuespecific processing of proglucagon. Mol.
Endocrinol. 10, 342–355 (1996).
Anini, Y. et al. Comparison of the postprandial
release of peptide YY and proglucagon-derived
peptides in the rat. Pflugers Arch. 438,
299–306 (1999).
Orskov, C. et al. Glucagon-like peptides GLP‑1
and GLP‑2, predicted products of the glucagon
gene, are secreted separately from pig small
intestine but not pancreas. Endocrinology 119,
1467–1475 (1986).
Larsson, L.‑I., Holst, J., Hakanson, R. 
Sundler, F. Distribution and properties of
glucagon immunoreactivity in the digestive tract
of various mammals: an immunohistochemical
and immunochemical study. Histochemistry 44,
281–290 (1975).
Eissele, R. et al. Glucagon-like peptide‑1 cells in
the gastrointestinal tract and pancreas of rat,
pig and man. Eur. J. Clin. Invest. 22, 283–291
(1992).
Reimann, F. et al. Glucose sensing in L cells:
a primary cell study. Cell Metab. 8, 532–539
(2008).
Geraedts, M. C. et al. Transformation of postingestive glucose responses after deletion of
sweet taste receptor subunits or gastric bypass
surgery. Am. J. Physiol. Endocrinol. Metab. 303,
E464–E474 (2012).
Tolhurst, G. et al. Short-chain fatty acids
stimulate glucagon-like peptide‑1 secretion via
the G‑protein‑coupled receptor FFAR2. Diabetes
61, 364–371 (2012).
Xiao, Q., Boushey, R., Drucker, D. J. 
Brubaker, P. L. Secretion of the intestinotropic
hormone glucagon-like peptide‑2 is differentially
regulated by nutrients in humans.
Gastroenterology 117, 99–105 (1999).
Anini, Y.  Brubaker, P. L. Muscaranic receptors
control glucagon-like peptide 1 secretion by
human endocrine L cells. Endocrinology 144,
3244–3250 (2003).

www.nature.com/nrgastro
© 2012 Macmillan Publishers Limited. All rights reserved
REVIEWS
60.	 Balks, H. J., Holst, J. J., Von zur Mühlen, A. 
Brabant, G. Rapid oscillations in plasma
glucagon-like peptide‑1 (GLP‑ 1) in humans:
cholinergic control of GLP‑1 secretion via
muscarinic receptors. J. Clin. Endocrinol. Metab.
82, 786–790 (1997).
61.	 Roberge, J. N.  Brubaker, P Secretion of
. L.
proglucagon-derived peptides in response to
intestinal luminal nutrients. Endocrinology 128,
3169–3174 (1991).
62.	 Parker, H. E. Predominant role of active versus
facilitative glucose transport for glucagon-like
peptide-1 secretion. Diabetologia 55,
2445–2455 (2012).
63.	 Gribble, F. M., Williams, L., Simpson, A. K. 
Reimann, F. A novel glucose-sensing mechanism
contributing to glucagon-like peptide‑1 secretion
from the GLUTag cell line. Diabetes 52,
1147–1154 (2003).
64.	 Iakoubov, R., Lauffer, L., Ahmed, A., Bazinet, R. P
.
 Brubaker, P Essential role for protein kinase
. L.
C zeta in oleic-acid induced glucagon-like
peptide‑1 secretion in vivo in the rat.
Endocrinology 152, 1244–1252 (2011).
65.	 Poreba, M. A. et al. Role of fatty acid transport
protein 4 in oleic acid-induced glucagon-like
peptide‑1 secretion. Am. J. Physiol. Endocrinol.
Metab. http://dx.doi.org/10.1152/
ajpendo.00116.2012.
66.	 Hirasawa, A. et al. Free fatty acids regulate gut
incretin glucagon-like peptide‑1 secretion
through GPR120. Nat. Med. 11, 90–94 (2005).
67.	 Edfalk, S., Steneberg, P  Edlund, H. Gpr40 is
.
expressed in enteroendocrine cells and
mediates FFA stimulation of incretin secretion.
Diabetes 57, 2280–2287 (2008).
68.	 Lauffer, L. M., Iakoubov, R.  Brubaker, P
. L.
GPR119 is essential for oleoylethanolamideinduced glucagon-like peptide‑1 secretion from
the intestinal enteroendocrine L‑cell. Diabetes
58, 1058–1066 (2009).
69.	 Thomas, C. et al. TGR5-mediated bile acid
sensing controls glucose homeostasis. Cell
Metab. 10, 167–177 (2009).
70.	 Hansen, K. B. et al. 2‑Oleoyl glycerol is a
GPR119 agonist and signals GLP‑1 release in
humans. J. Clin. Endocrinol. Metab. 96,
E1409–E1417 (2011).
71.	 Rocca, A. S., LaGreca, J., Kalitsky, J. 
Brubaker, P Monounsaturated fatty acids
. L.
improve glycemic tolerance through increased
secretion of glucagon-like peptide‑1.
Endocrinology 142, 1148–1155 (2001).
72.	 Thomsen, C. et al. Differential effects of
saturated and monounsaturated fatty acids on
postprandial lipemia and incretin responses in
healthy subjects. Am. J. Clin. Nutr. 69,
1135–1143 (1999).
73.	 Lim, G. E.  Brubaker, P Glucagon-like
. L.
peptide‑1 secretion by the L cell: a view from the
inside. Diabetes 55, S70–S77 (2006).
74.	 Ahren, B. Islet G protein-coupled receptors as
potential targets for treatment of type 2
diabetes. Nat. Rev. Drug Discov. 8, 369–385
(2009).
75.	 Anini, Y.  Brubaker, P Role of leptin in the
. L.
regulation of glucagon-like peptide‑1 secretion.
Diabetes 52, 252–259 (2003).
76.	 Lim, G. E. et al. Insulin regulates glucagon-like
peptide‑1 secretion from the enteroendocrine
L cell. Endocrinology 150, 580–591 (2009).
77.	 Rask, E. et al. Impaired incretin response after a
mixed meal is associated with insulin resistance
in nondiabetic men. Diabetes Care 24,
1640–1645 (2001).
78.	 Vilsboll, T., Krarup, T., Deacon, C. F., Madsbad, S.
 Holst, J. J. Reduced postprandial
concentrations of intact biologically active

79.	

80.	

81.	

82.	

83.	

84.	

85.	

86.	

87.	

88.	

89.	

90.	

91.	

92.	

93.	

glucagon-like peptide 1 in type 2 diabetic
patients. Diabetes 50, 609–613 (2001).
Mannucci, E. et al. Glucagon-like peptide (GLP)‑1
and leptin concentrations in obese patients with
Type 2 diabetes mellitus. Diabetes Med. 17,
713–719 (2000).
Vilsboll, T., Agerso, H., Krarup, T.  Holst, J. J.
Similar elimination rates of glucagon-like
peptide‑1 in obese type 2 diabetic patients and
healthy subjects. J. Clin. Endocrinol. Metab. 88,
220–224 (2003).
Tavares, W., Drucker, D. J.  Brubaker, P
. L.
Enzymatic- and renal-dependent catabolism of
the intestinotropic hormone glucagon-like
peptide‑2 in rats. Am. J. Physiol. Endocrinol.
Metab. 278, E134–E139 (2000).
Zhu, L. et al. The role of dipeptidyl peptidase IV in
the cleavage of glucagon family peptides: in vivo
metabolism of pituitary adenylate cyclase
activating polypeptide‑(1–38). J. Biol. Chem. 278,
22418–22423 (2003).
Kerr, B. D., Flatt, P  Gault, V. A. (D-Ser2)
. R.
Oxm[mPEG-PAL]: a novel chemically modified
analogue of oxyntomodulin with
antihyperglycaemic, insulinotropic and
anorexigenic actions. Biochem. Pharmacol. 80,
1727–1735 (2010).
Knudsen, L. B.  Pridal, L. Glucagon-like
peptide‑1‑(9–36) amide is a major metabolite of
glucagon-like peptide‑1‑(7–36) amide after in vivo
administration to dogs, and it acts as an
antagonist on the pancreatic receptor. Eur. J.
Pharmacol. 318, 429–435 (1996).
Shin, E. D., Estall, J. L., Izzo, A., Drucker, D. J. 
Brubaker, P Mucosal adaptation to enteral
. L.
nutrients is dependent on the physiologic actions
of glucagon-like peptide‑2 in mice.
Gastroenterology 128, 1340–1353 (2005).
Ban, K. et al. Cardioprotective and vasodilatory
actions of glucagon-like peptide 1 receptor are
mediated through both glucagon-like peptide 1
receptor-dependent and -independent pathways.
Circulation 117, 2340–2350 (2008).
Medeiros, M. D.  Turner, A. J. Processing and
metabolism of peptide-YY: pivotal roles of
dipeptidylpeptidase-IV, aminopeptidase‑P and
,
endopeptidase‑24.11. Endocrinology 134,
2088–2094 (1994).
Aaboe, K. et al. Twelve weeks treatment with the
DPP‑4 inhibitor, sitagliptin, prevents degradation
of peptide YY and improves glucose and nonglucose induced insulin secretion in patients
with type 2 diabetes mellitus. Diabetes Obes.
Metab. 12, 323–333 (2010).
Yoshinaga, K. et al. Structural requirements of
peptide YY for biological activity at enteric sites.
Am. J. Physiol. 263, G695–G701 (1992).
Dumont, Y., Fournier, A., St-Pierre, S.  Quirion, R.
Characterization of neuropeptide Y binding sites
in rat brain membrane preparations using [125I]
[Leu31, Pro34]peptide YY and [125I]peptide
YY3–36 as selective Y1 and Y2 radioligands.
J. Pharmacol. Exp. Ther. 272, 673–680 (1995).
Campos, R. V., Lee, Y. C.  Drucker, D. J.
Divergent tissue-specific and developmental
expression of receptors for glucagon and
glucagon-like peptide‑1 in the mouse.
Endocrinology 134, 2156–2164 (1994).
Vahl, T. P et al. Glucagon-like peptide‑1 (GLP‑1)
.
receptors expressed on nerve terminals in the
portal vein mediate the effects of endogenous
GLP‑1 on glucose tolerance in rats.
Endocrinology 148, 4965–4973 (2007).
Merchenthaler, I., Lane, M.  Shughrue, P
.
Distribution of pre‑pro‑glucagon and glucagon-like
peptide‑1 receptor messenger RNAs in the rat
central nervous system. J. Comp. Neurol. 403,
261–280 (1999).

NATURE REVIEWS | GASTROENTEROLOGY  HEPATOLOGY 	
© 2012 Macmillan Publishers Limited. All rights reserved

94.	 Baggio, L. L.  Drucker, D. J. Biology of
incretins: GLP‑1 and GIP. Gastroenterology 132,
2131–2157 (2007).
95.	 Brubaker, P. L. Minireview: update on incretin
biology: focus on glucagon-like peptide‑1.
Endocrinology 151, 1984–1989 (2010).
96.	 Salehi, M., Vahl, T. P.  D’Alessio, D. A.
Regulation of islet hormone release and gastric
emptying by endogenous glucagon-like peptide
1 after glucose ingestion. J. Clin. Endocrinol.
Metab. 93, 4909–4916 (2008).
97.	 Edwards, C. M. et al. Glucagon-like peptide 1
has a physiological role in the control of
postprandial glucose in humans: studies with
the antagonist exendin 9–39. Diabetes 48,
86–93 (1999).
98.	 Scrocchi, L. A. et al. Glucose intolerance but
normal satiety in mice with a null mutation in
the glucagon-like peptide 1 receptor gene. Nat.
Med. 2, 1254–1258 (1996).
99.	 Flint, A., Raben, A., Astrup, A.  Holst, J. J.
Glucagon-like peptide 1 promotes satiety and
suppresses energy intake in humans. J. Clin.
Invest. 101, 515–520 (1998).
100.	Naslund, E. et al. Energy intake and appetite are
suppressed by glucagon-like peptide‑1 (GLP‑1)
in obese men. Int. J. Obes. Relat. Metab. Disord.
23, 304–311 (1999).
101.	Rodriquez, D. F. et al. Peripheral versus central
effects of glucagon-like peptide‑1 receptor
agonists on satiety and body weight loss in
Zucker obese rats. Metabolism 49, 709–717
(2000).
102.	Gutzwiller, J. P. et al. Glucagon-like peptide‑1
promotes satiety and reduces food intake in
patients with diabetes mellitus type 2. Am. J.
Physiol. Regul. Integr. Comp. Physiol. 276,
R1541–R1544 (1999).
103.	Lovshin, J. A.  Drucker, D. J. Incretin-based
therapies for type 2 diabetes mellitus. Nat. Rev.
Endocrinol. 5, 262–269 (2009).
104.	Vilsboll, T., Christensen, M., Junker, A. E.,
Knop, F. K.  Gluud, L. L. Effects of glucagonlike peptide‑1 receptor agonists on weight loss:
systematic review and meta-analyses of
randomised controlled trials. BMJ 344, d7771
(2012).
105.	Day, J. W. et al. A new glucagon and GLP‑1 coagonist eliminates obesity in rodents. Nat.
Chem. Biol. 5, 749–757 (2009).
106.	Hughes, T. E. Emerging therapies for metabolic
diseases—the focus is on diabetes and obesity.
Curr. Opin. Chem. Biol. 13, 332–337 (2009).
107.	Thorens, B. Expression cloning of the pancreatic
β cell receptor for the gluco-incretin hormone
glucagon-like peptide 1. Proc. Natl Acad. Sci. USA
89, 8641–8645 (1992).
108.	Huypens, P., Ling, Z., Pipeleers, D.  Schuit, F.
Glucagon receptors on human islet cells
contribute to glucose competence of insulin
release. Diabetologia 43, 1012–1019 (2000).
109.	Bucinskaite, V. et al. Receptor-mediated
activation of gastric vagal afferents by glucagonlike peptide‑1 in the rat. Neurogastroenterol.
Motil. 21, 978–e78 (2009).
110.	Ahren, B. Sensory nerves contribute to insulin
secretion by glucagon-like peptide‑1 in mice.
Am. J. Physiol Regul. Integr. Comp. Physiol. 286,
R269–R272 (2004).
111.	Talsania, T., Anini, Y., Siu, S., Drucker, D. J. 
Brubaker, P. L. Peripheral exendin‑4 and peptide
YY(3–36) synergistically reduce food intake
through different mechanisms in mice.
Endocrinology 146, 3748–3756 (2005).
112.	Jelsing, J. et al. Liraglutide: short-lived effect on
gastric emptying-long lasting effects on body
weight. Diabetes Obes. Metab. 14, 531–538
(2012).

VOLUME 9  |  DECEMBER 2012  |  713
REVIEWS
113.	Waget, A. et al. Physiological and
pharmacological mechanisms through which the
DPP‑4 inhibitor sitagliptin regulates glycemia in
mice. Endocrinology 152, 3018–3029 (2011).
114.	Mussig, K. et al. Association of type 2 diabetes
candidate polymorphisms in KCNQ1 with incretin
and insulin secretion. Diabetes 58, 1715–1720
(2009).
115.	Beinborn, M., Worrall, C. I., McBride, E. W. 
Kopin, A. S. A human glucagon-like peptide‑1
receptor polymorphism results in reduced
agonist responsiveness. Regul. Pept. 130, 1–6
(2005).
116.	Schafer, S. A. et al. Impaired glucagon-like
peptide‑1‑induced insulin secretion in carriers of
transcription factor 7‑like 2 (TCF7L2) gene
polymorphisms. Diabetologia 50, 2443–2450
(2007).
117.	Schafer, S. A. et al. A common genetic variant in
WFS1 determines impaired glucagon-like
peptide‑1‑induced insulin secretion. Diabetologia
52, 1075–1082 (2009).
118.	Munroe, D. G. et al. Prototypic G protein-coupled
receptor for the intestinotrophic factor glucagonlike peptide 2. Proc. Natl Acad. Sci. USA 96,
1569–1573 (1999).
119.	Nelson, D. W., Sharp, J. W., Brownfield, M. S.,
Raybould, H. E.  Ney, D. M. Localization and
activation of glucagon-like peptide‑2 receptors
on vagal afferents in the rat. Endocrinology 148,
1954–1962 (2007).
120.	Drucker, D. J., Ehrlich, P Asa, S. L. 
.,
Brubaker, P Induction of intestinal epithelial
. L.
proliferation by glucagon-like peptide 2. Proc.
Natl Acad. Sci. USA 93, 7911–7916 (1996).
121.	Drucker, D. J. Biologic actions and therapeutic
potential of the proglucagon-derived peptides.
Nat. Clin. Pract. Endocrinol. Metab. 1, 22–31
(2005).
122.	Rowland, K. J.  Brubaker, P The “cryptic”
. L.
mechanism of action of glucagon-like peptide‑2.
Am. J. Physiol. Gastrointest. Liver Physiol. 301,
G1–G8 (2011).
123.	Nagell, C. F., Wettergren, A., Pedersen, J. F.,
Mortensen, D.  Holst, J. J. Glucagon-like
peptide‑2 inhibits antral emptying in man, but is
not as potent as glucagon-like peptide‑1. Scand.
J. Gastroenterol. 39, 353–358 (2004).
124.	Boushey, R. P Yusta, B.  Drucker, D. J.
.,
Glucagon-like peptide 2 decreases mortality and
reduces the severity of indomethacin-induced
murine enteritis. Am. J. Physiol. Endocrinol.
Metab. 277, E937–E947 (1999).
125.	Boushey, R. P Yusta, B.  Drucker, D. J.
.,
Glucagon-like peptide (GLP)‑2 reduces
chemotherapy-associated mortality and
enhances cell survival in cells expressing a
transfected GLP‑2 receptor. Cancer Res. 61,
687–693 (2001).
126.	L’Heureux, M.‑C.  Brubaker, P Glucagon-like
. L.
peptide‑2 and common therapeutics in a murine
model of ulcerative colitis. J. Pharm. Exp. Ther.
306, 347–354 (2003).
127.	Martin, G. R., Wallace, L. E.  Sigalet, D. L.
Glucagon-like peptide‑2 induces intestinal
adaptation in parenterally fed rats with short
bowel syndrome. Am. J. Physiol. Gastrointest.
Liver Physiol. 286, G964–G972 (2004).
128.	Shin, E. D., Drucker, D. J.  Brubaker, P
. L.
Glucagon-like peptide‑2: an update. Curr. Opin.
Endocrinol. Diab. 12, 63–71 (2005).
129.	Lee, S. J. et al. Disruption of the murine Glp2r
impairs paneth cell function and increases
susceptibility to small bowel enteritis.
Endocrinology 153, 1141–1151 (2012).
130.	Bahrami, J., Yusta, B.  Drucker, D. J. ErbB
activity links the glucagon-like peptide‑2 receptor
to refeeding-induced adaptation in the murine

714  |  DECEMBER 2012  |  VOLUME 9

small bowel. Gastroenterology 138, 2447–2456
(2010).
131.	Jeppesen, P. B. et al. Randomised placebocontrolled trial of teduglutide in reducing
parenteral nutrition and/or intravenous fluid
requirements in patients with short bowel
syndrome. Gut 60, 902–914 (2011).
132.	Buchman, A. L., Katz, S., Fang, J. C.,
Bernstein, C. N.  Abou-Assi, S. G. Teduglutide,
a novel mucosally active analog of glucagonlike peptide‑2 (GLP‑2) for the treatment of
moderate to severe Crohn’s disease. Inflamm.
Bowel. Dis. 16, 962–973 (2010).
133.	R  D Pipeline. NPS Pharmaceuticals [online],
http://www.npsp.com/page/rdpipeline/index.
v3page;jsessionid=1t4k7y0l3lntn (2012).
134.	Dube, P. E., Forse, C. L., Bahrami, J. 
Brubaker, P. L. Essential role of insulin-like
growth factor‑1 in the intestinal tropic effects
of glucagon-like peptide‑2 in mice.
Gastroenterology 131, 589–605 (2006).
135.	Rowland, K. J. et al. Loss of glucagon-like
peptide‑2‑induced proliferation following
intestinal epithelial insulin-like growth factor‑1
receptor deletion. Gastroenterology 141,
2166–2175 (2011).
136.	Yusta, B. et al. ErbB signaling is required for
the proliferative actions of GLP‑2 in the murine
gut. Gastroenterology 137, 986–996 (2009).
137.	Orskov, C. et al. GLP‑2 stimulates colonic
growth via KGF, released by subepithelial
myofibroblasts with GLP‑2 receptors. Regul.
Pept. 124, 105–112 (2005).
138.	Sigalet, D. L. et al. Enteric neural pathways
mediate the anti-inflammatory actions of
glucagon-like peptide 2. Am. J. Physiol.
Gastrointest. Liver Physiol. 293, G211–G221
(2007).
139.	Guan, X. et al. GLP‑2 receptor localizes to
enteric neurons and endocrine cells expressing
vasoactive peptides and mediates increased
blood flow. Gastroenterology 130, 150–164
(2006).
140.	Lovshin, J., Estall, J., Yusta, B., Brown, T. J. 
Drucker, D. J. Glucagon-like peptide (GLP)‑2
action in the murine central nervous system is
enhanced by elimination of GLP‑1 receptor
signaling. J. Biol. Chem. 276, 21489–21499
(2001).
141.	Schmidt, P. T. et al. Peripheral administration of
GLP‑2 to humans has no effect on gastric
emptying or satiety. Regul. Pept. 116, 21–25
(2003).
142.	Bataille, D., Gespach, C., Coudray, A. M. 
Rosselin, G. ‘Enteroglucagon’: A specific effect
on gastric glands isolated from the rat fundus.
Evidence for an ‘oxyntomodulin’ action. Biosci.
Rep. 1, 151–155 (1981).
143.	Greenberg, G. R., Fung, L.  Pokol-Daniel, S.
Regulation of somatostatin‑14 and ‑28
secretion by gastric acid in dogs: differential
role of cholecystokinin. Gastroenterology 105,
1387–1395 (1993).
144.	Dakin, C. L. et al. Oxyntomodulin inhibits food
intake in the rat. Endocrinology 142,
4244–4250 (2001).
145.	Cohen, M. A. et al. Oxyntomodulin suppresses
appetite and reduces food intake in humans.
J. Clin. Endocrinol. Metab. 88, 4696–4701
(2003).
146.	Wynne, K. et al. Subcutaneous oxyntomodulin
reduces body weight in overweight and obese
subjects: a double-blind, randomized,
controlled trial. Diabetes 54, 2390–2395
(2005).
147.	Wynne, K. et al. Oxyntomodulin increases
energy expenditure in addition to decreasing
energy intake in overweight and obese humans:



a randomised controlled trial. Int. J. Obes.
(Lond.) 30, 1729–1736 (2006).
148.	Liu, Y. L. et al. Subcutaneous oxyntomodulin
analogue administration reduces body weight in
lean and obese rodents. Int. J. Obes. (Lond.) 34,
1715–1725 (2010).
149.	MacNeil, D. J., Occi, J. L., Hey, P
. J., Strader, C. D.
 Graziano, M. P Cloning and expression of a
.
human glucagon receptor. Biochem. Biophys.
Res. Commun. 198, 328–334 (1994).
150.	Baggio, L. L., Huang, Q., Brown, T. J. 
Drucker, D. J. Oxyntomodulin and glucagon-like
peptide‑1 differentially regulate murine food
intake and energy expenditure. Gastroenterology
127, 546–558 (2004).
151.	Schepp, W. et al. Oxyntomodulin: a cAMPdependent stimulus of rat parietal cell function
via the receptor for glucagon-like peptide‑ 1
(7–36)NH2. Digestion 57, 398–405 (1996).
152.	Dakin, C. L. et al. Peripheral oxyntomodulin
reduces food intake and body weight gain in
rats. Endocrinology 145, 2687–2695 (2004).
153.	Kosinski, J. R. et al. The glucagon receptor is
involved in mediating the body weight-lowering
effects of oxyntomodulin. Obesity (Silver Spring)
20, 1566–1571 (2012).
154.	Habegger, K. M. et al. The metabolic actions of
glucagon revisited. Nat. Rev. Endocrinol. 6,
689–697 (2010).
155.	Pocai, A. et al. Glucagon-like peptide 1/glucagon
receptor dual agonism reverses obesity in mice.
Diabetes 58, 2258–2266 (2009).
156.	Maida, A., Lovshin, J. A., Baggio, L. L. 
Drucker, D. J. The glucagon-like peptide‑1
receptor agonist oxyntomodulin enhances betacell function but does not inhibit gastric
emptying in mice. Endocrinology 149,
5670–5678 (2008).
157.	Hager, J. et al. A missense mutation in the
glucagon receptor gene is associated with
non‑insulin‑dependent diabetes mellitus. Nat.
Genet. 9, 299–304 (1995).
158.	Anini, Y. et al. Oxyntomodulin inhibits pancreatic
secretion through the nervous system in rats.
Pancreas 20, 348–360 (2000).
159.	Tatemoto, K. Isolation and characterization of
peptide YY (PYY), a candidate gut hormone that
inhibits pancreatic exocrine secretion. Proc. Natl
Acad. Sci. USA 79, 2514–2518 (1982).
160.	Lundberg, J. M. et al. Localization of peptide YY
(PYY) in gastrointestinal endocrine cells and
effects on intestinal blood flow and motility.
Proc. Natl Acad. Sci. USA 79, 4471–4475
(1982).
161.	Kanatani, A. et al. Role of the Y1 receptor in the
regulation of neuropeptide Y‑mediated feeding:
comparison of wild-type, Y1 receptor-deficient,
and Y5 receptor-deficient mice. Endocrinology
141, 1011–1016 (2000).
162.	Cabrele, C.  Beck-Sickinger, A. G. Molecular
characterization of the ligand-receptor
interaction of the neuropeptide Y family. J. Pept.
Sci. 6, 97–122 (2000).
163.	Batterham, R. L. et al. Gut hormone PYY(3–36)
physiologically inhibits food intake. Nature 418,
650–654 (2002).
164.	Abbott, C. R. et al. Blockade of the neuropeptide
Y Y2 receptor with the specific antagonist
BIIE0246 attenuates the effect of endogenous
and exogenous peptide YY(3–36) on food
intake. Brain Res. 1043, 139–144 (2005).
165.	Koda, S. et al. The role of the vagal nerve in
peripheral PYY3‑36‑induced feeding reduction in
rats. Endocrinology 146, 2369–2375 (2005).
166.	Burdyga, G. et al. Cholecystokinin regulates
expression of Y2 receptors in vagal afferent
neurons serving the stomach. J. Neurosci. 28,
11583–11592 (2008).

www.nature.com/nrgastro
© 2012 Macmillan Publishers Limited. All rights reserved
REVIEWS
167.	Batterham, R. L. et al. Inhibition of food intake
in obese subjects by peptide YY3–36. N. Engl.
J. Med. 349, 941–948 (2003).
168.	Wynne, K.  Bloom, S. R. The role of
oxyntomodulin and peptide tyrosine-tyrosine
(PYY) in appetite control. Nat. Clin. Pract.
Endocrinol. Metab. 2, 612–620 (2006).
169.	Boggiano, M. M. et al. PYY3–36 as an antiobesity drug target. Obes. Rev. 6, 307–322
(2005).
170.	Ma, L. et al. Variations in peptide YY and Y2
receptor genes are associated with severe
obesity in Pima Indian men. Diabetes 54,
1598–1602 (2005).
171.	Lavebratt, C., Alpman, A., Persson, B., Arner, P. 
Hoffstedt, J. Common neuropeptide Y2 receptor
gene variant is protective against obesity
among Swedish men. Int. J. Obes. (Lond.) 30,
453–459 (2006).
172.	Ahituv, N. et al. A PYY Q62P variant linked to
human obesity. Hum. Mol. Genet. 15, 387–391
(2006).
173.	Neary, N. M. et al. Peptide YY3–36 and
glucagon-like peptide‑17‑36 inhibit food intake
additively. Endocrinology 146, 5120–5127
(2005).
174.	Field, B. C. et al. PYY3–36 and oxyntomodulin
can be additive in their effect on food intake in
overweight and obese humans. Diabetes 59,
1635–1639 (2010).
175.	Reidelberger, R. D., Haver, A. C., Apenteng, B. A.,
Anders, K. L.  Steenson, S. M. Effects of
exendin‑4 alone and with peptide YY(3–36) on
food intake and body weight in diet-induced
obese rats. Obesity (Silver Spring) 19, 121–127
(2011).

176.	Wettergren, A., Maina, P Boesby, S.  Holst, J. J.
.,
Glucagon-like peptide‑1 7–36 amide and peptide
YY have additive inhibitory effect on gastric acid
secretion in man. Scand. J. Gastroenterol. 32,
552–555 (1997).
177.	Baldassano, S., Bellanca, A., Serio, R.  Mule, F.
Food intake in lean and obese mice after
peripheral administration of glucagon-like
peptide‑2. J. Endocrinol. 213, 277–284 (2012).
178.	Lund, A., Vilsboll, T., Bagger, J. I., Holst, J. J. 
Knop, F. K. The separate and combined impact of
the intestinal hormones, GIP GLP‑1, and GLP‑2,
,
on glucagon secretion in type 2 diabetes. Am. J.
Physiol. Endocrinol. Metab. 300, E1038–E1046
(2011).
179.	Dixon, J. B., Straznicky, N. E., Lambert, E. A.,
Schlaich, M. P  Lambert, G. W. Surgical
.
approaches to the treatment of obesity. Nat. Rev.
Gastroenterol. Hepatol. 8, 429–437 (2011).
180.	Schauer, P et al. Bariatric surgery versus
. R.
intensive medical therapy in obese patients with
diabetes. N. Engl. J. Med. 366, 1567–1576
(2012).
181.	Peterli, R. et al. Metabolic and hormonal changes
after laparoscopic Roux‑en‑Y gastric bypass and
sleeve gastrectomy: a randomized, prospective
trial. Obes. Surg. 22, 740–748 (2012).
182.	Bayham, B. E., Greenway, F. L., Bellanger, D. E. 
O’Neil, C. E. Early resolution of type 2 diabetes
seen after Roux‑en‑Y gastric bypass and vertical
sleeve gastrectomy. Diabetes Technol. Ther. 14,
30–34 (2012).
183.	Cummings, D. E. et al. Plasma ghrelin levels after
diet-induced weight loss or gastric bypass
surgery. N. Engl. J. Med. 346, 1623–1630
(2002).

NATURE REVIEWS | GASTROENTEROLOGY  HEPATOLOGY 	
© 2012 Macmillan Publishers Limited. All rights reserved

184.	Korner, J. et al. Prospective study of gut hormone
and metabolic changes after adjustable gastric
banding and Roux‑en‑Y gastric bypass. Int. J.
Obes. (Lond.) 33, 786–795 (2009).
185.	Beckman, L. M., Beckman, T. R. 
Earthman, C. P Changes in gastrointestinal
.
hormones and leptin after Roux‑en‑Y gastric
bypass procedure: a review. J. Am. Diet. Assoc.
110, 571–584 (2010).
186.	Falken, Y., Hellstrom, P
. M., Holst, J. J. 
Naslund, E. Changes in glucose homeostasis
after Roux‑en‑Y gastric bypass surgery for
obesity at day three, two months, and one year
after surgery: role of gut peptides. J. Clin.
Endocrinol. Metab. 96, 2227–2235 (2011).
187.	Chandarana, K. et al. Diet and gastrointestinal
bypass-induced weight loss: the roles of ghrelin
and peptide YY. Diabetes 60, 810–818 (2011).
188.	Chambers, A. P et al. Weight-independent
.
changes in blood glucose homeostasis after
gastric bypass or vertical sleeve gastrectomy in
rats. Gastroenterology 141, 950–958 (2011).
Acknowledgements
C. X. Dong is supported by a graduate studentship
from the Banting and Best Diabetes Centre, University
of Toronto, Canada. P Brubaker is supported by the
. L.
Canada Research Chairs program. Studies on GLP‑1
and GLP‑2 in the Brubaker laboratory are supported
by operating grants from the Canadian Diabetes
Association (#2374) and the Canadian Institutes of
Health Research (#MOP‑9940), respectively.
Author contributions
Both authors contributed equally to all aspects of the
manuscript.

VOLUME 9  |  DECEMBER 2012  |  715

More Related Content

What's hot

GLP 1 E GIP: DIFERENÇAS E SEMELHANÇAS
GLP 1 E GIP: DIFERENÇAS E SEMELHANÇASGLP 1 E GIP: DIFERENÇAS E SEMELHANÇAS
GLP 1 E GIP: DIFERENÇAS E SEMELHANÇASRuy Pantoja
 
Occupational Therapy Leptin & Obesity
Occupational Therapy Leptin & ObesityOccupational Therapy Leptin & Obesity
Occupational Therapy Leptin & ObesityStephan Van Breenen
 
Neuroendocrine regulation of energy
Neuroendocrine regulation of energy Neuroendocrine regulation of energy
Neuroendocrine regulation of energy med_students0
 
Gut hormones - Appetite, Mood and Health
Gut hormones - Appetite, Mood and HealthGut hormones - Appetite, Mood and Health
Gut hormones - Appetite, Mood and HealthQuantified Self Dublin
 
Ghrelin Mathematical model Presentation iasi bio mathlab
Ghrelin Mathematical model Presentation iasi bio mathlabGhrelin Mathematical model Presentation iasi bio mathlab
Ghrelin Mathematical model Presentation iasi bio mathlabJorge Pires
 
Bhavsar et al Curr Diabetes Rev 2013
Bhavsar et al Curr Diabetes Rev 2013Bhavsar et al Curr Diabetes Rev 2013
Bhavsar et al Curr Diabetes Rev 2013Sunil Bhavsar
 
Leptin resistance in obesity
Leptin resistance in obesityLeptin resistance in obesity
Leptin resistance in obesityAbrahamOwodunni
 
Polimorfismos e obesidade
Polimorfismos e obesidadePolimorfismos e obesidade
Polimorfismos e obesidadeNutriline SRL
 
Gastroenterology paper- Homocysteine and SHP and FOXA1
Gastroenterology paper- Homocysteine and SHP and FOXA1Gastroenterology paper- Homocysteine and SHP and FOXA1
Gastroenterology paper- Homocysteine and SHP and FOXA1Attività scientifica
 
Austin Journal of Veterinary Science & Animal Husbandry
Austin Journal of Veterinary Science & Animal Husbandry Austin Journal of Veterinary Science & Animal Husbandry
Austin Journal of Veterinary Science & Animal Husbandry Austin Publishing Group
 
Biological and pharmaceutical importance of proteins
Biological and pharmaceutical importance of proteinsBiological and pharmaceutical importance of proteins
Biological and pharmaceutical importance of proteinsAsad Bilal
 
Final Report Anne Cox
Final Report Anne CoxFinal Report Anne Cox
Final Report Anne CoxAnne Cox
 
Alanine amino transferase_concentrations_are.17
Alanine amino transferase_concentrations_are.17Alanine amino transferase_concentrations_are.17
Alanine amino transferase_concentrations_are.17Giorgio Ibarra
 
4 5827774745222842168
4 58277747452228421684 5827774745222842168
4 5827774745222842168Osman Guure
 

What's hot (20)

GLP 1 E GIP: DIFERENÇAS E SEMELHANÇAS
GLP 1 E GIP: DIFERENÇAS E SEMELHANÇASGLP 1 E GIP: DIFERENÇAS E SEMELHANÇAS
GLP 1 E GIP: DIFERENÇAS E SEMELHANÇAS
 
Occupational Therapy Leptin & Obesity
Occupational Therapy Leptin & ObesityOccupational Therapy Leptin & Obesity
Occupational Therapy Leptin & Obesity
 
Metabolic abnormalities in obesity
Metabolic abnormalities in obesityMetabolic abnormalities in obesity
Metabolic abnormalities in obesity
 
Leptin ppt
Leptin pptLeptin ppt
Leptin ppt
 
Neuroendocrine regulation of energy
Neuroendocrine regulation of energy Neuroendocrine regulation of energy
Neuroendocrine regulation of energy
 
Gut hormones - Appetite, Mood and Health
Gut hormones - Appetite, Mood and HealthGut hormones - Appetite, Mood and Health
Gut hormones - Appetite, Mood and Health
 
Ghrelin Mathematical model Presentation iasi bio mathlab
Ghrelin Mathematical model Presentation iasi bio mathlabGhrelin Mathematical model Presentation iasi bio mathlab
Ghrelin Mathematical model Presentation iasi bio mathlab
 
Leptin
Leptin Leptin
Leptin
 
Bhavsar et al Curr Diabetes Rev 2013
Bhavsar et al Curr Diabetes Rev 2013Bhavsar et al Curr Diabetes Rev 2013
Bhavsar et al Curr Diabetes Rev 2013
 
Genetic Factors II
Genetic Factors IIGenetic Factors II
Genetic Factors II
 
Leptin resistance in obesity
Leptin resistance in obesityLeptin resistance in obesity
Leptin resistance in obesity
 
Adiponectin
AdiponectinAdiponectin
Adiponectin
 
Polimorfismos e obesidade
Polimorfismos e obesidadePolimorfismos e obesidade
Polimorfismos e obesidade
 
Gastroenterology paper- Homocysteine and SHP and FOXA1
Gastroenterology paper- Homocysteine and SHP and FOXA1Gastroenterology paper- Homocysteine and SHP and FOXA1
Gastroenterology paper- Homocysteine and SHP and FOXA1
 
Austin Journal of Veterinary Science & Animal Husbandry
Austin Journal of Veterinary Science & Animal Husbandry Austin Journal of Veterinary Science & Animal Husbandry
Austin Journal of Veterinary Science & Animal Husbandry
 
Biological and pharmaceutical importance of proteins
Biological and pharmaceutical importance of proteinsBiological and pharmaceutical importance of proteins
Biological and pharmaceutical importance of proteins
 
Final Report Anne Cox
Final Report Anne CoxFinal Report Anne Cox
Final Report Anne Cox
 
Alanine amino transferase_concentrations_are.17
Alanine amino transferase_concentrations_are.17Alanine amino transferase_concentrations_are.17
Alanine amino transferase_concentrations_are.17
 
Taurine
TaurineTaurine
Taurine
 
4 5827774745222842168
4 58277747452228421684 5827774745222842168
4 5827774745222842168
 

Similar to Nrgastro.2012.185

Efecto incretina fisologia
Efecto incretina fisologiaEfecto incretina fisologia
Efecto incretina fisologiayanetguzmanaybar
 
2003 role of incretins in glucose homeostasis and diabetes
2003 role of incretins in glucose homeostasis and diabetes2003 role of incretins in glucose homeostasis and diabetes
2003 role of incretins in glucose homeostasis and diabetesDr.Mudasir Bashir
 
41392_2022_Article_1149.pdf
41392_2022_Article_1149.pdf41392_2022_Article_1149.pdf
41392_2022_Article_1149.pdfRodrigoDalia1
 
Ghrelin - an orexigenic hormone
Ghrelin - an orexigenic hormoneGhrelin - an orexigenic hormone
Ghrelin - an orexigenic hormoneAmita Mevada
 
Brain as an endocrine organ
Brain as an endocrine organBrain as an endocrine organ
Brain as an endocrine organalaa wafa
 
Brain as an endocrine organ
Brain as an endocrine organBrain as an endocrine organ
Brain as an endocrine organalaa wafa
 
jcm-08-02203.pdf
jcm-08-02203.pdfjcm-08-02203.pdf
jcm-08-02203.pdfMohdHelali
 
Articulo glp1 pancreas
Articulo glp1 pancreasArticulo glp1 pancreas
Articulo glp1 pancreascrodrigoms
 
Git hormones-control-
Git hormones-control-Git hormones-control-
Git hormones-control-Mawara Ali
 
Gut hormone and its implication in glucose homeostasis
Gut hormone and its implication in glucose homeostasisGut hormone and its implication in glucose homeostasis
Gut hormone and its implication in glucose homeostasisDr. Lin
 
2013 List_The role of GH in adipose tissue-LiGHRKO mice
2013 List_The role of GH in adipose tissue-LiGHRKO mice2013 List_The role of GH in adipose tissue-LiGHRKO mice
2013 List_The role of GH in adipose tissue-LiGHRKO miceEdward List
 
2022 GIT Biochemistry.pptx
2022 GIT Biochemistry.pptx2022 GIT Biochemistry.pptx
2022 GIT Biochemistry.pptxGudetaNoto
 
Food-Matrix-and-Gherlin-Hormone
Food-Matrix-and-Gherlin-HormoneFood-Matrix-and-Gherlin-Hormone
Food-Matrix-and-Gherlin-HormoneAbdallah M. Ayoub
 
Genetics of Eating behavior
Genetics of Eating behaviorGenetics of Eating behavior
Genetics of Eating behaviorMARIA VRANCEANU
 
Polimorfismos y obesidad
Polimorfismos y obesidadPolimorfismos y obesidad
Polimorfismos y obesidadNutriline SRL
 
2009 List et al Diabetologia
2009 List et al Diabetologia2009 List et al Diabetologia
2009 List et al DiabetologiaEdward List
 

Similar to Nrgastro.2012.185 (20)

Laparoscopic Bariatric Surgery
Laparoscopic Bariatric SurgeryLaparoscopic Bariatric Surgery
Laparoscopic Bariatric Surgery
 
weight loss.pdf
weight loss.pdfweight loss.pdf
weight loss.pdf
 
Efecto incretina fisologia
Efecto incretina fisologiaEfecto incretina fisologia
Efecto incretina fisologia
 
2003 role of incretins in glucose homeostasis and diabetes
2003 role of incretins in glucose homeostasis and diabetes2003 role of incretins in glucose homeostasis and diabetes
2003 role of incretins in glucose homeostasis and diabetes
 
41392_2022_Article_1149.pdf
41392_2022_Article_1149.pdf41392_2022_Article_1149.pdf
41392_2022_Article_1149.pdf
 
Ghrelin - an orexigenic hormone
Ghrelin - an orexigenic hormoneGhrelin - an orexigenic hormone
Ghrelin - an orexigenic hormone
 
Brain as an endocrine organ
Brain as an endocrine organBrain as an endocrine organ
Brain as an endocrine organ
 
Brain as an endocrine organ
Brain as an endocrine organBrain as an endocrine organ
Brain as an endocrine organ
 
jcm-08-02203.pdf
jcm-08-02203.pdfjcm-08-02203.pdf
jcm-08-02203.pdf
 
Articulo glp1 pancreas
Articulo glp1 pancreasArticulo glp1 pancreas
Articulo glp1 pancreas
 
Git hormones-control-
Git hormones-control-Git hormones-control-
Git hormones-control-
 
Gut hormone and its implication in glucose homeostasis
Gut hormone and its implication in glucose homeostasisGut hormone and its implication in glucose homeostasis
Gut hormone and its implication in glucose homeostasis
 
2013 List_The role of GH in adipose tissue-LiGHRKO mice
2013 List_The role of GH in adipose tissue-LiGHRKO mice2013 List_The role of GH in adipose tissue-LiGHRKO mice
2013 List_The role of GH in adipose tissue-LiGHRKO mice
 
2022 GIT Biochemistry.pptx
2022 GIT Biochemistry.pptx2022 GIT Biochemistry.pptx
2022 GIT Biochemistry.pptx
 
Food-Matrix-and-Gherlin-Hormone
Food-Matrix-and-Gherlin-HormoneFood-Matrix-and-Gherlin-Hormone
Food-Matrix-and-Gherlin-Hormone
 
Genetics of Eating behavior
Genetics of Eating behaviorGenetics of Eating behavior
Genetics of Eating behavior
 
Presentation on growth harmone and prolactin converted
Presentation on growth harmone and prolactin convertedPresentation on growth harmone and prolactin converted
Presentation on growth harmone and prolactin converted
 
DDW
DDWDDW
DDW
 
Polimorfismos y obesidad
Polimorfismos y obesidadPolimorfismos y obesidad
Polimorfismos y obesidad
 
2009 List et al Diabetologia
2009 List et al Diabetologia2009 List et al Diabetologia
2009 List et al Diabetologia
 

More from Elsa von Licy

Styles of Scientific Reasoning, Scientific Practices and Argument in Science ...
Styles of Scientific Reasoning, Scientific Practices and Argument in Science ...Styles of Scientific Reasoning, Scientific Practices and Argument in Science ...
Styles of Scientific Reasoning, Scientific Practices and Argument in Science ...Elsa von Licy
 
Strategie Decisions Incertitude Actes conference fnege xerfi
Strategie Decisions Incertitude Actes conference fnege xerfiStrategie Decisions Incertitude Actes conference fnege xerfi
Strategie Decisions Incertitude Actes conference fnege xerfiElsa von Licy
 
Neuropsychophysiologie
NeuropsychophysiologieNeuropsychophysiologie
NeuropsychophysiologieElsa von Licy
 
L agressivite en psychanalyse (21 pages 184 ko)
L agressivite en psychanalyse (21 pages   184 ko)L agressivite en psychanalyse (21 pages   184 ko)
L agressivite en psychanalyse (21 pages 184 ko)Elsa von Licy
 
C1 clef pour_la_neuro
C1 clef pour_la_neuroC1 clef pour_la_neuro
C1 clef pour_la_neuroElsa von Licy
 
Vuillez jean philippe_p01
Vuillez jean philippe_p01Vuillez jean philippe_p01
Vuillez jean philippe_p01Elsa von Licy
 
Spr ue3.1 poly cours et exercices
Spr ue3.1   poly cours et exercicesSpr ue3.1   poly cours et exercices
Spr ue3.1 poly cours et exercicesElsa von Licy
 
Plan de cours all l1 l2l3m1m2 p
Plan de cours all l1 l2l3m1m2 pPlan de cours all l1 l2l3m1m2 p
Plan de cours all l1 l2l3m1m2 pElsa von Licy
 
Bioph pharm 1an-viscosit-des_liquides_et_des_solutions
Bioph pharm 1an-viscosit-des_liquides_et_des_solutionsBioph pharm 1an-viscosit-des_liquides_et_des_solutions
Bioph pharm 1an-viscosit-des_liquides_et_des_solutionsElsa von Licy
 
Poly histologie-et-embryologie-medicales
Poly histologie-et-embryologie-medicalesPoly histologie-et-embryologie-medicales
Poly histologie-et-embryologie-medicalesElsa von Licy
 
Methodes travail etudiants
Methodes travail etudiantsMethodes travail etudiants
Methodes travail etudiantsElsa von Licy
 
Atelier.etude.efficace
Atelier.etude.efficaceAtelier.etude.efficace
Atelier.etude.efficaceElsa von Licy
 
There is no_such_thing_as_a_social_science_intro
There is no_such_thing_as_a_social_science_introThere is no_such_thing_as_a_social_science_intro
There is no_such_thing_as_a_social_science_introElsa von Licy
 

More from Elsa von Licy (20)

Styles of Scientific Reasoning, Scientific Practices and Argument in Science ...
Styles of Scientific Reasoning, Scientific Practices and Argument in Science ...Styles of Scientific Reasoning, Scientific Practices and Argument in Science ...
Styles of Scientific Reasoning, Scientific Practices and Argument in Science ...
 
Strategie Decisions Incertitude Actes conference fnege xerfi
Strategie Decisions Incertitude Actes conference fnege xerfiStrategie Decisions Incertitude Actes conference fnege xerfi
Strategie Decisions Incertitude Actes conference fnege xerfi
 
Rainville pierre
Rainville pierreRainville pierre
Rainville pierre
 
Neuropsychophysiologie
NeuropsychophysiologieNeuropsychophysiologie
Neuropsychophysiologie
 
L agressivite en psychanalyse (21 pages 184 ko)
L agressivite en psychanalyse (21 pages   184 ko)L agressivite en psychanalyse (21 pages   184 ko)
L agressivite en psychanalyse (21 pages 184 ko)
 
C1 clef pour_la_neuro
C1 clef pour_la_neuroC1 clef pour_la_neuro
C1 clef pour_la_neuro
 
Hemostase polycop
Hemostase polycopHemostase polycop
Hemostase polycop
 
Antiphilos
AntiphilosAntiphilos
Antiphilos
 
Vuillez jean philippe_p01
Vuillez jean philippe_p01Vuillez jean philippe_p01
Vuillez jean philippe_p01
 
Spr ue3.1 poly cours et exercices
Spr ue3.1   poly cours et exercicesSpr ue3.1   poly cours et exercices
Spr ue3.1 poly cours et exercices
 
Plan de cours all l1 l2l3m1m2 p
Plan de cours all l1 l2l3m1m2 pPlan de cours all l1 l2l3m1m2 p
Plan de cours all l1 l2l3m1m2 p
 
M2 bmc2007 cours01
M2 bmc2007 cours01M2 bmc2007 cours01
M2 bmc2007 cours01
 
Feuilletage
FeuilletageFeuilletage
Feuilletage
 
Chapitre 1
Chapitre 1Chapitre 1
Chapitre 1
 
Biophy
BiophyBiophy
Biophy
 
Bioph pharm 1an-viscosit-des_liquides_et_des_solutions
Bioph pharm 1an-viscosit-des_liquides_et_des_solutionsBioph pharm 1an-viscosit-des_liquides_et_des_solutions
Bioph pharm 1an-viscosit-des_liquides_et_des_solutions
 
Poly histologie-et-embryologie-medicales
Poly histologie-et-embryologie-medicalesPoly histologie-et-embryologie-medicales
Poly histologie-et-embryologie-medicales
 
Methodes travail etudiants
Methodes travail etudiantsMethodes travail etudiants
Methodes travail etudiants
 
Atelier.etude.efficace
Atelier.etude.efficaceAtelier.etude.efficace
Atelier.etude.efficace
 
There is no_such_thing_as_a_social_science_intro
There is no_such_thing_as_a_social_science_introThere is no_such_thing_as_a_social_science_intro
There is no_such_thing_as_a_social_science_intro
 

Recently uploaded

Call Girls Near Hotel Marine Plaza ✔ 9820252231 ✔For 18+ VIP Call Girl At The...
Call Girls Near Hotel Marine Plaza ✔ 9820252231 ✔For 18+ VIP Call Girl At The...Call Girls Near Hotel Marine Plaza ✔ 9820252231 ✔For 18+ VIP Call Girl At The...
Call Girls Near Hotel Marine Plaza ✔ 9820252231 ✔For 18+ VIP Call Girl At The...call girls in ahmedabad high profile
 
Call Girl Number in Vashi Mumbai📲 9833363713 💞 Full Night Enjoy
Call Girl Number in Vashi Mumbai📲 9833363713 💞 Full Night EnjoyCall Girl Number in Vashi Mumbai📲 9833363713 💞 Full Night Enjoy
Call Girl Number in Vashi Mumbai📲 9833363713 💞 Full Night Enjoybabeytanya
 
High Profile Call Girls Coimbatore Saanvi☎️ 8250192130 Independent Escort Se...
High Profile Call Girls Coimbatore Saanvi☎️  8250192130 Independent Escort Se...High Profile Call Girls Coimbatore Saanvi☎️  8250192130 Independent Escort Se...
High Profile Call Girls Coimbatore Saanvi☎️ 8250192130 Independent Escort Se...narwatsonia7
 
♛VVIP Hyderabad Call Girls Chintalkunta🖕7001035870🖕Riya Kappor Top Call Girl ...
♛VVIP Hyderabad Call Girls Chintalkunta🖕7001035870🖕Riya Kappor Top Call Girl ...♛VVIP Hyderabad Call Girls Chintalkunta🖕7001035870🖕Riya Kappor Top Call Girl ...
♛VVIP Hyderabad Call Girls Chintalkunta🖕7001035870🖕Riya Kappor Top Call Girl ...astropune
 
Low Rate Call Girls Pune Esha 9907093804 Short 1500 Night 6000 Best call girl...
Low Rate Call Girls Pune Esha 9907093804 Short 1500 Night 6000 Best call girl...Low Rate Call Girls Pune Esha 9907093804 Short 1500 Night 6000 Best call girl...
Low Rate Call Girls Pune Esha 9907093804 Short 1500 Night 6000 Best call girl...Miss joya
 
Artifacts in Nuclear Medicine with Identifying and resolving artifacts.
Artifacts in Nuclear Medicine with Identifying and resolving artifacts.Artifacts in Nuclear Medicine with Identifying and resolving artifacts.
Artifacts in Nuclear Medicine with Identifying and resolving artifacts.MiadAlsulami
 
Bangalore Call Girls Majestic 📞 9907093804 High Profile Service 100% Safe
Bangalore Call Girls Majestic 📞 9907093804 High Profile Service 100% SafeBangalore Call Girls Majestic 📞 9907093804 High Profile Service 100% Safe
Bangalore Call Girls Majestic 📞 9907093804 High Profile Service 100% Safenarwatsonia7
 
Call Girls Service Surat Samaira ❤️🍑 8250192130 👄 Independent Escort Service ...
Call Girls Service Surat Samaira ❤️🍑 8250192130 👄 Independent Escort Service ...Call Girls Service Surat Samaira ❤️🍑 8250192130 👄 Independent Escort Service ...
Call Girls Service Surat Samaira ❤️🍑 8250192130 👄 Independent Escort Service ...CALL GIRLS
 
Russian Call Girls in Pune Tanvi 9907093804 Short 1500 Night 6000 Best call g...
Russian Call Girls in Pune Tanvi 9907093804 Short 1500 Night 6000 Best call g...Russian Call Girls in Pune Tanvi 9907093804 Short 1500 Night 6000 Best call g...
Russian Call Girls in Pune Tanvi 9907093804 Short 1500 Night 6000 Best call g...Miss joya
 
💎VVIP Kolkata Call Girls Parganas🩱7001035870🩱Independent Girl ( Ac Rooms Avai...
💎VVIP Kolkata Call Girls Parganas🩱7001035870🩱Independent Girl ( Ac Rooms Avai...💎VVIP Kolkata Call Girls Parganas🩱7001035870🩱Independent Girl ( Ac Rooms Avai...
💎VVIP Kolkata Call Girls Parganas🩱7001035870🩱Independent Girl ( Ac Rooms Avai...Taniya Sharma
 
Call Girls In Andheri East Call 9920874524 Book Hot And Sexy Girls
Call Girls In Andheri East Call 9920874524 Book Hot And Sexy GirlsCall Girls In Andheri East Call 9920874524 Book Hot And Sexy Girls
Call Girls In Andheri East Call 9920874524 Book Hot And Sexy Girlsnehamumbai
 
(Rocky) Jaipur Call Girl - 9001626015 Escorts Service 50% Off with Cash ON De...
(Rocky) Jaipur Call Girl - 9001626015 Escorts Service 50% Off with Cash ON De...(Rocky) Jaipur Call Girl - 9001626015 Escorts Service 50% Off with Cash ON De...
(Rocky) Jaipur Call Girl - 9001626015 Escorts Service 50% Off with Cash ON De...indiancallgirl4rent
 
Call Girls Service Pune Vaishnavi 9907093804 Short 1500 Night 6000 Best call ...
Call Girls Service Pune Vaishnavi 9907093804 Short 1500 Night 6000 Best call ...Call Girls Service Pune Vaishnavi 9907093804 Short 1500 Night 6000 Best call ...
Call Girls Service Pune Vaishnavi 9907093804 Short 1500 Night 6000 Best call ...Miss joya
 
Best Rate (Hyderabad) Call Girls Jahanuma ⟟ 8250192130 ⟟ High Class Call Girl...
Best Rate (Hyderabad) Call Girls Jahanuma ⟟ 8250192130 ⟟ High Class Call Girl...Best Rate (Hyderabad) Call Girls Jahanuma ⟟ 8250192130 ⟟ High Class Call Girl...
Best Rate (Hyderabad) Call Girls Jahanuma ⟟ 8250192130 ⟟ High Class Call Girl...astropune
 
Call Girls Horamavu WhatsApp Number 7001035870 Meeting With Bangalore Escorts
Call Girls Horamavu WhatsApp Number 7001035870 Meeting With Bangalore EscortsCall Girls Horamavu WhatsApp Number 7001035870 Meeting With Bangalore Escorts
Call Girls Horamavu WhatsApp Number 7001035870 Meeting With Bangalore Escortsvidya singh
 
Bangalore Call Girls Nelamangala Number 7001035870 Meetin With Bangalore Esc...
Bangalore Call Girls Nelamangala Number 7001035870  Meetin With Bangalore Esc...Bangalore Call Girls Nelamangala Number 7001035870  Meetin With Bangalore Esc...
Bangalore Call Girls Nelamangala Number 7001035870 Meetin With Bangalore Esc...narwatsonia7
 
Russian Call Girls in Bangalore Manisha 7001305949 Independent Escort Service...
Russian Call Girls in Bangalore Manisha 7001305949 Independent Escort Service...Russian Call Girls in Bangalore Manisha 7001305949 Independent Escort Service...
Russian Call Girls in Bangalore Manisha 7001305949 Independent Escort Service...narwatsonia7
 
College Call Girls Pune Mira 9907093804 Short 1500 Night 6000 Best call girls...
College Call Girls Pune Mira 9907093804 Short 1500 Night 6000 Best call girls...College Call Girls Pune Mira 9907093804 Short 1500 Night 6000 Best call girls...
College Call Girls Pune Mira 9907093804 Short 1500 Night 6000 Best call girls...Miss joya
 

Recently uploaded (20)

Call Girls Near Hotel Marine Plaza ✔ 9820252231 ✔For 18+ VIP Call Girl At The...
Call Girls Near Hotel Marine Plaza ✔ 9820252231 ✔For 18+ VIP Call Girl At The...Call Girls Near Hotel Marine Plaza ✔ 9820252231 ✔For 18+ VIP Call Girl At The...
Call Girls Near Hotel Marine Plaza ✔ 9820252231 ✔For 18+ VIP Call Girl At The...
 
Call Girl Number in Vashi Mumbai📲 9833363713 💞 Full Night Enjoy
Call Girl Number in Vashi Mumbai📲 9833363713 💞 Full Night EnjoyCall Girl Number in Vashi Mumbai📲 9833363713 💞 Full Night Enjoy
Call Girl Number in Vashi Mumbai📲 9833363713 💞 Full Night Enjoy
 
High Profile Call Girls Coimbatore Saanvi☎️ 8250192130 Independent Escort Se...
High Profile Call Girls Coimbatore Saanvi☎️  8250192130 Independent Escort Se...High Profile Call Girls Coimbatore Saanvi☎️  8250192130 Independent Escort Se...
High Profile Call Girls Coimbatore Saanvi☎️ 8250192130 Independent Escort Se...
 
♛VVIP Hyderabad Call Girls Chintalkunta🖕7001035870🖕Riya Kappor Top Call Girl ...
♛VVIP Hyderabad Call Girls Chintalkunta🖕7001035870🖕Riya Kappor Top Call Girl ...♛VVIP Hyderabad Call Girls Chintalkunta🖕7001035870🖕Riya Kappor Top Call Girl ...
♛VVIP Hyderabad Call Girls Chintalkunta🖕7001035870🖕Riya Kappor Top Call Girl ...
 
Low Rate Call Girls Pune Esha 9907093804 Short 1500 Night 6000 Best call girl...
Low Rate Call Girls Pune Esha 9907093804 Short 1500 Night 6000 Best call girl...Low Rate Call Girls Pune Esha 9907093804 Short 1500 Night 6000 Best call girl...
Low Rate Call Girls Pune Esha 9907093804 Short 1500 Night 6000 Best call girl...
 
Artifacts in Nuclear Medicine with Identifying and resolving artifacts.
Artifacts in Nuclear Medicine with Identifying and resolving artifacts.Artifacts in Nuclear Medicine with Identifying and resolving artifacts.
Artifacts in Nuclear Medicine with Identifying and resolving artifacts.
 
Bangalore Call Girls Majestic 📞 9907093804 High Profile Service 100% Safe
Bangalore Call Girls Majestic 📞 9907093804 High Profile Service 100% SafeBangalore Call Girls Majestic 📞 9907093804 High Profile Service 100% Safe
Bangalore Call Girls Majestic 📞 9907093804 High Profile Service 100% Safe
 
Call Girls Service Surat Samaira ❤️🍑 8250192130 👄 Independent Escort Service ...
Call Girls Service Surat Samaira ❤️🍑 8250192130 👄 Independent Escort Service ...Call Girls Service Surat Samaira ❤️🍑 8250192130 👄 Independent Escort Service ...
Call Girls Service Surat Samaira ❤️🍑 8250192130 👄 Independent Escort Service ...
 
Russian Call Girls in Pune Tanvi 9907093804 Short 1500 Night 6000 Best call g...
Russian Call Girls in Pune Tanvi 9907093804 Short 1500 Night 6000 Best call g...Russian Call Girls in Pune Tanvi 9907093804 Short 1500 Night 6000 Best call g...
Russian Call Girls in Pune Tanvi 9907093804 Short 1500 Night 6000 Best call g...
 
Russian Call Girls in Delhi Tanvi ➡️ 9711199012 💋📞 Independent Escort Service...
Russian Call Girls in Delhi Tanvi ➡️ 9711199012 💋📞 Independent Escort Service...Russian Call Girls in Delhi Tanvi ➡️ 9711199012 💋📞 Independent Escort Service...
Russian Call Girls in Delhi Tanvi ➡️ 9711199012 💋📞 Independent Escort Service...
 
💎VVIP Kolkata Call Girls Parganas🩱7001035870🩱Independent Girl ( Ac Rooms Avai...
💎VVIP Kolkata Call Girls Parganas🩱7001035870🩱Independent Girl ( Ac Rooms Avai...💎VVIP Kolkata Call Girls Parganas🩱7001035870🩱Independent Girl ( Ac Rooms Avai...
💎VVIP Kolkata Call Girls Parganas🩱7001035870🩱Independent Girl ( Ac Rooms Avai...
 
Call Girls In Andheri East Call 9920874524 Book Hot And Sexy Girls
Call Girls In Andheri East Call 9920874524 Book Hot And Sexy GirlsCall Girls In Andheri East Call 9920874524 Book Hot And Sexy Girls
Call Girls In Andheri East Call 9920874524 Book Hot And Sexy Girls
 
(Rocky) Jaipur Call Girl - 9001626015 Escorts Service 50% Off with Cash ON De...
(Rocky) Jaipur Call Girl - 9001626015 Escorts Service 50% Off with Cash ON De...(Rocky) Jaipur Call Girl - 9001626015 Escorts Service 50% Off with Cash ON De...
(Rocky) Jaipur Call Girl - 9001626015 Escorts Service 50% Off with Cash ON De...
 
Call Girls Service Pune Vaishnavi 9907093804 Short 1500 Night 6000 Best call ...
Call Girls Service Pune Vaishnavi 9907093804 Short 1500 Night 6000 Best call ...Call Girls Service Pune Vaishnavi 9907093804 Short 1500 Night 6000 Best call ...
Call Girls Service Pune Vaishnavi 9907093804 Short 1500 Night 6000 Best call ...
 
Best Rate (Hyderabad) Call Girls Jahanuma ⟟ 8250192130 ⟟ High Class Call Girl...
Best Rate (Hyderabad) Call Girls Jahanuma ⟟ 8250192130 ⟟ High Class Call Girl...Best Rate (Hyderabad) Call Girls Jahanuma ⟟ 8250192130 ⟟ High Class Call Girl...
Best Rate (Hyderabad) Call Girls Jahanuma ⟟ 8250192130 ⟟ High Class Call Girl...
 
Call Girls Horamavu WhatsApp Number 7001035870 Meeting With Bangalore Escorts
Call Girls Horamavu WhatsApp Number 7001035870 Meeting With Bangalore EscortsCall Girls Horamavu WhatsApp Number 7001035870 Meeting With Bangalore Escorts
Call Girls Horamavu WhatsApp Number 7001035870 Meeting With Bangalore Escorts
 
Escort Service Call Girls In Sarita Vihar,, 99530°56974 Delhi NCR
Escort Service Call Girls In Sarita Vihar,, 99530°56974 Delhi NCREscort Service Call Girls In Sarita Vihar,, 99530°56974 Delhi NCR
Escort Service Call Girls In Sarita Vihar,, 99530°56974 Delhi NCR
 
Bangalore Call Girls Nelamangala Number 7001035870 Meetin With Bangalore Esc...
Bangalore Call Girls Nelamangala Number 7001035870  Meetin With Bangalore Esc...Bangalore Call Girls Nelamangala Number 7001035870  Meetin With Bangalore Esc...
Bangalore Call Girls Nelamangala Number 7001035870 Meetin With Bangalore Esc...
 
Russian Call Girls in Bangalore Manisha 7001305949 Independent Escort Service...
Russian Call Girls in Bangalore Manisha 7001305949 Independent Escort Service...Russian Call Girls in Bangalore Manisha 7001305949 Independent Escort Service...
Russian Call Girls in Bangalore Manisha 7001305949 Independent Escort Service...
 
College Call Girls Pune Mira 9907093804 Short 1500 Night 6000 Best call girls...
College Call Girls Pune Mira 9907093804 Short 1500 Night 6000 Best call girls...College Call Girls Pune Mira 9907093804 Short 1500 Night 6000 Best call girls...
College Call Girls Pune Mira 9907093804 Short 1500 Night 6000 Best call girls...
 

Nrgastro.2012.185

  • 1. REVIEWS Ghrelin, the proglucagon-derived peptides and peptide YY in nutrient homeostasis Charlotte X. Dong and Patricia L. Brubaker Abstract | Dysregulation of nutrient homeostasis is implicated in the current epidemics of obesity and type 2 diabetes mellitus. The maintenance of homeostasis in the setting of repeated cycles of feeding and fasting occurs through complex interactions between metabolic, hormonal and neural factors. Although pancreatic islets, the liver, muscle, adipocytes and the central nervous system are all key players in this network, the gastrointestinal tract is the first tissue exposed to ingested nutrients and thus has an important role. This Review focuses on several of the endocrine hormones released by the gastrointestinal tract prior to or during nutrient ingestion that have key roles in maintaining energy balance. These hormones include the gastric orexigenic hormone, ghrelin, and the distal L cell anorexigenic and metabolic hormones, glucagon-like peptide (GLP)‑1, GLP‑2, oxyntomodulin and peptide YY. Each of these hormones exerts a distinct set of biological actions to maintain nutrient homeostasis, the properties of which are currently, or might soon be, exploited in the clinic for the treatment of obesity and type 2 diabetes mellitus. Dong, C. X. & Brubaker, P Nat. Rev. Gastroenterol. Hepatol. 9, 705–715 (2012); published online 2 October 2012; . L. doi:10.1038/nrgastro.2012.185 Introduction A complex interplay of metabolic, hormonal and neural factors is required to maintain nutrient homeostasis under a wide variety of environmental conditions includ­ ing, most notably, the daily cycle of feeding and fasting. Although pancreatic islets, the liver, muscle, adipocytes and the central nervous system (CNS) are all key players in this network, the gastrointestinal tract is the first tissue affected by nutrient ingestion. As dysregulation of nutri­ ent homeostasis might be linked to the current epi­demics of obesity and type 2 diabetes mellitus, this Review focuses on several of the hormones released by the gastro­intestinal tract prior to or during nutrient ingestion that have key roles in maintaining energy balance. Many gastrointestinal hormones have now been iden­ tified,1 making the gut the largest endocrine organ of the body; most of these hormones are produced by entero­ endocrine cells scattered throughout the gastro­intestinal epithelium. 2 These cells were originally classified as being either ‘open’ or ‘closed’ to the intestinal lumen on the basis of the presence or absence of microvilli on the luminal surface.3 Some enteroendocrine cells are also directly innervated and/or extend long ‘axonal-like’ pro­ cesses into the lamina propria, both of which probably permit direct interactions with the nervous system.4,5 These findings are consistent with the expression of multiple entero­ ndocrine hormone receptors by the e Competing interests C. X. Dong declares no competing interests. P Brubaker . L. declares associations with the following companies: Eli Lilly; Merck, Sharp & Dome; NPS Pharmaceuticals. See the article online for full details of the relationships. vagus nerve.6 Open-type enteroendocrine cells seem to ‘sense’ luminal nutrients through multiple mechanisms, including both classic transporters and membranebound nutrient receptors, through which intracellular signalling pathways are activated.7 Collectively, these findings indicate the existence of a highly inter-related network between luminal nutrients, gut hormones and the nervous system. Two enteroendocrine cells have been selected for this Review on the basis of the demonstrated importance of their constituent hormones to whole body energy homeo­ tasis, as well as on the basis of their actual or s potential use in the treatment of the prevalent syndromes of nutrient dysregulation, obesity and type 2 diabetes mellitus. These two cells are the proximal X/A-like cell, which releases ghrelin, and the distal L cell, which pro­ duces the proglucagon-­ erived peptides, glucagon-like d peptide (GLP)‑1, GLP‑2 and oxynto­ odulin, as well as m peptide YY (PYY). Of note, although all of these peptides are also synthesized in the brain, the focus of this Review is on the gastro­ ntestinal tract as the major source of i these hormones in the peripheral circulation. Finally, the focus of this Review on these specific peptides does not discount the key contributions of other gut hormones to nutrient homeostasis, including glucose-dependent insulino­ ropic peptide (GIP) and cholecystokinin.8,9 t Indeed, develop­ ental, functional and compensatory m relationships between ghrelin, GLP‑1, GLP‑2, PYY, GIP and cholecystokinin have been reported,8,10,11 indicat­ ing that the gastrointestinal hormones function in a highly integrated fashion to optimize nutrient digestion, absorption and assimilation. NATURE REVIEWS | GASTROENTEROLOGY & HEPATOLOGY © 2012 Macmillan Publishers Limited. All rights reserved Department of Physiology, Medical Sciences Building, University of Toronto, 1 King’s College Circle, Toronto, ON M5S 1A8, Canada (C. X. Dong, P. L. Brubaker). Correspondence to: P Brubaker . L. p.brubaker@utoronto.ca VOLUME 9  |  DECEMBER 2012  |  705
  • 2. REVIEWS Key points ■■ Enteroendocrine cells in the gastrointestinal tract produce various hormones that have essential roles in the maintenance of nutrient homeostasis in both the fasting and fed states ■■ Ghrelin is an orexigenic peptide released by the proximal X/A-like cells prior to nutrient ingestion; the enteroendocrine L cells release glucagon-like peptide (GLP)‑1, GLP‑2, oxyntomodulin and peptide YY (PYY) in response to nutrient ingestion ■■ GLP‑1 is a key incretin hormone, which also inhibits glucagon release, gastric emptying and appetite; agents that increase GLP‑1 action are used to treat type 2 diabetes mellitus ■■ GLP‑2 regulates nutrient absorption by the gastrointestinal tract through enhancement of intestinal growth, nutrient digestion and transport, as well as blood flow ■■ The physiologic role of oxyntomodulin remains to be established, but exogenous administration suppresses food intake; PYY3–36NH2 has an important role as an anorexigenic gut hormone ■■ Surgically induced alterations in the levels of ghrelin and/or GLP‑1, oxyntomodulin and PYY improve glycaemic control and induce weight loss; pharmacological manipulation of these gut hormones might prove beneficial The X/A-like cell Ghrelin was first discovered in 1999 as a growthhormone-­releasing peptide.12 The following character­ istics make ghrelin unique amongst the gut hormones: first, it is located primarily in the proximal gut; second, it is produced by both closed-type and open-type entero­ endocrine cells; third, it is acylated; fourth, levels are increased in the fasting state; and fifth, it is orexigenic. As a consequence, the regulation of ghrelin synthesis, secretion and biological activities differs markedly from that of GLP‑1, GLP‑2, oxyntomodulin and PYY. The rat X/A-like cell (known as the P/D 1 cell in humans) was identified as the ghrelin-producing cell in 2000, having originally been named ‘X’ for its unknown hormone product and ‘A-like’ for the morphologic simi­ larity of its secretory granules with those of the pancre­ atic A cell.13,14 X/A-like cells represent ~20% of the total number of endocrine cells of the gastric corpus mucosa in rats, with profoundly reduced cell numbers along the aboral axis.15 Interestingly, the proximal X/A-like cells are almost exclusively closed-type enteroendocrine cells, whereas >60% of the distal X/A-like cells (that is, ileal, cecal and colonic) are open type.15 Although the func­ tional importance of these different cell types remains unclear, their morphologic characteristics suggest that these cells might be differentially regulated, such that the distal, but not the proximal, cells might be responsive to the presence of nutrients in the intestinal lumen. Synthesized by post-translational processing of proghrelin through the actions of prohormone con­ vertase (PC) 1/3,16 ghrelin is modified on Ser3 through covalent linkage to a medium-chain (C8–C10) fatty acid by ghrelin O‑acyltransferase (GOAT).17 This acyla­ tion is required for the activity of ghrelin on the growth hormone secretagogue receptor, now also known as the ghrelin receptor (GRLN‑R),12 and is a unique posttranslational modification amongst all of the known peptide hormones. Hence, GOAT levels determine the proportion of bioactive ghrelin that is released by the X/A-like cell. 706  |  DECEMBER 2012  |  VOLUME 9 Circulating levels of ghrelin increase before a meal in both rodents and humans, and are suppressed by nutri­ ent ingestion (Figure 1).18,19 Given the closed nature of the proximal X/A-like cell, it has proven difficult to elu­ cidate the exact mechanisms regulating ghrelin release in response to fasting and nutrient ingestion. Nonetheless, in vivo studies have implicated both sympathetic and parasympathetic neurons as stimulatory effectors of the X/A-like cell, as well as hyperglycaemia, insulin and somato­ tatin from gastric D cells as inhibitory regulators s of ghrelin release.20–24 Studies on primary rodent gastric X/A-like cell cultures have confirmed the stimulatory effects of epinephrine and norepinephrine on ghrelin release.25,26 Furthermore, 1 mM glucose (comparable to hypo­ lycaemia) enhanced ghrelin release, whereas g 5 mM ‘normoglycaemic’ glucose had no effect and 10 mM glucose inhibited ghrelin release; inhibitory effects of insulin were also observed but only in the setting of low or normal glucose levels.25,26 These findings are consistent with the demonstration that these cells express glucosesensing machinery, including transporters, hexokinases and the KATP channel, as well as a functional insulin recep­ tor/phosphatidylinositol 3 (PI3)-kinase/Akt signalling pathway.25,26 Hence, the available data suggest that ghrelin release before a meal is coupled to activation of the sym­ pathetic nervous system. Conversely, the prandial suppres­ sion of ghrelin secretion is probably mediated, at least in part, by increasing levels of glucose and/or insulin. Finally, given the original identification of ghrelin-producing cells as ‘A-like’ on the basis of their morphological characteris­ tics,13,14 it is interesting that the regulation of ghrelin secre­ tion by X/A-like cells bears a striking resemblance to that of pancreatic glucagon by the A cell, which is also stimu­ lated by the sympathetic nervous system and inhibited by high levels of both glucose and insulin.27,28 The finding of a lineage relationship between pancreatic islet ghrelin and A cells further suggests that the gastric X/A-like and islet A cells might also be related,29 although transgenic expres­ sion studies have not identified any such link to date.29–31 Nonetheless, these findings suggest that, under both fasting and fed conditions, ghrelin and glucagon secretion are coordinated to maintain nutrient homeostasis. The orexigenic action of ghrelin and the require­ ment for the GRLN‑R in nutrient homeostasis has been demon­strated in rodents and humans using a wide variety of gain-of-function and loss-of-function approaches, and is mediated not only by increased food intake, but also via decreased energy expenditure.32–36 As ghrelin crosses the blood–brain barrier, related in part to its acylation status,37 the orexigenic effects of this gut hormone are thought to be mediated through direct effects on neurons in the central feeding centre, including those expressing the orexins.32,38 However, the GRLN‑R is also expressed by the vagus nerve and, although controversial, one study has demonstrated that both truncal vagotomy and vagal afferent ablation can prevent the orexigenic actions of peripherally administered ghrelin on food intake.39 Studies have also demonstrated that ghrelin has a role in glucose homeostasis through expression of the GRLN‑R in pancreatic islets, the liver and, to a much www.nature.com/nrgastro © 2012 Macmillan Publishers Limited. All rights reserved
  • 3. REVIEWS lesser extent, muscle. 40 Consistent with these find­ ings, ghrelin has been shown to both inhibit glucose-­ stimulated insulin secretion and decrease insulin sensitivity in normal mice, with improved glucose toler­ ance found in knockout animals.34,36 Administration of a GOAT inhibitor also enhances glucose tolerance in mice, in a ghrelin-­ ependent manner.35 However, the glycae­ d mic actions of ghrelin might also be mediated through its effects as a growth hormone secretagogue, as GOAT knockout mice subjected to severe caloric restriction experience lethal hypoglycaemia, even in the setting of profoundly reduced insulin levels, and ghrelin replace­ ment prevents the mortality through increased growth hormone levels.41 Together, these findings suggest the existence of a metabolic negative feedback loop that regulates circulating ghrelin levels, probably through modulation of blood glucose levels. Given the important role of ghrelin in stimulat­ ing appetite, it is not surprising that ghrelin levels are decreased in obese individuals, consistent with negative feedback regulation under conditions of positive energy balance. 42–44 However, concentrations of circulating ghrelin are reduced even further by insulin resistance that is associated with either obesity or type 2 diabe­ tes mellitus.43,45 These in vivo data are in keeping with reports that ghrelin release by primary X/A-like cells is inhibited by physiological concentrations of insulin in the setting of low glucose levels,25,26 but not with the finding that insulin resistance in these cells prevents insulin-induced suppression of ghrelin.26 Furthermore, the suppression of endogenous ghrelin levels in all of these conditions raises the question as to whether antagonism of the ghrelin system will provide additional benefit to body weight regulation in any of these condi­ tions. Collectively, therefore, it remains to be established whether manipulation of ghrelin signalling will be of clinical benefit in conditions characterized by obesity. No strong link has been found between body weight dysregulation and polymorphisms in the ghrelin– GRLN‑R axis.46 However, increased ghrelin levels have been reported in individuals with anorexia, which sug­ gests a role for ghrelin resistance in this condition.47 A number of clinical trials have now examined the effects of ghrelin administration in a variety of conditions char­ acterized by reduced food intake due to cachexia; to date, results from these trials have been generally positive.48 Conversely, ghrelin receptor antagonists have been pro­ posed for use in obesity,49 with a reduction in body weight associated with decreased food intake demonstrated in diet-induced obese mice; enhanced glycaemic control during an intraperitoneal glucose tolerance test was also noted in these animals.32 Similar findings have been reported for a GOAT inhibitor.35 However, results from clinical trials using either approach are still pending. The L cell In contrast to ghrelin, peptides synthesized by the intes­ tinal L cell have the following characteristics: first, they are produced predominantly in the distal small intestine (that is, the ileum) and colon; second, they are found Norepinephrine Glucose Insulin IR β1R GLUT1/4/5 PI3K cAMP Glucokinase or hexokinase AKT ?? Low concentrations of glucose High concentrations of glucose PKA Ghrelin Figure 1 | The X/A-like cell. Under fasting conditions (for example, low levels of insulin and glucose), the X/A-like cell is stimulated by the sympathetic nervous system through release of norepinephrine and activation of the β1R, which is known to be coupled to cAMP and PKA signalling. Low concentrations of glucose also directly stimulate ghrelin release, at least in vitro. Under fed conditions (for example, high levels of insulin and glucose), insulin inhibits ghrelin release through the canonical PI3K/Akt pathway, whereas glucose inhibits secretion through a pathway that might involve glucose transporters and glucokinase or hexokinase. Abbreviations: β1R, β1 adrenergic receptor; GLUT1/4/5, glucose transporter 1, 4 or 5; IR, insulin receptor; PI3K, phosphatidylinositol‑3 kinase‑1; PKA, protein kinase A. only in open-type L cells; third, their levels are increased by feeding; and fourth, they are anorexigenic. GLP‑1, GLP‑2 and oxyntomodulin are synthesized by PC1/3-mediated cleavage of proglucagon, and are cosecreted by the L cell.50–52 Although glicentin and two intervening peptides are also liberated from proglucagon in the L cell, no physiological actions have been delin­ eated for these hormones and we do not discuss them further in this Review. The highest density of L cells is in the distal gut, but they are also localized in the upper gastro­ ntestinal tract of rats and humans, although i in markedly lower numbers than in the distal gut.53,54 Interestingly, findings have indicated that ~20% percent of proximal L cells co-express GIP, and 40% express cholecystokinin.11,55 By contrast, GIP and cholecysto­ kinin expression by distal L cells is fairly low, but ~45% of the ileal and ~70% of the colonic L cells co-express PYY, the functions of which are described below. 11,55 Furthermore, a study has indicated that the phenotype of the colonic L cell is altered by abnormal carbohydrate flux to this tissue, such that the cell increases its capacity to respond to the presence of these luminal nutrients.56 Compared with proximal L cells, colonic L cells might NATURE REVIEWS | GASTROENTEROLOGY HEPATOLOGY © 2012 Macmillan Publishers Limited. All rights reserved VOLUME 9  |  DECEMBER 2012  |  707
  • 4. REVIEWS Insulin Acetylcholine Leptin IR M1R LR PI3K MEK AKT ERK1/2 STAT3 GLP1/2 Oxyntomodulin PYY Ca2+ PKCζ PKA/PKC GPRs Fatty acids Oleoylethanoloamide 2-Oleoylglycerol ∆ψ FATP4 Oleic acid SGLT1 Glucose Na+ Figure 2 | The L cell. In the fed state, secretion by the L cell is rapidly stimulated by the vagus, through release of acetylcholine and activation of M1R. After passage down the gastrointestinal tract, luminal nutrients then enhance secretion through a variety of pathways: GPRs for long and short-chain fatty acids, oleoylethanoloamide and 2‑oleoylglycerol (leading to activation of PKA or PKC); FATP4 for oleic acid (leading to PKCζ signalling); and SGLT1 for sodium and glucose (resulting in depolarization (Ψ)-induced calcium signalling). Insulin and leptin also enhance secretion by the intestinal L cell in a positive-feedback fashion, through canonical signalling pathways. Abbreviations: ERK1/2, extracellular signal-regulated kinase 1/2; FATP4, fatty acid transport protein 4; GLP‑1/2, glucagon-like peptide 1 or 2; GPRs, G protein-coupled receptors; IR, insulin receptor; LR; leptin receptor isoform b; M1R, the M1 muscarinic receptor; MEK, MAPK/ERK kinase; PI3K, phosphatidylinositol‑3 kinase‑1; PKA, protein kinase A; PKC, protein kinase C; PYY, peptide YY; SGLT1, sodium-glucose transporter 1; STAT3, signal transducer activator of transcription 3. also respond preferentially to short-chain fatty acids,57 which is consistent with the high density of gut bacteria in this organ. The physiological importance of the differ­ ences between the proximal and distal L cell populations, as well as their developmental aetiology, clearly requires further examination. Hormone secretion by the L cell is stimulated by nutri­ ent ingestion; plasma levels of these hormones exhibit an early rise followed by a prolonged plateau phase.58 Studies 708  |  DECEMBER 2012  |  VOLUME 9 in rodents have demonstrated that the early L cell response is mediated indirectly by proximal activation of the vagus nerve, leading to muscarinic receptor signalling in the distal L cell; some evidence exists for a similar pathway in humans (Figure 2).4,59,60 By contrast, the prolonged secretory phase is triggered by nutrients in the gut lumen that act directly on ileal and/or colonic L cells through a variety of mechanisms, including the following: sig­ nalling coupled through transporters (for example, for glucose: sodium–glucose transporter‑1 and, to a lesser extent, KATP channels, leading to Na+ and K+-induced depolarization, respectively; and for oleic acid: fatty acid transport protein 4 and atypical protein kinase C‑ζ);7,61–65 or G‑protein-coupled receptor signalling (for example, for short-chain fatty acids: GRP40 and GRP43; for long-chain fatty acids: GPR40 and GPR120; for oleoylethanol­ mide a and 2‑oleoylglycerol: GPR119; and for glucose: sweet taste receptors). The bile acid receptor, TGR5, is also expressed by the distal L cell.56,57,66–70 Thus, luminal nutrient compo­ sition has a key role in the stimulation of L cell secretion, with diets enriched in oleic acid, for example, increasing hormone release to a greater extent than diets high in saturated fatty acids.71,72 Although L cell secretion is also known to be modulated by a number of hormones and neuropeptides (for example, cholecysto­ inin, gastrink releasing peptide, GIP, insulin, leptin and somatostatin),73 the physiological role of many of these factors and how their actions are coupled to nutrient ingestion remain to be fully elucidated. Finally, just as the X/A-like cell shares many regulatory features with the pancreatic A cell, so too is hormone secretion by the L cell similar to that of islet β cells, particularly with respect to the pathways regulating glucose and fatty acid responses.74 Not only is hormone secretion by the gut and pancreatic islets thereby coor­ dinated in the fed state, but many of these pathways are currently under investigation as coordinated L and β‑cell secretagogues, as discussed in more detail below. Both insulin and leptin also serve as GLP‑1 secreta­ gogues, with the effects of insulin seeming to form a positive feedback loop between the L cell and islet β cell.75,76 The concomitant findings that conditions of leptin and insulin resistance impair the L cell response to oral glucose are consistent with reports of reduced GLP‑1 levels in individuals with obesity and insulin resistance and/or type 2 diabetes mellitus, respectively,77–79 and indi­ cate that, like the X/A-like cell, the L cell is dysregulated by perturbations in nutrient homeostasis. Once released into the circulation, GLP‑1 and GLP‑2 are cleaved by dipeptidylpeptidase 4 (DPP‑4), render­ ing short bioactive half-lives of ~1–2 min and ~7 min, respectively. 53,80,81 Oxyntomodulin has intermediate susceptibility to degradation by DPP‑4, at least in vitro, and a DPP‑4-resistant analogue has been reported to demon­ trate enhanced bioactivity compared with the s native peptide in vivo.82,83 In the case of both GLP‑1 and GLP‑2, the cleavage product serves as a partial agonist or antagonist at the respective receptor,84,85 although trun­ cated GLP‑1 also seems to mediate some of the cardio­ protective actions of GLP‑1 through a yet-to-be-identified receptor.86 Finally, PYY is also degraded by DPP‑4,87,88 www.nature.com/nrgastro © 2012 Macmillan Publishers Limited. All rights reserved
  • 5. REVIEWS with the half-lives of both PYY1–36NH2 and PYY3–36NH2 being ~13 min,89 but, in contrast to the inactivation of GLP‑1, GLP‑2 and oxynto­ odulin, DPP‑4-mediated cleavage of m PYY is actually required for its biological activity at the Y2 receptor.90 Thus, the truncated peptide, PYY3–36NH2, mediates the effects of PYY as a satiety factor.87 Together, therefore, GLP‑1, GLP‑2, oxyntomodulin and PYY are all released by the intestinal L cell. However, these L cell hormones have distinct profiles of biological actions, which ultimately integrate at the level of whole body nutrient homeostasis. GLP‑1 As a result of the widespread distribution of the GLP‑1 receptor, in both peripheral tissues and the CNS,91–93 cir­ culating GLP‑1 exerts a diverse array of effects on nutrient homeostasis. The major biological actions of GLP‑1 are antidiabetic, and are most notably exerted through the pancreatic β cell. Hence, GLP‑1 is an incretin that enhances glucose-dependent insulin secretion, as well as stimulat­ ing insulin biosynthesis and, in rodents, promoting β‑cell proliferation while inhibiting apoptosis.94,95 GLP‑1 also inhibits release of glucagon by pancreatic α cells, delays nutrient absorption through inhibition of gastric emptying and intestinal motility, and enhances insulin sensitivity at the level of both the liver and peripheral tissues—although the majority of its effects on glycaemia seem to be medi­ ated through changes in islet hormone secretion.96 The beneficial effects of circulating endogenous and exogenous GLP‑1 or its analogues on regulation of glucose have also been confirmed in studies using both antagonist and receptor knockout approaches.97,98 However, peripherally administered GLP‑1 and its analogues also have anorexi­ genic actions, inducing satiety and inhibiting food intake in normal and obese rodents and humans, as well as in patients with type 2 diabetes mellitus.99–102 As a consequence of the reported biological actions of GLP‑1, several long-acting, DPP‑4-resistant GLP‑1based drugs have now been licensed for clinical use in the treatment of type 2 diabetes mellitus, the major effects of which are a reduction in haemoglobin A1c (HbA1c, a glycated haemoglobin) levels in association with weight loss.103 Of note, although GLP‑1R agonists do induce weight loss in overweight and obese individuals with and without type 2 diabetes mellitus, the overall effect is fairly modest, averaging 2.9 kg.104 However, a GLP‑1– glucagon co-agonist that takes advantage of the inhibi­ tory effects of GLP‑1 on food intake and the stimulatory effects of glucagon on energy expenditure, while at the same time balancing the hypoglycaemic and hyper­ glycaemic effects of the two peptides, respectively, indi­ cates that novel approaches to the use of GLP‑1 could optimize its use in obesity. 105 Conversely, although DPP‑4 inhibitors are used clinically to increase circulat­ ing levels of endo­ enous GLP‑1, as well as of the other g major incretin, GIP, in patients with type 2 diabetes mel­ litus,103 the use of these agents is associated with reduced levels of HbA1c but not weight loss, the latter possibly owing to concomitantly reduced production of bioactive PYY3–36NH2, as discussed above. An alternative approach to the use of GLP‑1 in the treatment of patients with type 2 diabetes mellitus and/or obesity is enhancement of endogenous GLP‑1 secretion by activation of novel L cell secretory pathways and, in particular, those medi­ ating the responses to fatty acids, including GPR40 and GPR119.74,106 Such agents would have the advantage of concomitantly increasing the release of all of the L cell hormones, of which oxynto­ odulin and PYY3–36NH2 are m also anorexigenic (discussed in more detail below). Although GLP‑1 is released into the circulation as a hormone, its short half-life suggests that at least some of its physiological actions might be mediated indirectly, prob­ ably through the GLP‑1R expressed on afferent neurons. Hence, despite the well-established direct effects of GLP‑1 on β cells in vitro,107,108 the in vivo actions of GLP‑1 to stimulate insulin release have been reported to require afferent neurons.92 Indeed, GLP‑1 increases vagal afferent firing in rats.109 However, in direct comparative studies, the requirement for sensory afferents in the actions of GLP‑1 on both insulin release and food intake in mice was observed at low, but not high, doses of GLP‑1.110,111 Whether some of the anorexigenic actions of GLP‑1 are also mediated via suppression of gastric emptying remains unclear.112 Nonetheless, these findings collectively suggest that the physiological effects of endogenously secreted GLP‑1, which includes the actions of GLP‑1 in patients treated with DPP‑4 inhibitors, are mediated indirectly by local effects of GLP‑1 on its receptor expressed by gastro­ intestinal and/or vagal afferent neurons.113 By contrast, the pharmacologic actions of GLP‑1 and long-acting GLP‑1R agonists seem to be exerted, at least in part, through direct effects on the GLP‑1R in β cells and the brain. Genetic studies have demonstrated that poly­ morphisms in the voltage-gated K + channel gene, KCNQ1, are associated with reduced secretion of GLP‑1, whereas variants in the genes for the GLP‑1R, the β‑cell transcription factor, TCF7L2, and the neuroendocrine transmembrane protein, Wolfram syndrome 1, lead to reduced GLP‑1-stimulated insulin release.114–117 Hence, inter­ ndividual differences in GLP‑1 release or func­ i tion might be linked to alterations in the effectiveness of DPP‑4 inhibitors and GLP‑1R agonists, respectively, in patients with type 2 diabetes mellitus, a notion that has yet to be rigorously assessed. GLP‑2 In contrast to GLP‑1, the actions of GLP‑2 are more restricted owing to the highly concentrated expression of the GLP‑2R in the gastrointestinal tract; these receptors are expressed in only a few other tissues, including the vagus nerve and brain.118,119 GLP‑2 is, therefore, known foremost for its effects in the gastrointestinal tract. Both GLP‑2 and its degradation-resistant analogues increase small and large intestinal weight, crypt-villus height and mucosal surface area through stimulation of crypt cell proliferation and inhibition of epithelial cell apop­ tosis.120–122 GLP‑2 also enhances nutrient absorption through various actions, including increasing expression of digestive enzymes and nutrient transporters, as well as stimulating intestinal blood flow.120–122 Of note, although NATURE REVIEWS | GASTROENTEROLOGY HEPATOLOGY © 2012 Macmillan Publishers Limited. All rights reserved VOLUME 9  |  DECEMBER 2012  |  709
  • 6. REVIEWS GLP‑2 also inhibits gastric emptying, these effects are minor compared with the actions of GLP‑1 on gastric emptying.123 Furthermore, in a wide variety of animal models of intestinal damage and insufficiency, GLP‑2 exerts protective effects on the gastrointestinal epithelium by reducing inflammation and increasing barrier func­ tion, as well as by preventing and repairing damage.124–127 However, in contrast to the profound pharmacological actions of GLP‑2, the role of the endogenous hormone seems to be more limited, with only a small or no reduc­ tion in bowel size noted upon administration of a GLP‑2R antagonist and in GLP‑2R null mice.128,129 Nonetheless, endogenous GLP‑2 is required for the intestinal adaptive responses to nutrient ingestion after a prolonged fast, as well as for protection from chemically induced enteri­ tis.85,130 Importantly, the majority of the major findings on GLP‑2 in rodent models have also been confirmed in humans, most notably in patients with intestinal insuffi­ ciency owing to short bowel syndrome or damage associ­ ated with Crohn’s disease.131,132 Clinical trials are ongoing, and a long-acting analogue of GLP‑2 has been approved in Europe for use in patients with short bowel syndrome.133 Despite extensive characterization of the effects of GLP‑2, the mechanistic basis of the action of GLP‑2 is only poorly understood. The GLP‑2R is not localized to the major site of action of GLP‑2 (that is, intestinal epithelial cells) but rather it is localized to the intestinal subepithelial myofibroblasts, enteric nervous system and diffuse entero­ endocrine cells;122 the actions of GLP‑2 must therefore be exerted indirectly by the release of paracrine factors from these cells. Downstream players that have been identified to date include: insulin-like growth factor (IGF)‑1 and IGF‑2, and the ErbB network for crypt cell proliferation in the small and large intestine;130,134–136 keratinocyte growth factor, for large bowel growth;137 vasoactive intestinal poly­ peptide, for anti-inflammatory actions in the small and large intestine;138 and endothelial nitric oxide synthase, for intestinal blood flow.139 Finally, and again in contrast to the actions of GLP‑1, the known effects of GLP‑2 do not seem to be mediated through afferent pathways, as activa­ tion of this receptor is not linked to any change in vagal firing, and centrally administered but not peripherally administered GLP‑2 inhibits food intake.109,140,141 Oxyntomodulin Although oxyntomodulin was originally named for its inhibitory effects on gastric acid secretion,142 these actions are relatively weak compared with those exerted by other gut peptides, such as somatostatin.143 Hence, the biologi­ cal role of oxyntomodulin remained somewhat uncer­ tain until ~10 years ago, when it was reported to inhibit food intake in both rats and humans when administered exogenously.144,145 Indeed, a number of studies have now demonstrated that repeated peripheral administration of either oxyntomodulin or its long-acting analogues induces weight loss in obese rats, mice and humans, with this effect being mediated through both suppression of food intake and increased energy expenditure.146–148 Interestingly, early studies on oxyntomodulin and gastric acid secretion suggested the existence of a receptor 710  |  DECEMBER 2012  |  VOLUME 9 for this hormone through which glucagon might also act, although with less potency.142 Although no specific oxyntomodulin receptor has been identified to date, this hormone has been established to be an agonist for both the GLP‑1R and the glucagon receptor. Hence, oxynto­ modulin has EC50s (half maximal effective concentra­ tions) for the GLP‑1R and glucagon receptor that are approximately 100-fold and 10-fold greater than those of GLP‑1 and glucagon, respectively.144,149,150 Consistent with these findings, the actions of oxyntomodulin on the gastric glands are now known to be mediated through GLP‑1R, as are the effects of peripheral oxyntomodulin to suppress food intake.151,152 However, the stimulatory effect of oxyntomodulin on energy expenditure might also be dependent upon the glucagon receptor, as this is also a well-established effect of glucagon.153,154 Hence, not only DPP‑4-resistant analogues of oxyntomodulin, but also dual GLP‑1R/glucagon receptor oxynto­modulin analogues have been developed as possible thera­ eutic p agents for the treatment of obesity. 83,148,155 Notably, oxynto­modulin also improves glycaemic control, through GLP‑1R-dependent increase of glucose-stimulated insulin secretion.156 Although this effect might be expected to be counter-balanced by oxyntomodulin-induced activation of the glucagon receptor, the available data suggest that the anti-hyperglycaemic effects of oxyntomodulin prevail in the setting of dual receptor activation.153 Furthermore, the mechanism of action of oxyntomodulin also implies that polymorphisms in either the GLP-1R, the glucagon receptor and/or their signalling pathways might influence the effectiveness of these drugs.115–117,157 Finally, the cellular pathway(s) by which oxynto­ modulin inhibits food intake are thought to include both the CNS and the vagus nerve. Peripheral admin­ istration of oxynto­ odulin activates a variety of central m nuclei involved in feeding, including the arcuate nucleus that abuts areas of the hypothalamus characterized by a leaky blood–brain barrier.150,152 Oxyntomodulin also stimulates release of the anorexigenic neuropeptide, α‑melanocyte-stimulating hormone, from hypothalamic explants in vitro, which is consistent with possible direct effects on central feeding centers.152 However, activation of the nucleus of the solitary tract by peripheral oxynto­ modulin is also in keeping with a role for vagal sensing of this hormone.150 Although studies of oxyntomodulin actions on food intake following vagal disruption have yet to be conducted, the inhibitory actions of oxynto­ modulin on pancreatic exocrine secretion are abrogated in vagotomised rats, which indicates that signalling can occur through this peripheral neural pathway.158 PYY Which prohormone convertase cleaves proPYY to PYY1–36NH2 is not known, although it is presumed to be PC1/3 owing to the known actions of this enzyme on pro­ glucagon in the intestinal L cell.50 PYY1–36NH2 was first iden­ tified as a gut peptide with tyrosine (‘Y’) at its N‑terminal and C‑terminal ends, and was initially characterized as an inhibitor of both pancreatic exocrine secretion and intestinal motility.159,160 However, although PYY1–36NH2 www.nature.com/nrgastro © 2012 Macmillan Publishers Limited. All rights reserved
  • 7. REVIEWS binds to multiple receptors of the neuropeptide Y recep­ tor (YR) family, its cleavage by DPP‑4 in the circula­ tion converts it to a Y2-receptor-selective agonist.87,90 Thus, compared with both full-length PYY 1–36NH2 and neuropeptide Y, which stimulate food intake through the Y1R and possibly the Y5R, the Y2R-selectivity of PYY3–36NH2 and its agonists enables suppression of food intake through inhibition of neuropeptide Y release in the arcuate nucleus, while Y2R specific antagonists prevent the anorexigenic actions of PYY3–36NH2.161–164 Consistent with these findings, initial studies in mice have suggested that the actions of PYY3–36NH2 are not mediated by periph­ eral sensory afferent nerves such as the vagus.111 However, in the rat, the Y2R is expressed by the vagus nerve, and peripherally administered PYY3–36NH2 stimulates gastric vagal afferent firing, while vagotomy prevents the anorexi­ genic actions of PYY3–36NH2.165 Thus, the mechanism of action of PYY3–36NH2 might exhibit species-­ pecific dif­ s ferences. Alternatively, as the Y2R expressed by rat vagal afferent neurons is regulated by cholecystokinin in a nutri­ ent-dependent manner,166 experimental differences could also account for these apparent discrepancies. Although great interest was generated by the first reports of the anorexigenic actions of PYY3–36NH2 in rodents as well as normal-weight and obese humans,163,167 these findings have been the subject of some controversy in the literature, with both confirmatory and negative findings reported.111,168,169 The reason for these discrepant findings remains elusive. Nonetheless, both a rare PYY mutation and Y2R variants have been linked to altera­ tions in body weight in humans.170–172 Hence, long-acting PYY3–36NH2 analogues, as well as Y2R agonists, are cur­ rently under development for the treatment of obesity, and many clinical trials are ongoing. Combined effects Although the L cell hormones are co-secreted, few studies have examined the combined effects of simultaneous increased levels of these hormones. With respect to feed­ back suppression of food intake, additive and/or syner­ gistic effects of PYY3–36NH2 with oxyntomodulin, as well as of PYY3–36NH2 with GLP‑1 or its analogues have been reported.111,173–175 Furthermore, PYY3–36NH2 and GLP‑1 also have additive effects to delay gastric emptying.176 Finally, GLP‑1 and GLP‑2 do not demonstrate additive effects to suppress food intake,177 but do exert opposing effects on glucagon release, with the inhibitory effect of GLP‑1 predominating.178 No studies to date have examined the physiological consequences of increasing the levels of all four of these hormones, or of their effects when given in combination with all of the other gut hormones released by nutrient intake. Such investigations will clearly be nec­ essary to fully understand the integrated contribution of the L cell to nutrient homeostasis, particularly if L cell secretagogues are to be used for the treatment of type 2 diabetes mellitus and/or obesity, as discussed above. Clinical relevance Two successful, surgical treatments for obesity—Rouxen‑Y gastric bypass (RYGB) and sleeve gastrectomy—also improve or reverse the symptoms of type 2 diabetes mel­ litus. RYGB markedly reduces the size of the stomach and diverts ingested nutrients and pancreato-biliary flow directly to the distal small intestine, whereas sleeve gas­ trectomy introduces gastric restriction only.179 Despite the differences between these surgical interventions, the results of clinical trials indicate that both proce­ dures result in equivalent improvements in glycaemic control and identical weight loss 1 year after surgery.180,181 Remarkably, the reductions in glucose levels occur rapidly after surgery, often prior to the changes in body weight.182 The success of RYGB for both resolution of type 2 diabetes mellitus and weight loss has largely been attributed to an increase in the release of peptides from the L cells, with the effects to reduce ghrelin release being controversial.181,183–187 The concomitant diversion of bile flow to the distal small intestine might also contribute to the increase in L cell secretion following RYGB through activation of the L cell bile acid receptor, TGR5.69 Indeed, a study has demonstrated that enhanced release of GLP‑1 following gastric bypass in obese rats is responsible for the weight-loss-independent improvement in glucose tolerance associated with this procedure.188 By contrast, sleeve gastrectomy has been reported to cause greater decreases in ghrelin levels and increased cholecystokinin release than found with RYGB, in addition to increasing L cell secretion.181 Thus, although differing somewhat in their mechanisms of action, RYGB and sleeve gastrec­ tomy are unique approaches that manipulate the physio­ logical actions of the gut hormones to regulate nutrient homeostasis through their combined effects on gut nutrient handling, glucose tolerance and food intake. Conclusion This Review has focused on the hormones produced by the enteroendocrine X/A-like cell (ghrelin) and the L cell (GLP‑1, GLP‑2, oxyntomodulin and PYY). These peptides have key roles in the integration of food intake with metabolism, as well as providing feedback signals to the brain to induce satiety during feeding. However, it is recognized that the gastrointestinal tract releases many other hormones that also have important roles in nutri­ ent homeostasis including, most notably, GIP and chol­ ecystokinin. Elucidation of the combined actions of all of these hormones will be essential to develop a comprehen­ sive understanding of the integrative role that the gastro­ intestinal tract has in maintaining nutrient homeostasis during fasting and in response to feeding. Review criteria PubMed was searched for full-length articles using the keywords “ghrelin”, “GLP‑1”, “GLP‑2”, “oxyntomodulin”, “enteroglucagon” and “PYY”, alone and in combination with key author names and/or the keywords “secretion”, “food intake”, “obesity”, “insulin resistance”, “type 2 diabetes” and/or “review”; review articles were used to further search for original papers. All years were included in the search. Articles published in languages other than English were excluded from the analysis. NATURE REVIEWS | GASTROENTEROLOGY HEPATOLOGY © 2012 Macmillan Publishers Limited. All rights reserved VOLUME 9  |  DECEMBER 2012  |  711
  • 8. REVIEWS 1. 2. 3. 4. 5. 6. 7. 8. 9. 10. 11. 12. 13. 14. 15. 16. 17. 18. 19. 20. 21. Rehfeld, J. F. A centenary of gastrointestinal endocrinology. Horm. Metab. Res. 36, 735–741 (2004). Ferri, G.‑L. et al. Tissue localization and relative distribution of regulatory peptides in separated layers from the human bowel. Gastroenterology 84, 777–786 (1983). Larsson, L. I. Peptide secretory pathways in GI tract: cytochemical contributions to regulatory physiology of the gut. Am. J. Physiol. 239, G237–G246 (1980). Anini, Y., Hansotia, T. Brubaker, P Muscarinic . L. receptors control postprandial release of glucagon-like peptide‑1: in vivo and in vitro studies in rats. Endocrinology 143, 2420–2426 (2002). Bohorquez, D. V. Liddle, R. A. Axon-like basal processes in enteroendocrine cells: characteristics and potential targets. Clin. Transl. Sci. 4, 387–391 (2011). Dockray, G. J. The versatility of the vagus. Physiol. Behav. 97, 531–536 (2009). Reimann, F., Tolhurst, G. Gribble, F. M. G‑protein‑coupled receptors in intestinal chemosensation. Cell Metab. 15, 421–431 (2012). Dockray, G. J. Cholecystokinin. Curr. Opin. Endocrinol. Diabetes Obes. 19, 8–12 (2012). Cho, Y. M. Kieffer, T. J. K‑cells and glucosedependent insulinotropic polypeptide in health and disease. Vitam. Horm. 84, 111–150 (2010). Ali, S., Lamont, B. J., Charron, M. J. Drucker, D. J. Dual elimination of the glucagon and GLP‑1 receptors in mice reveals plasticity in the incretin axis. J. Clin. Invest. 121, 1917–1929 (2011). Habib, A. M. et al. Overlap of endocrine hormone expression in the mouse intestine revealed by transcriptional profiling and flow cytometry. Endocrinology 153, 3054–3065 (2012). Kojima, M. et al. Ghrelin is a growth‑hormone‑releasing acylated peptide from stomach. Nature 402, 656–660 (1999). Date, Y. et al. Ghrelin, a novel growth hormonereleasing acylated peptide, is synthesized in a distinct endocrine cell type in the gastrointestinal tracts of rats and humans. Endocrinology 141, 4255–4261 (2000). Rindi, G. et al. Characterisation of gastric ghrelin cells in man and other mammals: studies in adult and fetal tissues. Histochem. Cell Biol. 117, 511–519 (2002). Sakata, I. et al. Ghrelin-producing cells exist as two types of cells, closed- and opened-type cells, in the rat gastrointestinal tract. Peptides 23, 531–536 (2002). Zhu, X., Cao, Y., Voogd, K. Steiner, D. F. On the processing of proghrelin to ghrelin. J. Biol. Chem. 281, 38867–38870 (2006). Kirchner, H. et al. GOAT links dietary lipids with the endocrine control of energy balance. Nat. Med. 15, 741–745 (2009). Tschop, M., Smiley, D. L. Heiman, M. L. Ghrelin induces adiposity in rodents. Nature 407, 908–913 (2000). Cummings, D. E. et al. A preprandial rise in plasma ghrelin levels suggests a role in meal initiation in humans. Diabetes 50, 1714–1719 (2001). Mundinger, T. O., Cummings, D. E. Taborsky, G. J. Jr. Direct stimulation of ghrelin secretion by sympathetic nerves. Endocrinology 147, 2893–2901 (2006). Ao, Y. et al. Brainstem thyrotropin-releasing hormone regulates food intake through vagaldependent cholinergic stimulation of ghrelin secretion. Endocrinology 147, 6004–6010 (2006). 712  |  DECEMBER 2012  |  VOLUME 9 22. Shimada, M. et al. Somatostatin suppresses ghrelin secretion from the rat stomach. Biochem. Biophys. Res. Commun. 302, 520–525 (2003). 23. McCowen, K. C., Maykel, J. A., Bistrian, B. R. Ling, P . R. Circulating ghrelin concentrations are lowered by intravenous glucose or hyperinsulinemic euglycemic conditions in rodents. J. Endocrinol. 175, R7–R11 (2002). 24. Saad, M. F. et al. Insulin regulates plasma ghrelin concentration. J. Clin. Endocrinol. Metab. 87, 3997–4000 (2002). 25. Sakata, I. et al. Glucose-mediated control of ghrelin release from primary cultures of gastric mucosal cells. Am. J. Physiol. Endocrinol. Metab. 302, E1300–E1310 (2012). 26. Gagnon, J. Anini, Y. Insulin and norepinephrine regulate ghrelin secretion from a rat primary stomach cell culture. Endocrinology 153, 3646–3656 (2012). 27. Taborsky, G. J. Jr. The physiology of glucagon. J. Diabetes Sci. Technol. 4, 1338–1344 (2010). 28. Walker, J. N. et al. Regulation of glucagon secretion by glucose: paracrine, intrinsic or both? Diabetes Obes. Metab. 13 (Suppl. 1), 95–105 (2011). 29. Kordowich, S., Collombat, P Mansouri, A. ., Serup, P Arx and Nkx2.2 compound deficiency . redirects pancreatic alpha- and beta-cell differentiation to a somatostatin/ghrelin coexpressing cell lineage. BMC Dev. Biol. 11, 52–67 (2011). 30. Ariyasu, H. et al. A postweaning reduction in circulating ghrelin temporarily alters growth hormone (GH) responsiveness to GH‑releasing hormone in male mice but does not affect somatic growth. Endocrinology 151, 1743–1750 (2010). 31. Sakata, I. et al. Characterization of a novel ghrelin cell reporter mouse. Regul. Pept. 155, 91–98 (2009). 32. Esler, W. P et al. Small-molecule ghrelin receptor . antagonists improve glucose tolerance, suppress appetite, and promote weight loss. Endocrinology 148, 5175–5185 (2007). 33. Pfluger, P et al. Simultaneous deletion of . T. ghrelin and its receptor increases motor activity and energy expenditure. Am. J. Physiol. Gastrointest. Liver Physiol. 294, G610–G618 (2008). 34. Kirchner, H., Tong, J., Tschop, M. H. Pfluger, P Ghrelin and PYY in the regulation of . T. energy balance and metabolism: lessons from mouse mutants. Am. J. Physiol. Endocrinol. Metab. 298, E909–E919 (2010). 35. Barnett, B. P et al. Glucose and weight control in . mice with a designed ghrelin O‑acyltransferase inhibitor. Science 330, 1689–1692 (2010). 36. Stengel, A. Tache, Y. Ghrelin—a pleiotropic hormone secreted from endocrine x/a-like cells of the stomach. Front. Neurosci. 6, 24 (2012). 37. Banks, W. A., Tschop, M., Robinson, S. M. Heiman, M. L. Extent and direction of ghrelin transport across the blood–brain barrier is determined by its unique primary structure. J. Pharmacol. Exp. Ther. 302, 822–827 (2002). 38. Toshinai, K. et al. Ghrelin-induced food intake is mediated via the orexin pathway. Endocrinology 144, 1506–1512 (2003). 39. Date, Y. et al. The role of the gastric afferent vagal nerve in ghrelin-induced feeding and growth hormone secretion in rats. Gastroenterology 123, 1120–1128 (2002). 40. Gnanapavan, S. et al. The tissue distribution of the mRNA of ghrelin and subtypes of its receptor, GHS‑R, in humans. J. Clin. Endocrinol. Metab. 87, 2988–2991 (2002). 41. Zhao, T. J. et al. Ghrelin O‑acyltransferase (GOAT) is essential for growth hormone-mediated 42. 43. 44. 45. 46. 47. 48. 49. 50. 51. 52. 53. 54. 55. 56. 57. 58. 59. survival of calorie-restricted mice. Proc. Natl Acad. Sci. USA 107, 7467–7472 (2010). Tschop, M. et al. Circulating ghrelin levels are decreased in human obesity. Diabetes 50, 707–709 (2001). Shiiya, T. et al. Plasma ghrelin levels in lean and obese humans and the effect of glucose on ghrelin secretion. J. Clin. Endocrinol. Metab. 87, 240–244 (2002). Marzullo, P. et al. The relationship between active ghrelin levels and human obesity involves alterations in resting energy expenditure. J. Clin. Endocrinol. Metab. 89, 936–939 (2004). McLaughlin, T., Abbasi, F., Lamendola, C., Frayo, R. S. Cummings, D. E. Plasma ghrelin concentrations are decreased in insulinresistant obese adults relative to equally obese insulin-sensitive controls. J. Clin. Endocrinol. Metab. 89, 1630–1635 (2004). Ukkola, O. et al. Role of ghrelin polymorphisms in obesity based on three different studies. Obes. Res. 10, 782–791 (2002). Otto, B. et al. Weight gain decreases elevated plasma ghrelin concentrations of patients with anorexia nervosa. Eur. J. Endocrinol. 145, 669–673 (2001). Akamizu, T. Kangawa, K. The physiological significance and potential clinical applications of ghrelin. Eur. J. Intern. Med. 23, 197–202 (2012). Patterson, M., Bloom, S. R. Gardiner, J. V. Ghrelin and appetite control in humans-potential application in the treatment of obesity. Peptides 32, 2290–2294 (2011). Dhanvantari, S., Seidah, N. G. Brubaker, P. L. Role of prohormone convertases in the tissuespecific processing of proglucagon. Mol. Endocrinol. 10, 342–355 (1996). Anini, Y. et al. Comparison of the postprandial release of peptide YY and proglucagon-derived peptides in the rat. Pflugers Arch. 438, 299–306 (1999). Orskov, C. et al. Glucagon-like peptides GLP‑1 and GLP‑2, predicted products of the glucagon gene, are secreted separately from pig small intestine but not pancreas. Endocrinology 119, 1467–1475 (1986). Larsson, L.‑I., Holst, J., Hakanson, R. Sundler, F. Distribution and properties of glucagon immunoreactivity in the digestive tract of various mammals: an immunohistochemical and immunochemical study. Histochemistry 44, 281–290 (1975). Eissele, R. et al. Glucagon-like peptide‑1 cells in the gastrointestinal tract and pancreas of rat, pig and man. Eur. J. Clin. Invest. 22, 283–291 (1992). Reimann, F. et al. Glucose sensing in L cells: a primary cell study. Cell Metab. 8, 532–539 (2008). Geraedts, M. C. et al. Transformation of postingestive glucose responses after deletion of sweet taste receptor subunits or gastric bypass surgery. Am. J. Physiol. Endocrinol. Metab. 303, E464–E474 (2012). Tolhurst, G. et al. Short-chain fatty acids stimulate glucagon-like peptide‑1 secretion via the G‑protein‑coupled receptor FFAR2. Diabetes 61, 364–371 (2012). Xiao, Q., Boushey, R., Drucker, D. J. Brubaker, P. L. Secretion of the intestinotropic hormone glucagon-like peptide‑2 is differentially regulated by nutrients in humans. Gastroenterology 117, 99–105 (1999). Anini, Y. Brubaker, P. L. Muscaranic receptors control glucagon-like peptide 1 secretion by human endocrine L cells. Endocrinology 144, 3244–3250 (2003). www.nature.com/nrgastro © 2012 Macmillan Publishers Limited. All rights reserved
  • 9. REVIEWS 60. Balks, H. J., Holst, J. J., Von zur Mühlen, A. Brabant, G. Rapid oscillations in plasma glucagon-like peptide‑1 (GLP‑ 1) in humans: cholinergic control of GLP‑1 secretion via muscarinic receptors. J. Clin. Endocrinol. Metab. 82, 786–790 (1997). 61. Roberge, J. N. Brubaker, P Secretion of . L. proglucagon-derived peptides in response to intestinal luminal nutrients. Endocrinology 128, 3169–3174 (1991). 62. Parker, H. E. Predominant role of active versus facilitative glucose transport for glucagon-like peptide-1 secretion. Diabetologia 55, 2445–2455 (2012). 63. Gribble, F. M., Williams, L., Simpson, A. K. Reimann, F. A novel glucose-sensing mechanism contributing to glucagon-like peptide‑1 secretion from the GLUTag cell line. Diabetes 52, 1147–1154 (2003). 64. Iakoubov, R., Lauffer, L., Ahmed, A., Bazinet, R. P . Brubaker, P Essential role for protein kinase . L. C zeta in oleic-acid induced glucagon-like peptide‑1 secretion in vivo in the rat. Endocrinology 152, 1244–1252 (2011). 65. Poreba, M. A. et al. Role of fatty acid transport protein 4 in oleic acid-induced glucagon-like peptide‑1 secretion. Am. J. Physiol. Endocrinol. Metab. http://dx.doi.org/10.1152/ ajpendo.00116.2012. 66. Hirasawa, A. et al. Free fatty acids regulate gut incretin glucagon-like peptide‑1 secretion through GPR120. Nat. Med. 11, 90–94 (2005). 67. Edfalk, S., Steneberg, P Edlund, H. Gpr40 is . expressed in enteroendocrine cells and mediates FFA stimulation of incretin secretion. Diabetes 57, 2280–2287 (2008). 68. Lauffer, L. M., Iakoubov, R. Brubaker, P . L. GPR119 is essential for oleoylethanolamideinduced glucagon-like peptide‑1 secretion from the intestinal enteroendocrine L‑cell. Diabetes 58, 1058–1066 (2009). 69. Thomas, C. et al. TGR5-mediated bile acid sensing controls glucose homeostasis. Cell Metab. 10, 167–177 (2009). 70. Hansen, K. B. et al. 2‑Oleoyl glycerol is a GPR119 agonist and signals GLP‑1 release in humans. J. Clin. Endocrinol. Metab. 96, E1409–E1417 (2011). 71. Rocca, A. S., LaGreca, J., Kalitsky, J. Brubaker, P Monounsaturated fatty acids . L. improve glycemic tolerance through increased secretion of glucagon-like peptide‑1. Endocrinology 142, 1148–1155 (2001). 72. Thomsen, C. et al. Differential effects of saturated and monounsaturated fatty acids on postprandial lipemia and incretin responses in healthy subjects. Am. J. Clin. Nutr. 69, 1135–1143 (1999). 73. Lim, G. E. Brubaker, P Glucagon-like . L. peptide‑1 secretion by the L cell: a view from the inside. Diabetes 55, S70–S77 (2006). 74. Ahren, B. Islet G protein-coupled receptors as potential targets for treatment of type 2 diabetes. Nat. Rev. Drug Discov. 8, 369–385 (2009). 75. Anini, Y. Brubaker, P Role of leptin in the . L. regulation of glucagon-like peptide‑1 secretion. Diabetes 52, 252–259 (2003). 76. Lim, G. E. et al. Insulin regulates glucagon-like peptide‑1 secretion from the enteroendocrine L cell. Endocrinology 150, 580–591 (2009). 77. Rask, E. et al. Impaired incretin response after a mixed meal is associated with insulin resistance in nondiabetic men. Diabetes Care 24, 1640–1645 (2001). 78. Vilsboll, T., Krarup, T., Deacon, C. F., Madsbad, S. Holst, J. J. Reduced postprandial concentrations of intact biologically active 79. 80. 81. 82. 83. 84. 85. 86. 87. 88. 89. 90. 91. 92. 93. glucagon-like peptide 1 in type 2 diabetic patients. Diabetes 50, 609–613 (2001). Mannucci, E. et al. Glucagon-like peptide (GLP)‑1 and leptin concentrations in obese patients with Type 2 diabetes mellitus. Diabetes Med. 17, 713–719 (2000). Vilsboll, T., Agerso, H., Krarup, T. Holst, J. J. Similar elimination rates of glucagon-like peptide‑1 in obese type 2 diabetic patients and healthy subjects. J. Clin. Endocrinol. Metab. 88, 220–224 (2003). Tavares, W., Drucker, D. J. Brubaker, P . L. Enzymatic- and renal-dependent catabolism of the intestinotropic hormone glucagon-like peptide‑2 in rats. Am. J. Physiol. Endocrinol. Metab. 278, E134–E139 (2000). Zhu, L. et al. The role of dipeptidyl peptidase IV in the cleavage of glucagon family peptides: in vivo metabolism of pituitary adenylate cyclase activating polypeptide‑(1–38). J. Biol. Chem. 278, 22418–22423 (2003). Kerr, B. D., Flatt, P Gault, V. A. (D-Ser2) . R. Oxm[mPEG-PAL]: a novel chemically modified analogue of oxyntomodulin with antihyperglycaemic, insulinotropic and anorexigenic actions. Biochem. Pharmacol. 80, 1727–1735 (2010). Knudsen, L. B. Pridal, L. Glucagon-like peptide‑1‑(9–36) amide is a major metabolite of glucagon-like peptide‑1‑(7–36) amide after in vivo administration to dogs, and it acts as an antagonist on the pancreatic receptor. Eur. J. Pharmacol. 318, 429–435 (1996). Shin, E. D., Estall, J. L., Izzo, A., Drucker, D. J. Brubaker, P Mucosal adaptation to enteral . L. nutrients is dependent on the physiologic actions of glucagon-like peptide‑2 in mice. Gastroenterology 128, 1340–1353 (2005). Ban, K. et al. Cardioprotective and vasodilatory actions of glucagon-like peptide 1 receptor are mediated through both glucagon-like peptide 1 receptor-dependent and -independent pathways. Circulation 117, 2340–2350 (2008). Medeiros, M. D. Turner, A. J. Processing and metabolism of peptide-YY: pivotal roles of dipeptidylpeptidase-IV, aminopeptidase‑P and , endopeptidase‑24.11. Endocrinology 134, 2088–2094 (1994). Aaboe, K. et al. Twelve weeks treatment with the DPP‑4 inhibitor, sitagliptin, prevents degradation of peptide YY and improves glucose and nonglucose induced insulin secretion in patients with type 2 diabetes mellitus. Diabetes Obes. Metab. 12, 323–333 (2010). Yoshinaga, K. et al. Structural requirements of peptide YY for biological activity at enteric sites. Am. J. Physiol. 263, G695–G701 (1992). Dumont, Y., Fournier, A., St-Pierre, S. Quirion, R. Characterization of neuropeptide Y binding sites in rat brain membrane preparations using [125I] [Leu31, Pro34]peptide YY and [125I]peptide YY3–36 as selective Y1 and Y2 radioligands. J. Pharmacol. Exp. Ther. 272, 673–680 (1995). Campos, R. V., Lee, Y. C. Drucker, D. J. Divergent tissue-specific and developmental expression of receptors for glucagon and glucagon-like peptide‑1 in the mouse. Endocrinology 134, 2156–2164 (1994). Vahl, T. P et al. Glucagon-like peptide‑1 (GLP‑1) . receptors expressed on nerve terminals in the portal vein mediate the effects of endogenous GLP‑1 on glucose tolerance in rats. Endocrinology 148, 4965–4973 (2007). Merchenthaler, I., Lane, M. Shughrue, P . Distribution of pre‑pro‑glucagon and glucagon-like peptide‑1 receptor messenger RNAs in the rat central nervous system. J. Comp. Neurol. 403, 261–280 (1999). NATURE REVIEWS | GASTROENTEROLOGY HEPATOLOGY © 2012 Macmillan Publishers Limited. All rights reserved 94. Baggio, L. L. Drucker, D. J. Biology of incretins: GLP‑1 and GIP. Gastroenterology 132, 2131–2157 (2007). 95. Brubaker, P. L. Minireview: update on incretin biology: focus on glucagon-like peptide‑1. Endocrinology 151, 1984–1989 (2010). 96. Salehi, M., Vahl, T. P. D’Alessio, D. A. Regulation of islet hormone release and gastric emptying by endogenous glucagon-like peptide 1 after glucose ingestion. J. Clin. Endocrinol. Metab. 93, 4909–4916 (2008). 97. Edwards, C. M. et al. Glucagon-like peptide 1 has a physiological role in the control of postprandial glucose in humans: studies with the antagonist exendin 9–39. Diabetes 48, 86–93 (1999). 98. Scrocchi, L. A. et al. Glucose intolerance but normal satiety in mice with a null mutation in the glucagon-like peptide 1 receptor gene. Nat. Med. 2, 1254–1258 (1996). 99. Flint, A., Raben, A., Astrup, A. Holst, J. J. Glucagon-like peptide 1 promotes satiety and suppresses energy intake in humans. J. Clin. Invest. 101, 515–520 (1998). 100. Naslund, E. et al. Energy intake and appetite are suppressed by glucagon-like peptide‑1 (GLP‑1) in obese men. Int. J. Obes. Relat. Metab. Disord. 23, 304–311 (1999). 101. Rodriquez, D. F. et al. Peripheral versus central effects of glucagon-like peptide‑1 receptor agonists on satiety and body weight loss in Zucker obese rats. Metabolism 49, 709–717 (2000). 102. Gutzwiller, J. P. et al. Glucagon-like peptide‑1 promotes satiety and reduces food intake in patients with diabetes mellitus type 2. Am. J. Physiol. Regul. Integr. Comp. Physiol. 276, R1541–R1544 (1999). 103. Lovshin, J. A. Drucker, D. J. Incretin-based therapies for type 2 diabetes mellitus. Nat. Rev. Endocrinol. 5, 262–269 (2009). 104. Vilsboll, T., Christensen, M., Junker, A. E., Knop, F. K. Gluud, L. L. Effects of glucagonlike peptide‑1 receptor agonists on weight loss: systematic review and meta-analyses of randomised controlled trials. BMJ 344, d7771 (2012). 105. Day, J. W. et al. A new glucagon and GLP‑1 coagonist eliminates obesity in rodents. Nat. Chem. Biol. 5, 749–757 (2009). 106. Hughes, T. E. Emerging therapies for metabolic diseases—the focus is on diabetes and obesity. Curr. Opin. Chem. Biol. 13, 332–337 (2009). 107. Thorens, B. Expression cloning of the pancreatic β cell receptor for the gluco-incretin hormone glucagon-like peptide 1. Proc. Natl Acad. Sci. USA 89, 8641–8645 (1992). 108. Huypens, P., Ling, Z., Pipeleers, D. Schuit, F. Glucagon receptors on human islet cells contribute to glucose competence of insulin release. Diabetologia 43, 1012–1019 (2000). 109. Bucinskaite, V. et al. Receptor-mediated activation of gastric vagal afferents by glucagonlike peptide‑1 in the rat. Neurogastroenterol. Motil. 21, 978–e78 (2009). 110. Ahren, B. Sensory nerves contribute to insulin secretion by glucagon-like peptide‑1 in mice. Am. J. Physiol Regul. Integr. Comp. Physiol. 286, R269–R272 (2004). 111. Talsania, T., Anini, Y., Siu, S., Drucker, D. J. Brubaker, P. L. Peripheral exendin‑4 and peptide YY(3–36) synergistically reduce food intake through different mechanisms in mice. Endocrinology 146, 3748–3756 (2005). 112. Jelsing, J. et al. Liraglutide: short-lived effect on gastric emptying-long lasting effects on body weight. Diabetes Obes. Metab. 14, 531–538 (2012). VOLUME 9  |  DECEMBER 2012  |  713
  • 10. REVIEWS 113. Waget, A. et al. Physiological and pharmacological mechanisms through which the DPP‑4 inhibitor sitagliptin regulates glycemia in mice. Endocrinology 152, 3018–3029 (2011). 114. Mussig, K. et al. Association of type 2 diabetes candidate polymorphisms in KCNQ1 with incretin and insulin secretion. Diabetes 58, 1715–1720 (2009). 115. Beinborn, M., Worrall, C. I., McBride, E. W. Kopin, A. S. A human glucagon-like peptide‑1 receptor polymorphism results in reduced agonist responsiveness. Regul. Pept. 130, 1–6 (2005). 116. Schafer, S. A. et al. Impaired glucagon-like peptide‑1‑induced insulin secretion in carriers of transcription factor 7‑like 2 (TCF7L2) gene polymorphisms. Diabetologia 50, 2443–2450 (2007). 117. Schafer, S. A. et al. A common genetic variant in WFS1 determines impaired glucagon-like peptide‑1‑induced insulin secretion. Diabetologia 52, 1075–1082 (2009). 118. Munroe, D. G. et al. Prototypic G protein-coupled receptor for the intestinotrophic factor glucagonlike peptide 2. Proc. Natl Acad. Sci. USA 96, 1569–1573 (1999). 119. Nelson, D. W., Sharp, J. W., Brownfield, M. S., Raybould, H. E. Ney, D. M. Localization and activation of glucagon-like peptide‑2 receptors on vagal afferents in the rat. Endocrinology 148, 1954–1962 (2007). 120. Drucker, D. J., Ehrlich, P Asa, S. L. ., Brubaker, P Induction of intestinal epithelial . L. proliferation by glucagon-like peptide 2. Proc. Natl Acad. Sci. USA 93, 7911–7916 (1996). 121. Drucker, D. J. Biologic actions and therapeutic potential of the proglucagon-derived peptides. Nat. Clin. Pract. Endocrinol. Metab. 1, 22–31 (2005). 122. Rowland, K. J. Brubaker, P The “cryptic” . L. mechanism of action of glucagon-like peptide‑2. Am. J. Physiol. Gastrointest. Liver Physiol. 301, G1–G8 (2011). 123. Nagell, C. F., Wettergren, A., Pedersen, J. F., Mortensen, D. Holst, J. J. Glucagon-like peptide‑2 inhibits antral emptying in man, but is not as potent as glucagon-like peptide‑1. Scand. J. Gastroenterol. 39, 353–358 (2004). 124. Boushey, R. P Yusta, B. Drucker, D. J. ., Glucagon-like peptide 2 decreases mortality and reduces the severity of indomethacin-induced murine enteritis. Am. J. Physiol. Endocrinol. Metab. 277, E937–E947 (1999). 125. Boushey, R. P Yusta, B. Drucker, D. J. ., Glucagon-like peptide (GLP)‑2 reduces chemotherapy-associated mortality and enhances cell survival in cells expressing a transfected GLP‑2 receptor. Cancer Res. 61, 687–693 (2001). 126. L’Heureux, M.‑C. Brubaker, P Glucagon-like . L. peptide‑2 and common therapeutics in a murine model of ulcerative colitis. J. Pharm. Exp. Ther. 306, 347–354 (2003). 127. Martin, G. R., Wallace, L. E. Sigalet, D. L. Glucagon-like peptide‑2 induces intestinal adaptation in parenterally fed rats with short bowel syndrome. Am. J. Physiol. Gastrointest. Liver Physiol. 286, G964–G972 (2004). 128. Shin, E. D., Drucker, D. J. Brubaker, P . L. Glucagon-like peptide‑2: an update. Curr. Opin. Endocrinol. Diab. 12, 63–71 (2005). 129. Lee, S. J. et al. Disruption of the murine Glp2r impairs paneth cell function and increases susceptibility to small bowel enteritis. Endocrinology 153, 1141–1151 (2012). 130. Bahrami, J., Yusta, B. Drucker, D. J. ErbB activity links the glucagon-like peptide‑2 receptor to refeeding-induced adaptation in the murine 714  |  DECEMBER 2012  |  VOLUME 9 small bowel. Gastroenterology 138, 2447–2456 (2010). 131. Jeppesen, P. B. et al. Randomised placebocontrolled trial of teduglutide in reducing parenteral nutrition and/or intravenous fluid requirements in patients with short bowel syndrome. Gut 60, 902–914 (2011). 132. Buchman, A. L., Katz, S., Fang, J. C., Bernstein, C. N. Abou-Assi, S. G. Teduglutide, a novel mucosally active analog of glucagonlike peptide‑2 (GLP‑2) for the treatment of moderate to severe Crohn’s disease. Inflamm. Bowel. Dis. 16, 962–973 (2010). 133. R D Pipeline. NPS Pharmaceuticals [online], http://www.npsp.com/page/rdpipeline/index. v3page;jsessionid=1t4k7y0l3lntn (2012). 134. Dube, P. E., Forse, C. L., Bahrami, J. Brubaker, P. L. Essential role of insulin-like growth factor‑1 in the intestinal tropic effects of glucagon-like peptide‑2 in mice. Gastroenterology 131, 589–605 (2006). 135. Rowland, K. J. et al. Loss of glucagon-like peptide‑2‑induced proliferation following intestinal epithelial insulin-like growth factor‑1 receptor deletion. Gastroenterology 141, 2166–2175 (2011). 136. Yusta, B. et al. ErbB signaling is required for the proliferative actions of GLP‑2 in the murine gut. Gastroenterology 137, 986–996 (2009). 137. Orskov, C. et al. GLP‑2 stimulates colonic growth via KGF, released by subepithelial myofibroblasts with GLP‑2 receptors. Regul. Pept. 124, 105–112 (2005). 138. Sigalet, D. L. et al. Enteric neural pathways mediate the anti-inflammatory actions of glucagon-like peptide 2. Am. J. Physiol. Gastrointest. Liver Physiol. 293, G211–G221 (2007). 139. Guan, X. et al. GLP‑2 receptor localizes to enteric neurons and endocrine cells expressing vasoactive peptides and mediates increased blood flow. Gastroenterology 130, 150–164 (2006). 140. Lovshin, J., Estall, J., Yusta, B., Brown, T. J. Drucker, D. J. Glucagon-like peptide (GLP)‑2 action in the murine central nervous system is enhanced by elimination of GLP‑1 receptor signaling. J. Biol. Chem. 276, 21489–21499 (2001). 141. Schmidt, P. T. et al. Peripheral administration of GLP‑2 to humans has no effect on gastric emptying or satiety. Regul. Pept. 116, 21–25 (2003). 142. Bataille, D., Gespach, C., Coudray, A. M. Rosselin, G. ‘Enteroglucagon’: A specific effect on gastric glands isolated from the rat fundus. Evidence for an ‘oxyntomodulin’ action. Biosci. Rep. 1, 151–155 (1981). 143. Greenberg, G. R., Fung, L. Pokol-Daniel, S. Regulation of somatostatin‑14 and ‑28 secretion by gastric acid in dogs: differential role of cholecystokinin. Gastroenterology 105, 1387–1395 (1993). 144. Dakin, C. L. et al. Oxyntomodulin inhibits food intake in the rat. Endocrinology 142, 4244–4250 (2001). 145. Cohen, M. A. et al. Oxyntomodulin suppresses appetite and reduces food intake in humans. J. Clin. Endocrinol. Metab. 88, 4696–4701 (2003). 146. Wynne, K. et al. Subcutaneous oxyntomodulin reduces body weight in overweight and obese subjects: a double-blind, randomized, controlled trial. Diabetes 54, 2390–2395 (2005). 147. Wynne, K. et al. Oxyntomodulin increases energy expenditure in addition to decreasing energy intake in overweight and obese humans: a randomised controlled trial. Int. J. Obes. (Lond.) 30, 1729–1736 (2006). 148. Liu, Y. L. et al. Subcutaneous oxyntomodulin analogue administration reduces body weight in lean and obese rodents. Int. J. Obes. (Lond.) 34, 1715–1725 (2010). 149. MacNeil, D. J., Occi, J. L., Hey, P . J., Strader, C. D. Graziano, M. P Cloning and expression of a . human glucagon receptor. Biochem. Biophys. Res. Commun. 198, 328–334 (1994). 150. Baggio, L. L., Huang, Q., Brown, T. J. Drucker, D. J. Oxyntomodulin and glucagon-like peptide‑1 differentially regulate murine food intake and energy expenditure. Gastroenterology 127, 546–558 (2004). 151. Schepp, W. et al. Oxyntomodulin: a cAMPdependent stimulus of rat parietal cell function via the receptor for glucagon-like peptide‑ 1 (7–36)NH2. Digestion 57, 398–405 (1996). 152. Dakin, C. L. et al. Peripheral oxyntomodulin reduces food intake and body weight gain in rats. Endocrinology 145, 2687–2695 (2004). 153. Kosinski, J. R. et al. The glucagon receptor is involved in mediating the body weight-lowering effects of oxyntomodulin. Obesity (Silver Spring) 20, 1566–1571 (2012). 154. Habegger, K. M. et al. The metabolic actions of glucagon revisited. Nat. Rev. Endocrinol. 6, 689–697 (2010). 155. Pocai, A. et al. Glucagon-like peptide 1/glucagon receptor dual agonism reverses obesity in mice. Diabetes 58, 2258–2266 (2009). 156. Maida, A., Lovshin, J. A., Baggio, L. L. Drucker, D. J. The glucagon-like peptide‑1 receptor agonist oxyntomodulin enhances betacell function but does not inhibit gastric emptying in mice. Endocrinology 149, 5670–5678 (2008). 157. Hager, J. et al. A missense mutation in the glucagon receptor gene is associated with non‑insulin‑dependent diabetes mellitus. Nat. Genet. 9, 299–304 (1995). 158. Anini, Y. et al. Oxyntomodulin inhibits pancreatic secretion through the nervous system in rats. Pancreas 20, 348–360 (2000). 159. Tatemoto, K. Isolation and characterization of peptide YY (PYY), a candidate gut hormone that inhibits pancreatic exocrine secretion. Proc. Natl Acad. Sci. USA 79, 2514–2518 (1982). 160. Lundberg, J. M. et al. Localization of peptide YY (PYY) in gastrointestinal endocrine cells and effects on intestinal blood flow and motility. Proc. Natl Acad. Sci. USA 79, 4471–4475 (1982). 161. Kanatani, A. et al. Role of the Y1 receptor in the regulation of neuropeptide Y‑mediated feeding: comparison of wild-type, Y1 receptor-deficient, and Y5 receptor-deficient mice. Endocrinology 141, 1011–1016 (2000). 162. Cabrele, C. Beck-Sickinger, A. G. Molecular characterization of the ligand-receptor interaction of the neuropeptide Y family. J. Pept. Sci. 6, 97–122 (2000). 163. Batterham, R. L. et al. Gut hormone PYY(3–36) physiologically inhibits food intake. Nature 418, 650–654 (2002). 164. Abbott, C. R. et al. Blockade of the neuropeptide Y Y2 receptor with the specific antagonist BIIE0246 attenuates the effect of endogenous and exogenous peptide YY(3–36) on food intake. Brain Res. 1043, 139–144 (2005). 165. Koda, S. et al. The role of the vagal nerve in peripheral PYY3‑36‑induced feeding reduction in rats. Endocrinology 146, 2369–2375 (2005). 166. Burdyga, G. et al. Cholecystokinin regulates expression of Y2 receptors in vagal afferent neurons serving the stomach. J. Neurosci. 28, 11583–11592 (2008). www.nature.com/nrgastro © 2012 Macmillan Publishers Limited. All rights reserved
  • 11. REVIEWS 167. Batterham, R. L. et al. Inhibition of food intake in obese subjects by peptide YY3–36. N. Engl. J. Med. 349, 941–948 (2003). 168. Wynne, K. Bloom, S. R. The role of oxyntomodulin and peptide tyrosine-tyrosine (PYY) in appetite control. Nat. Clin. Pract. Endocrinol. Metab. 2, 612–620 (2006). 169. Boggiano, M. M. et al. PYY3–36 as an antiobesity drug target. Obes. Rev. 6, 307–322 (2005). 170. Ma, L. et al. Variations in peptide YY and Y2 receptor genes are associated with severe obesity in Pima Indian men. Diabetes 54, 1598–1602 (2005). 171. Lavebratt, C., Alpman, A., Persson, B., Arner, P. Hoffstedt, J. Common neuropeptide Y2 receptor gene variant is protective against obesity among Swedish men. Int. J. Obes. (Lond.) 30, 453–459 (2006). 172. Ahituv, N. et al. A PYY Q62P variant linked to human obesity. Hum. Mol. Genet. 15, 387–391 (2006). 173. Neary, N. M. et al. Peptide YY3–36 and glucagon-like peptide‑17‑36 inhibit food intake additively. Endocrinology 146, 5120–5127 (2005). 174. Field, B. C. et al. PYY3–36 and oxyntomodulin can be additive in their effect on food intake in overweight and obese humans. Diabetes 59, 1635–1639 (2010). 175. Reidelberger, R. D., Haver, A. C., Apenteng, B. A., Anders, K. L. Steenson, S. M. Effects of exendin‑4 alone and with peptide YY(3–36) on food intake and body weight in diet-induced obese rats. Obesity (Silver Spring) 19, 121–127 (2011). 176. Wettergren, A., Maina, P Boesby, S. Holst, J. J. ., Glucagon-like peptide‑1 7–36 amide and peptide YY have additive inhibitory effect on gastric acid secretion in man. Scand. J. Gastroenterol. 32, 552–555 (1997). 177. Baldassano, S., Bellanca, A., Serio, R. Mule, F. Food intake in lean and obese mice after peripheral administration of glucagon-like peptide‑2. J. Endocrinol. 213, 277–284 (2012). 178. Lund, A., Vilsboll, T., Bagger, J. I., Holst, J. J. Knop, F. K. The separate and combined impact of the intestinal hormones, GIP GLP‑1, and GLP‑2, , on glucagon secretion in type 2 diabetes. Am. J. Physiol. Endocrinol. Metab. 300, E1038–E1046 (2011). 179. Dixon, J. B., Straznicky, N. E., Lambert, E. A., Schlaich, M. P Lambert, G. W. Surgical . approaches to the treatment of obesity. Nat. Rev. Gastroenterol. Hepatol. 8, 429–437 (2011). 180. Schauer, P et al. Bariatric surgery versus . R. intensive medical therapy in obese patients with diabetes. N. Engl. J. Med. 366, 1567–1576 (2012). 181. Peterli, R. et al. Metabolic and hormonal changes after laparoscopic Roux‑en‑Y gastric bypass and sleeve gastrectomy: a randomized, prospective trial. Obes. Surg. 22, 740–748 (2012). 182. Bayham, B. E., Greenway, F. L., Bellanger, D. E. O’Neil, C. E. Early resolution of type 2 diabetes seen after Roux‑en‑Y gastric bypass and vertical sleeve gastrectomy. Diabetes Technol. Ther. 14, 30–34 (2012). 183. Cummings, D. E. et al. Plasma ghrelin levels after diet-induced weight loss or gastric bypass surgery. N. Engl. J. Med. 346, 1623–1630 (2002). NATURE REVIEWS | GASTROENTEROLOGY HEPATOLOGY © 2012 Macmillan Publishers Limited. All rights reserved 184. Korner, J. et al. Prospective study of gut hormone and metabolic changes after adjustable gastric banding and Roux‑en‑Y gastric bypass. Int. J. Obes. (Lond.) 33, 786–795 (2009). 185. Beckman, L. M., Beckman, T. R. Earthman, C. P Changes in gastrointestinal . hormones and leptin after Roux‑en‑Y gastric bypass procedure: a review. J. Am. Diet. Assoc. 110, 571–584 (2010). 186. Falken, Y., Hellstrom, P . M., Holst, J. J. Naslund, E. Changes in glucose homeostasis after Roux‑en‑Y gastric bypass surgery for obesity at day three, two months, and one year after surgery: role of gut peptides. J. Clin. Endocrinol. Metab. 96, 2227–2235 (2011). 187. Chandarana, K. et al. Diet and gastrointestinal bypass-induced weight loss: the roles of ghrelin and peptide YY. Diabetes 60, 810–818 (2011). 188. Chambers, A. P et al. Weight-independent . changes in blood glucose homeostasis after gastric bypass or vertical sleeve gastrectomy in rats. Gastroenterology 141, 950–958 (2011). Acknowledgements C. X. Dong is supported by a graduate studentship from the Banting and Best Diabetes Centre, University of Toronto, Canada. P Brubaker is supported by the . L. Canada Research Chairs program. Studies on GLP‑1 and GLP‑2 in the Brubaker laboratory are supported by operating grants from the Canadian Diabetes Association (#2374) and the Canadian Institutes of Health Research (#MOP‑9940), respectively. Author contributions Both authors contributed equally to all aspects of the manuscript. VOLUME 9  |  DECEMBER 2012  |  715