You are on page 1of 11

Neuropharmacology 63 (2012) 46e56

Contents lists available at SciVerse ScienceDirect

Neuropharmacology
journal homepage: www.elsevier.com/locate/neuropharm

Invited review

The role of the gut/brain axis in modulating food intake


Amir H. Sam, Rachel C. Troke, Tricia M. Tan, Gavin A. Bewick*
Section of Investigative Medicine, Imperial College London, London, W12 0NN, UK

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

Article history: Peptide hormones released from the gastrointestinal tract communicate information about the current
Received 4 August 2011 state of energy balance to the brain. These hormones regulate appetite and energy expenditure via the
Received in revised form vagus nerve or by acting on key brain regions implicated in energy homeostasis such as the hypothal-
28 September 2011
amus and brainstem. This review gives an overview of the main gut hormones implicated in the regu-
Accepted 13 October 2011
lation of food intake. Research in this area has provided novel targets for the pharmacological treatment
of obesity.
Keywords:
This article is part of a Special Issue entitled ‘Central Control Food Intake’
Peptide YY (PYY)
Glucagon like peptide-1 (GLP-1)
Crown Copyright Ó 2011 Published by Elsevier Ltd. All rights reserved.
Oxyntomodulin
Cholecystokinin
Ghrelin
Obesity

1. Introduction an increased risk of both all cause, and cause-specific mortality


(Prospective_Studies_Collaboration, 2009; Ringback Weitoft et al.,
Hormones released from the gut and adipose tissue play an 2008; Zheng et al., 2011). Peripheral signals controlling satiety
important role in the regulation of food intake and energy expen- may present potential targets for developing novel anti-obesity
diture. These hormones may exert their effects both via the vagus therapies. The focus of this review is to provide a synopsis of the
nerve and by acting directly on areas of the brain implicated in the gut-brain cross talk involved in the regulation of food intake.
control of energy homeostasis. Peripheral signals from the gut and
adipose tissue constitute feedback mechanisms allowing mainte-
nance of a steady body weight, despite daily variations in energy 2. Neuroendocrine control of appetite (Fig. 1)
expenditure and nutrient intake.
The role of peripheral hormones and the gut/brain axis in the The hypothalamus and the brainstem are the main central
regulation of appetite has become an area of interest in recent nervous system regions responsible for the regulation of energy
years, owing to the growing global obesity crisis. Obesity is homeostasis. These brain areas receive peripheral neural and
a major worldwide public health problem, with a significant hormonal signals that relay information about acute nutritional
burden of morbidity and mortality, as well as having substantial state and adiposity (Murphy and Bloom, 2006). Neural afferents and
economic consequences. It is an important risk factor for type 2 hormonal signals from the periphery are integrated with higher
diabetes, ischaemic heart disease, stroke and cancer, and carries brain centre signals (e.g. relaying reward drive and mood) to regu-
late appetite and control energy expenditure (Schwartz et al., 2000).
The arcuate nucleus (ARC) of the hypothalamus is believed to
play a crucial role in the regulation of food intake and energy
Abbreviations: AgRP, agouti related peptide; ARC, arcuate nucleus; a-MSH, homeostasis. The ARC contains two populations of neurons with
alpha-melanocyte stimulating hormone; AP, area postrema; CART, cocaine and
opposing effects on food intake (Chaudhri et al., 2008). Medially
amphetamine regulated transcript; CCK, cholecystokinin; DPP-IV, dipeptidyl
peptidase-4; GLP-1, glucagon like peptide-1; GLP-2, glucagon like peptide-2; NPY, located orexigenic neurons (i.e. those stimulating appetite) express
neuropeptide Y; POMC, pro-opiomelanocortin; NT, neurotensin; NTS, nucleus neuropeptide Y (NPY) and Agouti-related protein (AgRP) (Bewick
tractus solitarius; OEA, Oleoylethanolamine; OXM, oxyntomodulin; PP, pancreatic et al., 2005; Broberger et al., 1998; Hahn et al., 1998). Anorexi-
polypeptide; PYY, peptide tyrosine tyrosine; PVN, paraventricular nucleus; RYGB, genic neurons (i.e. those inhibiting appetite) in the lateral ARC
roux-en-Y gastric bypass; SFO, sub fornical organ; VIP, vasoactive intestinal
polypeptide.
express alpha-melanocyte stimulating hormone (alpha-MSH)
* Corresponding author. Tel.: þ44 2083833086; fax: þ44 2083838320. derived from pro-opiomelanocortin (POMC), and cocaine- and
E-mail address: g.bewick@imperial.ac.uk (G.A. Bewick). amphetamine-regulated transcript (CART) (Elias et al., 1998).

0028-3908/$ e see front matter Crown Copyright Ó 2011 Published by Elsevier Ltd. All rights reserved.
doi:10.1016/j.neuropharm.2011.10.008
A.H. Sam et al. / Neuropharmacology 63 (2012) 46e56 47

Fig. 1. Gut-brain axis: regulation of food intake. Nutrients created by the digestion of food are proposed to activate G protein coupled receptors on the luminal side of ene-
troendocrine cells e.g. the L-cell. This stimulates the release of gut hormones which may influence food intake at three sites: the vagus nerve, brainstem and hypothalamus. Within
the arcuate nucleus of the hypothalamus two neuronal populations are thought to be critical conduits through which peripheral signals are integrated to alter the drive to eat, the
orexigenic NPY/AgRP neurons and the anorexigenic POMC neurons. Further connections between hypothalamic nuclei and higher brain centres may exist which control the hedonic
aspects of food ingestion. ARC (arcuate nucleus), AgRP (agouti related peptide), GLP-1 (glucagon like peptide-1), NPY (neuropeptide Y), POMC (propiomelanocortin), PVN (para-
ventricular nucleus), PYY (peptide YY).

The ARC is adjacent to the median eminence, a ‘circum- cells undergo terminal differentiation as they migrate up from the
ventricular organ’ with fenestrated capillaries and hence an crypts to the epithelial surface (Tsubouchi and Leblond, 1979).
incomplete bloodebrain barrier (Peruzzo et al., 2000). Circulating Immunohistochemical studies have shown that enter-
hormones can therefore influence the activity of the arcuate nucleus oendocrine cells frequently express two or more products. PYY has
neurons directly, after passing across the median eminence. Gut been shown to co-localise with glucagon-like peptide-1 (GLP-1),
hormones, released from the gastrointestinal tract on a meal to meal neurotensin (NT), and cholecystokinin (CCK) in enteroendocrine
basis, signal short-term nutrient availability to the ARC. Other cells in the colon (Roth et al., 1992). Approximately 50% of CCK, and
circulating factors such as insulin and leptin (a circulating peptide NT cells co express PYY (Roth et al., 1992). Substance P/serotonin
released from adipose tissue) relay information regarding long-term cells are commonly found in the same crypts as PYY/GLP-1/CCK/NT
energy stores (Porte et al., 2002). cells and most of the substance P cells also contain serotonin.
Gastrointestinal vagal afferents are activated by mechanore- However, co-expression of substance P/serotonin in the same cells
ceptors and chemoreceptors, and converge in the nucleus of the as PYY/GLP-1/CCK/NT is only rarely observed (Roth et al., 1992).
tractus solitarius (NTS) of the brainstem. Neuronal projections from This suggests the existence of two separate branches of enter-
the NTS, in turn, carry signals to the hypothalamus. Circulating oendocrine L-cell differentiation, with one branch resulting in cells
factors such as gut hormones may influence the NTS neurons producing substance P and serotonin, and the other resulting in
through the adjacent circumventricular organ, the ‘area postrema’ GLP-1, PYY, NT, and CCK-producing cells (Roth et al., 1992).
(AP). Ablation of both the AP and another circumventricular organ, Chemosensing of gut luminal contents by the enteroendocrine
the sub fornical organ (SFO), has been shown to influence the action GI cells plays a critical role in the control of functions such as
of the gut hormone Peptide tyrosine tyrosine (PYY) (Baraboi et al., digestion, pancreatic secretion, food intake, and metabolic regula-
2010). Gut hormones also alter the activity of the ascending vagal tion. Evidence that endocrine cells can directly sense luminal
pathways from the gut to the brainstem (Jobst et al., 2004). contents has been demonstrated in PYY and GLP-1 expressing L
Hence, the hypothalamic ARC orexigenic and anorexigenic cells. It has been shown that both human and rodent intestinal L
neurons are influenced by numerous neural and hormonal inputs. cells express receptors previously identified in the oral epithelium
These ARC neurons in turn project to a number of extra-hypothalamic for detecting sweet (T1R2 and T1R3) and bitter sensation (T2R), as
and intra-hypothalamic regions, including in particular the hypo- well as amino acids (T1R1 and T1R3) (Jang et al., 2007; Rozengurt
thalamic paraventricular nucleus (PVN), where some of the impor- et al., 2006). The gustducin G protein associated with these taste
tant efferent pathways regulating energy expenditure arise. receptors has also been identified in L cells of the gut (Jang et al.,
2007; Rozengurt et al., 2006). The activation of these receptors
3. Enteroendocrine cells of the gastrointestinal tract leads to an increase in intracellular calcium levels and release of
gastrointestinal peptides from enteroendocrine cells (Rozengurt
There are at least 15 different types of enteroendocrine cells et al., 2006). The presence of the sweet taste receptor subunit
diffusely distributed throughout the gastrointestinal epithelium T1R3 and gustducin may also underlie a luminal glucose sensing
(Sjolund et al., 1983). These cells produce and release a variety of mechanism, since activation of these receptors mediates the post-
hormones and signalling molecules, which together constitute the prandial release of GLP-1 from intestinal L cells (Jang et al., 2007).
largest endocrine organ in the body (Cheng and Leblond, 1974; Fatty acids derived from digestion of dietary fats appear to be
Sjolund et al., 1983). The enteroendocrine cells are derived from sensed via separate mechanisms. The short-chain fatty acid
multipotent stem cells, located towards the base of the intestinal receptors GPR43 and GPR41 are expressed in PYY-containing
crypts. The different enteroendocrine cell subpopulations are enteroendocrine L cells (Karaki et al., 2006; Tazoe et al., 2009).
constantly re-established by the differentiation of enteroendocrine Short-chain fatty acids have been shown to increase PYY secretion
cells arising from these stem cells. The majority of enteroendocrine in rats when delivered directly into the colon (Cherbut et al., 1998;
48 A.H. Sam et al. / Neuropharmacology 63 (2012) 46e56

Fu-Cheng et al., 1995). GPR119 is another G protein coupled (Tschop et al., 2004), many groups have subsequently validated its
receptor found in intestinal endocrine cells as well as pancreatic anorectic effects (Chelikani et al., 2005; Vrang et al., 2006).
beta cells (Chu et al., 2008, 2007). Administration of oleoyletha- Demonstration of the anorectic effect of PYY in rodents is depen-
nolamide (OEA), an endogenous long chain fatty acid derivative, dent upon the full acclimatization of the animals to the handling
and other GPR119 agonists increases GLP-1 secretion, both in vitro and injection procedures, and is lost in the presence of even mild
and in vivo in rodents (Chu et al., 2008; Lauffer et al., 2009). stressors (Abbott et al., 2006; Halatchev et al., 2004). This is because
The enteroendocrine L cells therefore have the capacity to stress can reduce baseline food intake, making it difficult for
integrate complex nutrient sensing in the gut and to respond anorectic agents to further suppress appetite. Intravenous admin-
appropriately by releasing gut hormones. In addition to chemical istration of PYY inhibits food intake in humans and it is equally
stimulation, the endocrine cells of the gut also respond to neural effective in normal and obese subjects (Batterham et al., 2003a).
and physical stimulation of the cell by releasing peptide containing The anorectic effects of PYY3e36 appear to be mediated centrally
granules at the basolateral side of the cell. These peptides can have via the ARC, as peripheral administration of PYY3e36 has been
an endocrine role, a local paracrine role, and/or activate receptors shown to increase c-fos expression in this hypothalamic nucleus
present on nerves innervating the GI mucosa (Cummings and (Batterham et al., 2002). As discussed previously, the ARC contains
Overduin, 2007). two neuronal populations, orexigenic NPY/AgRP, and anorexigenic
POMC/CART neurons. There is conflicting data regarding the effects
4. Gut hormones regulating food intake of PYY3e36 on these neurons. Peripheral administration of PYY3e36
has been reported to decrease expression and release of NPY whilst
The gastrointestinal tract releases more than 20 different activating POMC neurons (Batterham et al., 2002). However, other
regulatory peptide hormones that influence a number of physio- investigators have reported that PYY3e36 inhibits POMC neurons
logical processes. Gut hormones act on tissues such as exocrine via postsynaptic Y2R (Ghamari-Langroudi et al., 2005). Moreover,
glands, smooth muscle and the peripheral nervous system (Murphy POMC knockout mice maintain their acute anorectic response to
and Bloom, 2006). The release of gut hormones such as PYY, GLP-1, peripherally administered PYY3e36, suggesting that POMC is not
and oxyntomodulin (OXM) is stimulated by distension of the critical to the inhibitory effects of PYY3e36 on feeding. PYY1e36 and
stomach and interactions between nutrients and the luminal wall PYY3e36 exert their effects through the neuropeptide Y family of
of the intestine (Adrian et al., 1985; Le Quellec et al., 1992). receptors (Larhammar, 1996). PYY1e36 binds with similar affinity to
Gut hormones are believed to contribute to the short-term feel- all of the Y receptors, however PYY3e36 is a selective high affinity
ings of satiety and hunger (Badman and Flier, 2005). These peptides agonist at the Y2 receptor subtype (Y2R) (Grandt et al., 1992). The
may reduce food intake by decreasing hypothalamic orexigenic sig- Y2R is thought to be the receptor responsible for mediating the
nalling, and increasing anorectic signalling (Batterham et al., 2002; reduction of food intake by PYY. It is an auto-inhibitory pre-
Jobst et al., 2004). Another effect of these peptides is to mediate synaptic receptor found on NPY neurons within the ARC (Broberger
inhibitory feedback mechanisms on intestinal transit, contributing to et al., 1997), and deficiency of the Y2R abolishes the anorectic effects
prolonged gastric distension, and increased satiety between meals of PYY (Batterham et al., 2002). Furthermore, the anorectic effects
(Lin et al., 1996; Wen et al., 1995). These combined CNS effects and of PYY3e36 are attenuated by Y2R antagonists (Abbott et al., 2005b).
‘intestinal brake’ mechanisms, mediated by gut peptides such as CCK, PYY3e36 is therefore thought to reduce food intake through acti-
PYY, GLP-1 and OXM, facilitate the control of food intake and post- vation of the Y2R.
prandial transit through the gastrointestinal tract. A vagal brainstem mediated pathway may also be involved in
the actions of circulating PYY3e36. PYY is present in myenteric
5. Peptide tyrosine tyrosine (PYY) nervous plexus neurons innervating the gastrointestinal tract and
the Y2R receptor has been identified on the vagus nerve (Koda et al.,
PYY, like NPY and Pancreatic Polypeptide (PP), belongs to the ‘PP- 2005). The effects of peripheral administration of PYY3e36 on both
fold’ family of proteins. These peptides are 36-amino acids in length food intake (Abbott et al., 2005a; Koda et al., 2005), and on the
and share a common tertiary structural motif known as the PP-fold. activation of ARC feeding neurons, are abolished following bilateral
C-terminal amidation of these proteins is a necessary requirement sub-diaphragmatic total truncal vagotomy or transectioning of the
for biological activity. PYY exists endogenously in two forms: PYY1e36 brainstemehypothalamic pathway in rodents (Abbott et al., 2005a),
and PYY3e36 (Grandt et al., 1994). The enzymatic cleavage of secreted supporting a role for the vagus in appetite regulation.
PYY1e36 at the amino terminal by the cell surface enzyme dipeptidyl PYY has been shown to have an effect on intestinal motility. Close
peptidase IV (DPP-IV) gives rise to PYY3e36 (Medeiros and Turner, intra-arterial administration of PYY in cats causes an inhibition of
1994), the predominant form of circulating PYY immunoreactivity. jejunal and colonic motility (Lundberg et al., 1982). PYY also delays
Low levels of PYY are detected in enteroendocrine cells in the gastric emptying, decreases pancreatic secretions, and increases
stomach, and levels increase distally along the small and large absorption of fluids and electrolytes from the ileum in humans
intestine, reaching their highest levels in cells in the colon and (Savage et al., 1987; Symersky et al., 2005). It is possible that some of
rectum (Adrian et al., 1985). Endogenous circulating concentrations the anorectic effects of PYY may be mediated by the gastric disten-
of PYY are lowest in the fasting state, and rise post-prandially as sion resulting from delayed gastric emptying.
PYY is released from enteroendocrine L cells lining the distal GI Interestingly, it has recently been shown that acute effects of
tract in proportion to caloric intake (Adrian et al., 1985) Plasma gastrointestinal bypass surgery on body weight are lost in PyyKO
levels of PYY rise within 30 min of a meal, and in humans, circu- mice (Chandarana et al., 2011), and that wild-type mice losing weight
lating levels plateau at 1e2 h post-prandially, remaining elevated after gastrointestinal bypass surgery exhibit increased colonic Pyy
for up to 6 h (Batterham et al., 2003a). Protein rich meals cause the expression and circulating fasting PYY levels (Chandarana et al.,
greatest increase in PYY levels compared to other macronutrients 2011). This suggests that PYY plays a key role in mediating the early
(Batterham et al., 2006; Pedersen-Bjergaard et al., 1996). weight loss that occurs following gastrointestinal bypass surgery.
Peripheral administration of PYY3e36 reduces food intake and The effects of PYY3e36 on satiety and central control of appetite
weight gain in rodents (Batterham et al., 2002; Challis et al., 2003; are clear. Most are mediated via anorectic neuronal populations in
Chelikani et al., 2005; Vrang et al., 2006), and although there was the ARC, but vagal/brainstem-mediated pathways and peripheral
initial controversy regarding the effects of PYY3e36 on food intake effects of PYY on gastric emptying and intestinal motility may also
A.H. Sam et al. / Neuropharmacology 63 (2012) 46e56 49

play a part. High plasma concentrations of PYY result in nausea, but mellitus. Exenatide has also been shown to reduce body weight in
the importance of PYY3e36 at physiological levels in the regulation treated diabetics in phase III clinical trials (Buse et al., 2004;
of energy intake make it a prime focus for new obesity therapies, DeFronzo et al., 2005; Kendall et al., 2005). The weight loss asso-
targeted either at PYY itself, or against the Y2 receptor. ciated with exenatide is considered a significant advantage as many
anti-diabetic treatments are commonly associated with weight
6. Glucagon-like peptide-1 (GLP-1) gain. Nausea is a relatively common side effect of the treatment.
However, it does not seem to be intrinsically linked to the effects on
GLP-1 is a 30 amino acid peptide resulting from cleavage of the appetite (Murphy and Bloom, 2006). Whilst GLP-1 has been
preproglucagon precursor molecule (Dhanvantari et al., 1996). The developed as a treatment for diabetes due to its incretin properties,
two bioactive forms of GLP-1, GLP-17e37 and GLP-17e36 amide, are the observed effects of GLP-1 on satiety and weight loss are
released into the circulation from L cells of the gastrointestinal tract a valuable secondary effect. Indeed recent data suggests liraglutide
in response to an oral glucose load (Herrmann et al., 1995). Physi- maybe useful for the treatment of obesity, causing sustained weight
ologically, GLP-1 is an important incretin, stimulating glucose- loss over 2 years but with a 50% rate of nausea and vomiting in the
dependent insulin release (MacDonald et al., 2002). In addition to 3.0 mg/day group in the first year (Astrup et al., 2011).
its incretin effect, GLP-1 also inhibits the secretion of glucagon,
thereby inhibiting endogenous glucose production (Willms et al., 7. Oxyntomodulin (OXM)
1996). The net effect is to reduce blood glucose following a meal.
GLP-1 also delays gastric emptying (Schirra et al., 2006), and OXM, like GLP-1, is also a product of the preproglucagon
increases satiety (Punjabi et al., 2011). precursor molecule. It is a 37 amino acid peptide released post-
Like PYY, GLP-1 has been shown to act centrally in hypothalamic prandially from L cells in proportion to caloric intake (Le Quellec
nuclei known to be implicated in the control of appetite including et al., 1992). OXM delays gastric emptying and reduces gastric
the ARC, PVN and supraoptic nucleus (Shughrue et al., 1996). Both acid secretion (Schjoldager et al., 1989), and has been shown
acute peripheral and central administration of GLP-1 reduce food acutely to decrease food intake and in the longer term to decrease
intake in rats (Tang-Christensen et al., 2001; Turton et al., 1996) and weight gain in rodents (Dakin et al., 2001, 2004). In addition,
chronic administration of GLP-1 reduces weight gain (Meeran et al., chronic administration of OXM causes rats to lose more weight than
1999). The intravenous administration of GLP-1 to normal and pair-fed controls, suggesting an increase in energy expenditure
obese humans decreases food intake in a dose dependent manner (Dakin et al., 2002). OXM also reduces food intake in normal weight
(Verdich et al., 2001) as well as reducing gastric emptying (Naslund human volunteers when administered intravenously or subcuta-
et al., 1999; Nauck et al., 1997). These effects are thought to be neously (Cohen et al., 2003). Given preprandially to obese subjects
mediated through vagal and brainstem pathways since peripheral it reduces both food intake and body weight (Wynne et al., 2005).
administration of GLP-1 activates neurons within the brainstem in As in rats, there is evidence that OXM may also increase energy
rats (Imeryuz et al., 1997). Furthermore, this increase in neuronal expenditure in humans (Wynne et al., 2006).
activity, and the anorectic effects of GLP-1, are abolished following Although OXM has some agonist activity at the glucagon receptor,
vagotomy in rodents (Abbott et al., 2005a; Imeryuz et al., 1997), there is evidence that its anorectic effect is predominantly mediated
reinforcing the argument for vagal involvement. More recently, via the GLP-1 receptor (Baggio et al., 2004; Dakin et al., 2001). The
techniques such as functional magnetic resonance imaging (fMRI) anorectic effects of OXM are abolished in GLP-1 receptor knockout
have confirmed a change in signal intensity within neuronal pop- mice (Baggio et al., 2004) and in the presence of the GLP-1 receptor
ulations in the VMH and the PVN following peripheral adminis- antagonist exendin 9e39 (Dakin et al., 2004), suggesting that OXM
tration of GLP-1 (Parkinson et al., 2009). This adds further weight to acts via the GLP-1 receptor. OXM has a roughly 50-fold lower affinity
the evidence that appetite and feeding are regulated by GLP-1 via for the GLP-1 receptor than GLP-1 itself, but despite this, it reduces
these hypothalamic and brainstem areas. food intake with similar potency (Dakin et al., 2001). Furthermore,
GLP-1 is rapidly degraded in the circulation by the enzyme DPP- although the administration of exendin 9e39 directly into the ARC
IV, making native GLP-1 unsuitable for therapeutic use. Longer blocks the anorectic effects of OXM, it does not block those of GLP-1
acting GLP-1 mimetics have been developed as new treatments for (Dakin et al., 2004). Therefore, it is possible that OXM may act via an
type 2 diabetes (Joy et al., 2005). Exendin-4 is a naturally occurring as yet unidentified receptor or a specific subpopulation of the GLP-1
GLP-1 mimetic isolated from the venom of Heloderma suspectum, receptor. Studies using manganese-enhanced magnetic resonance
a lizard native to several southwestern American states (Eng et al., imaging MRI (MEMRI) have shown that intraperitoneal administra-
1992). A truncated form of this peptide, exendin 9e39, acts as tion of OXM causes a reduced rate of signal enhancement, reflecting
a competitive antagonist at the GLP-1 receptor. Acute intra- a reduction in neuronal activity, in the ARC, PVN, and supraoptic
cerebroventricular administration of exendin 9e39 increases food nucleus. This pattern of activation was distinct from that of GLP-1
intake, and chronic administration increases body weight in rats under the same conditions (Chaudhri et al., 2006), implying that
(Meeran et al., 1999; Turton et al., 1996). Endogenous peripheral these two hormones act via different hypothalamic pathways.
GLP-1 may physiologically reduce appetite and food intake after
a meal. However, GLP-1 receptor knockout mice do not have altered 8. Cholecystokinin (CCK)
food intake or body weight (Drucker, 2006). This may be because
developmental changes compensate for the lack of GLP-1 signal- CCK is released post-prandially from the small intestine
ling, or may reflect that GLP-1 has a more important physiological (Murphy and Bloom, 2006), and has also been shown to co-localise
role in controlling blood glucose than in regulating food intake. with PYY in L cells (Roth et al., 1992). Two types of CCK receptor
The discovery of exendin-4 has led to the development of have been identified in the CNS and peripheral tissues. CCK1
a synthetic version, exenatide. Exenatide has a much longer in vivo receptors are present in peripheral tissues such as the pancreas,
half-life than GLP-1 and is the first incretin mimetic approved for gallbladder, and on vagal afferent nerve fibres innervating the gut
the treatment of type 2 diabetes (Drucker and Nauck, 2006). Exe- (Moran and Kinzig, 2004). Furthermore, CCK1 receptors have been
natide is effective at stimulating insulin release, suppressing identified in areas within the CNS involved in the regulation of food
glucagon and lowering blood glucose. Clinical trials have demon- intake such as the NTS, AP, and dorsomedial hypothalamus (Moran
strated that exenatide is useful for the treatment of type 2 diabetes et al., 1986). The CCK2 receptor has a different distribution, and is
50 A.H. Sam et al. / Neuropharmacology 63 (2012) 46e56

found in the cortex, hypothalamus, vagal afferents and gastric Intravenous infusion of PP at doses that achieve normal post-
mucosa (Moran et al., 1986), once again encompassing several areas prandial plasma concentrations reduces appetite in lean humans
known to be involved in appetite regulation. and inhibition of food intake persists for 24 h after infusion
CCK is released post-prandially in response to saturated fat, long (Batterham et al., 2003b). PP has also been shown to reduce food
chain fatty acids, amino acids and small peptides that would nor- intake at lower infusion rates (Jesudason et al., 2007). Furthermore,
mally result from protein digestion (Liddle et al., 1985; Rehfeld pancreatic polypeptide has been shown to reduce food intake in
et al., 2003). CCK release and signalling via the CCK1 receptors in patients with obesity secondary to PradereWilli syndrome
response to these long chain fatty acids mediates stimulation of (Berntson et al., 1993).
PYY release and inhibition of ghrelin (an orexigenic gut hormone) PP has also been implicated in energy homeostasis, with exog-
in human subjects (Degen et al., 2007). Both of these hormones enous administration of PP causing an increase in oxygen
would then further act to suppress appetite and inhibit food intake. consumption (Asakawa et al., 2003), thus implying that part of the
Post-prandial secretion of CCK also triggers the release of pancre- effect of PP on body weight may be due to increased energy
atic enzymes, and of bile salts from the gallbladder, into the expenditure. It has also been shown to increase spontaneous
duodenum, promoting protein and fat digestion (Liddle et al., 1985; locomotor activity in mice (Liu et al., 2008).
Rehfeld et al., 2003). In addition, CCK regulates other gastrointes- PP binds to all the members of the Y receptor family, but has the
tinal functions including gastric emptying (Fried et al., 1991). highest affinity for the Y4 receptor subtype (Michel et al., 1998). The
The effects of CCK on appetite are well documented. Peripheral effects of PP are likely to be mediated by both the hypothalamus
administration of CCK in rodents results in a dose dependant and brainstem (Asakawa et al., 2003). The Y4 receptor mRNA has
reduction in food intake, decreasing both meal size and duration been shown to be present in the appetite-regulating areas of the
(Antin et al., 1975). CCK administration is also associated with an brainstem notably the AP and in the ARC (Larsen and Kristensen,
increase in post-prandial satiety behaviours such as increased 1997; Parker and Herzog, 1999). Functional imaging with MEMRI
grooming and decreased locomotor activity (Antin et al., 1975). The following peripheral administration of PP has also suggested
Otsuka Long-Evans Tokushima Fatty rat was bred to produce a central mode of action, demonstrating a significant reduction in
a polygenic model of diabetes. These rats lack the CCK1 receptor, signal intensity in the ARC, VMH and PVN of fasted mice. This
are hyperphagic and obese, suggesting that CCK has a physiological reduction in signal intensity implies a reduction in activity of
role in the regulation of food intake (Bi and Moran, 2002). However, neurons in these areas central to the control of appetite. The
CCK1 receptor knockout mice do not differ significantly in body decrease in signal intensity correlated with a reduction in food
weight from wild types (Kopin et al., 1999). This discrepancy has intake (Hankir et al., 2011). It has also been suggested that inhibi-
been suggested to be due to a number of reasons. The most likely tion of food intake by PP may be secondary to an effect on gastric
explanation is the potential for the OLETF phenotype to be driven emptying, and therefore increased gastric distension. However,
by other, as yet unidentified, genetic mutations in addition to the while some investigators have reported an inhibition of gastric
loss of CCK1. These data suggest that CCK may play a role in acute emptying, others have failed to show this (Adrian et al., 1981;
rather than long-term energy homeostasis. It has been reported Batterham et al., 2003b; Schmidt et al., 2005). Furthermore, the
that endogenous circulating concentrations of CCK cannot activate anorectic effects of PP in humans appear to be independent of
vagal circuits, suggesting that the actions of CCK on food intake changes in gastric motility (Batterham et al., 2003b). These data
might be paracrine or neurocrine rather than endocrine (Moran, have lead to a concerted effort to develop long acting PP analogues,
2000). In humans, intravenous administration of physiological which have completed Phase I trials (Tan et al., 2011).
doses of CCK reduces food intake and increases the perception of
fullness (Lieverse et al., 1995). Unfortunately, the therapeutic 10. Glucagon
potential of CCK as a treatment for obesity is limited by nausea and
tachyphylaxis of the anorectic effects associated with chronic Glucagon is a 29 amino acid peptide secreted from the a-cells of
administration (Covasa et al., 2001). the pancreatic islets of Langerhans. It is a further product of pre-
proglucagon cleavage alongside OXM and GLP-1. Glucagon is
9. Pancreatic polypeptide (PP) released into the portal vein in fasted states and also in response to
exercise, and acts on the liver to promote hepatic glycogenolysis
PP is an amidated 36-amino acid peptide and belongs the ‘PP- and gluconeogenesis and maintain glycaemic balance (Stevenson
fold’ family of peptides. It is released post-prandially under vagal et al., 1987; Striffler et al., 1981; Studer et al., 1984; Wasserman
control by pancreatic islet PP cells (Adrian et al., 1976; Larsson et al., et al., 1989).
1975; Schwartz et al., 1978). PP is comparable to other anorectic Glucagon mediates its effects via the glucagon receptor, a 7-
intestinal peptides such as PYY, being secreted in proportion to transmembrane G protein coupled receptor which has a wide
caloric intake. Circulating levels rise after meals and remain tissue distribution. It is expressed in the gut, adrenal glands, brain,
elevated for up to 6 h post-prandially (Adrian et al., 1976). heart, pancreas, spleen and in adipocytes, but is predominantly
The functions of PP in regulating gallbladder motility and found in the liver and kidney (Svoboda et al., 1994).
pancreatic secretions have previously been well characterized As a potential treatment for obesity, glucagon has been shown to
(Adrian et al., 1979, 1976; Greenberg et al., 1979). PP has been increase energy expenditure in rats, and also in humans during
shown to augment basal insulin and glucagon release in rats and insulin deficiency (Davidson et al., 1960; Nair, 1987). It also signif-
this is thought to be via a direct paracrine effect on the pancreatic icantly reduces food intake, with a subjective reduction of appetite
beta and alpha cells respectively (Szecowka et al., 1983). Intraper- in man (Schulman et al., 1957). Infusion of glucagon into the portal
itoneal injection of PP acutely reduces food intake in fasted mice vein but not the inferior vena cava causes a reduction in meal size in
(Asakawa et al., 2003), an effect that remains apparent for 24 h after rats (Geary et al., 1993; Le Sauter et al., 1991).
injection. Furthermore, chronic administration of PP over 6 days in Glucagon presents an interesting prospect in the treatment of
ob/ob mice significantly reduces body weight gain and improves obesity due to its effect on increasing energy expenditure, and
glucose profile (Asakawa et al., 2003). Pancreatic polypeptide-over- increasing satiety. It has been demonstrated that the potential
expressing mice with supraphysiological PP concentrations have unfavourable effect on glucose tolerance due to glucagon’s actions
reduced food intake and gain less weight (Ueno et al., 1999). on hepatic glycogenolysis and gluconeogenesis is effectively
A.H. Sam et al. / Neuropharmacology 63 (2012) 46e56 51

counteracted by dual agonism at the glucagon and GLP-1 receptors peptides in the regulation of food intake and energy homeostasis
(Day et al., 2009; Pocai et al., 2009). The data from these studies remains unclear.
demonstrated highly effective weight loss in diet-induced obese NT was first isolated from hypothalamic tissue, but is widely
mice whilst avoiding the hyperglycaemia that might be expected distributed throughout the central nervous system. However, the
from agonism at the glucagon receptor. majority of NT is found within enteroendocrine cells of the GI tract
(Carraway and Leeman, 1976). NT regulates a number of digestive
11. Ghrelin processes, including gastrointestinal motility, and pancreatic and
biliary secretion (Kitabgi, 2006). It also has trophic effects on the
Ghrelin is a 28-amino acid acylated peptide secreted from the pancreas and small intestine (Feurle et al., 1987; Wood et al.,
stomach. It was originally identified as an endogenous ligand for 1988a,b). Plasma levels of NT increase after a meal, with intra-
the ‘growth hormone secretagogue’ receptor (GHS-R) and is luminal fat being the most potent stimulus (Rosell and Rokaeus,
a growth-hormone-releasing peptide (Kojima et al., 1999). 1979). Peripheral administration of neurotensin decreases food
Ghrelin is the only orexigenic gut hormone (Bewick et al., 2009), intake and grooming behaviour in rats only at large doses (Sandoval
causing an increase in food intake and weight gain in rodents and Kulkosky, 1992). Therefore at physiological levels, neurotensin
following both peripheral and central administration (Lawrence is unlikely to play a major role in appetite regulation. Although
et al., 2002; Tschop et al., 2000; Wren et al., 2000). Intravenous neurotensin acutely reduces food intake when administered cen-
administration of ghrelin has also been shown to stimulate gastric trally in rats or peripherally in mice, chronic administration to mice
acid secretion and motility in rats (Masuda et al., 2000). In normal has no significant effect on food intake or body weight (Cooke et al.,
subjects, ghrelin levels are highest in the fasted state (Toshinai 2009). The lack of chronic effects on body weight suggests that NT
et al., 2001), and levels are chronically higher in people with is unlikely to be useful as a treatment for obesity.
weight loss due to anorexia nervosa or dietary reduction (Ariyasu Intracerebroventricular injection of glucagon-like peptide-2
et al., 2001; Cummings et al., 2002; Shiiya et al., 2002). In (GLP-2) into rats inhibits food intake. In contrast, GLP-2 adminis-
contrast to other gut hormones, plasma ghrelin levels decrease tered peripherally does not inhibit food intake in rodents or
after meals (Ariyasu et al., 2001; Tschop et al., 2001) and are low in humans (Schmidt et al., 2003; Scott et al., 1998). GLP-2 appears to
obese subjects (Shiiya et al., 2002). Ghrelin concentrations are also play a more important physiological role as an intestinal growth
reduced after gastric bypass surgery, and this may contribute to factor (Scott et al., 1998).
weight loss in such patients (Cummings et al., 2002). Amylin is a peptide co-secreted with insulin by pancreatic beta
Ghrelin and its receptor are both expressed in vagal afferents in cells. Injection of amylin or amylin agonist has been shown to
mice (Page et al., 2007), and blockade of the gastric vagal afferent reduce food intake in a number of species, including humans
has been shown to abolish ghrelin-induced feeding, growth (Chance et al., 1993; Lutz et al., 1994; Ratner et al., 2005; Rushing
hormone secretion, and activation of NPY-producing and growth- et al., 2000; Whitehouse et al., 2002). The amylin receptor
hormone-releasing hormone producing neurons in rats (Date et al., agonist pramlintide has been shown to cause weight loss in dia-
2002). Central administration of ghrelin has been shown to betic humans (Ratner et al., 2005; Whitehouse et al., 2002).
increase pancreatic exocrine secretion via the vagus in conscious Vasoactive intestinal polypeptide (VIP) has been shown to
rats (Sato et al., 2003). reduce appetite, in addition to its well-characterized effects on the
Ghrelin receptors are found in the ARC of the hypothalamus, and cardiovascular system and gastrointestinal motility and secretion.
c-fos expression is increased in the ARC after peripheral adminis- Intracerebroventricular administration of VIP has been shown to
tration of ghrelin (Hewson and Dickson, 2000). In rodents who had cause a potent short-lived decrease in food intake and an increase
undergone ablation of the ARC, administration of ghrelin did not in activity and energy expenditure in rats. Treatment of hypotha-
stimulate food intake (Tamura et al., 2002). When given centrally, lamic explants with VIP stimulated the release of the anorexigenic
ghrelin also stimulates c-fos expression in other nuclei known to be peptide a-MSH (Ghourab et al., 2011). These studies suggest
involved in appetite control including the PVN, dorsomedial a possible endogenous role for VIP in the hypothalamic control of
nucleus, and lateral hypothalamus as well as in the AP and NTS in energy homeostasis.
the brainstem (Lawrence et al., 2002). Hypothalamic levels of
orexigenic peptides NPY and AgRP mRNA were also increased after 13. Gut hormones and the treatment of obesity
chronic central administration of ghrelin (Kamegai et al., 2001).
Diet-induced obesity is associated with a blunting of ghrelin’s Lifestyle and dietary modification alone are inadequate for the
orexigenic effect. There has therefore been recent interest in the successful treatment of the majority of obese individuals. However,
interaction between the ghrelin system and macronutrients. High despite an increasingly high demand for intervention, the field of
fat feeding has been shown to render NPY/AgRP neurones relatively obesity therapeutics has limited options to offer these patients. The
ghrelin resistant (Briggs et al., 2010), and diets high in fat have been history of obesity pharmacotherapy is littered with examples of
shown to directly inhibit the hyperphagic effect of ghrelin drugs withdrawn from the market due to adverse effects out-
(Gardiner et al., 2010; Perez-Tilve et al., 2011). These data have weighing the beneficial effects of weight loss. Recent examples
significant implications for developing anti-obesity treatments include Sibutramine, a norepinephrine and serotonin reuptake
targeting the ghrelin system and suggest success of these inhibitor, and Rimonabant, which is a cannabinoid-1 receptor
approaches could depend on the fat content of the diet the patient blocker. Sibutramine was withdrawn after it was found to increase
consumes. More recently, ghrelin has been shown to engage heart rate and blood pressure in some subjects, and was associated
neurons in the ventral tegmental area of the brain and may provide with an increased risk of stroke and non-fatal myocardial infarction
a link between the gut and neuronal control of stress-induced in patients with pre-existing cardiovascular conditions (James et al.,
eating of ‘comfort foods’ (Chuang et al., 2011). 2010), whilst Rimonabant was withdrawn amidst concerns
regarding adverse psychiatric events (Christensen et al., 2007). The
12. Other gut peptides only currently licensed product in the UK is Orlistat, a pancreatic
lipase inhibitor which prevents fat absorption and confers a modest
A number of other gut-derived peptides have been shown to weight loss of 2.9 kg more than placebo over the course of a year
reduce food intake. However, the physiological role of these (Rucker et al., 2007).
52 A.H. Sam et al. / Neuropharmacology 63 (2012) 46e56

The only obesity treatment that has been shown to confer long- insulinotropic action (Chu et al., 2007). It is less clear if these
term, sustained weight loss and a decrease in overall mortality is compounds will be effective as anti-obesity agents, but some
bariatric surgical intervention (Adams et al., 2007; Sjostrom et al., agonists have been shown to significantly reduce food intake, for
2007). Several surgical procedures are available to achieve weight example PSN632408 (Overton et al., 2006). GPR119 is currently the
loss. Gastric banding restricts the amount of food that can be only target for which synthetic modulators stimulate both incretin
comfortably ingested and increases the satiating effect of food and insulin release. This highly beneficial profile has generated
(Dixon et al., 2005). A more efficient reduction in appetite and great industry interest with at least 9 companies actively working
weight loss is seen with surgical procedures that involve gastro- in this area. Initial clinical trials have been successful with respect
intestinal bypass, such as Roux-en-Y Gastric bypass (RYGB) (Adams to the anti-diabetic indication (Jones et al., 2009; Shah and
et al., 2007; Kenler et al., 1990; Sjostrom et al., 2007). Weight loss is Kowalski, 2010).
normally associated with reduced plasma levels of the adipocyte-
derived anorectic hormone leptin, causing increased hunger 14. Conclusion
(Chan et al., 2003). However, following RYGB, despite significant
reductions in body weight and leptin levels, appetite is markedly Obesity has emerged as a major global healthcare challenge. The
reduced (Kenler et al., 1990). significant mortality and morbidity associated with obesity has
A significant difference in the effects of different forms of bariatric inspired a vast amount of research directed towards developing safe
surgery on type 2 diabetes has been shown (Gan et al., 2007). RYGB and efficacious weight loss agents. The beneficial effects of centrally
ameliorates coexistent type 2 diabetes mellitus before substantial acting weight loss agents have been negated by their potentially
weight loss has occurred and more rapidly than gastric banding. The hazardous effects on mood, reward, dependence and autonomic
differences between gastric banding and RYGB may be due to alter- tone. Gut hormones, as outlined in this article, play an important
ations in the anorectic and incretin gut hormone profile that is seen role in the homeostatic control of food intake and offer an alterna-
following RYGB, but not following gastric banding (Kellum et al., tive to centrally acting drugs. In particular, recent data suggests that
1990; le Roux et al., 2006). Experimental evidence suggests that mimicking the gut hormone profile observed following gastric
these anorectic gut hormones may mediate the effects of RYGB on bypass may offer viable new treatments for obesity. This can be
appetite and body weight (le Roux et al., 2006; le Roux et al., 2007). accomplished either through the use of long acting peptide based
Post-prandial PYY and GLP-1 levels begin to rise as early as 2 days therapies, alone or in combination, or with gut hormone secreta-
following gastric bypass in humans (le Roux et al., 2007), and gogues. We believe the most promising avenues for future exploi-
secretory products of enteroendocrine L cells, including PYY and GLP- tation lie in the development of longer acting GLP-1 agonists,
1 remain elevated two years after bypass surgery (le Roux et al., 2010). combination therapies including GLP-1/glucagon co-agonists and
Inhibiting gut hormone release with somatostatin analogue octreo- gut hormone secretagogues targeting L-cell nutrient receptors
tide increases the food intake after gastric bypass surgery but not (eg. GPR119 agonists) either as small molecules or as functional
following gastric banding (le Roux et al., 2007), further suggesting foods. Thus, our emerging understanding of the physiological
that these hormones play a critical role. Determining the mechanisms systems in the gut which regulate food intake has identified novel
behind this sustained reduction in appetite may identify pathways targets for the treatment of obesity and its related co-morbidities.
that can be targeted by anti-obesity agents. To this end there has been We believe that in time these approaches will develop clinically
recent concerted effort to mimic the rise in gut hormones following useful compounds which will offer a real answer to the ever
gut bypass by either the development of peptide based analogues or growing burden of obesity.
by the design of small molecule drugs which target nutrient sensing
receptors on the enteroendocrine L-cell.
Acknowledgements
Long acting versions of PYY and OXM are being actively pursued
by the pharmaceutical industry, such as Pfizer’s OAP-189, we await
The Section is funded by grants from the MRC, BBSRC, NIHR, an
the dissemination of data from ongoing trials. Given that gut
Integrative Mammalian Biology (IMB) Capacity Building Award, an
hormones are co-released one logical approach would be the
FP7-HEALTH-2009-241592 EuroCHIP grant and is supported by the
development of combination therapies. Indeed data suggests that
NIHR Imperial Biomedical Research Centre Funding Scheme. Amir
co-administration of gut hormones can have additive effects on
H. Sam is funded by a Wellcome Trust Research Training Fellow-
food intake inhibition, for example PYY þ GLP-1 (Neary et al., 2005)
ship. Tricia Tan is funded by Developmental Pathway Funding and
or PYY þ OXM (Field et al., 2010). Such combination approaches
Developmental Clinical Schemes from the Medical Research
may prove more effective than individual administration. Very
Council. Gavin A Bewick is funded by an EFSD Paul Langerhans
recently the development of chimeric agonists has emerged as
grant and by an NIHR career development fellowship.
a novel form of combination therapy (Day et al., 2009). GLP-1/
glucagon co-agonists combine the appetite suppressive effects of
GLP-1 and glucagon with the energy expenditure promoting effects References
of glucagon. Whilst at the same time GLP-1’s insulinotropic effects
Abbott, C.R., Monteiro, M., Small, C.J., Sajedi, A., Smith, K.L., Parkinson, J.R.,
inhibit the detrimental hyperglycaemic effects of glucagon. Mar- Ghatei, M.A., Bloom, S.R., 2005a. The inhibitory effects of peripheral adminis-
cadia Ltd., now a subsidiary of Roche, first reported the beneficial tration of peptide YY(3-36) and glucagon-like peptide-1 on food intake are
effects of this approach and their compound is now undergoing attenuated by ablation of the vagal-brainstem-hypothalamic pathway. Brain
Res. 1044, 127e131.
clinical trials. In addition, Zealand Pharma is also developing Abbott, C.R., Small, C.J., Kennedy, A.R., Neary, N.M., Sajedi, A., Ghatei, M.A.,
a similar compound, ZP-2929, in partnership with Boehringer Bloom, S.R., 2005b. Blockade of the neuropeptide Y Y2 receptor with the
Mannheim. Time will tell if the promising pre-clinical data trans- specific antagonist BIIE0246 attenuates the effect of endogenous and exogenous
peptide YY(3-36) on food intake. Brain Res. 1043, 139e144.
lates in to clinical benefit (Daugaard et al., 2010).
Abbott, C.R., Small, C.J., Sajedi, A., Smith, K.L., Parkinson, J.R., Broadhead, L.L.,
Considerable energy has also been directed toward the devel- Ghatei, M.A., Bloom, S.R., 2006. The importance of acclimatisation and habit-
opment of gut hormone secretagogues. The most well charac- uation to experimental conditions when investigating the anorectic effects of
terised class being agonists of GPR119. These compounds have been gastrointestinal hormones in the rat. Int. J. Obes. (Lond) 30, 288e292.
Adams, T.D., Gress, R.E., Smith, S.C., Halverson, R.C., Simper, S.C., Rosamond, W.D.,
shown to release both GLP-1 and PYY (Chu et al., 2008). Their anti- Lamonte, M.J., Stroup, A.M., Hunt, S.C., 2007. Long-term mortality after gastric
diabetic effects are well defined; stimulation of GLP-1 and a direct bypass surgery. N. Engl. J. Med. 357, 753e761.
A.H. Sam et al. / Neuropharmacology 63 (2012) 46e56 53

Adrian, T.E., Bloom, S.R., Bryant, M.G., Polak, J.M., Heitz, P.H., Barnes, A.J., 1976. Chandarana, K., Gelegen, C., Karra, E., Choudhury, A.I., Drew, M.E., Fauveau, V.,
Distribution and release of human pancreatic polypeptide. Gut 17, 940e944. Viollet, B., Andreelli, F., Withers, D.J., Batterham, R.L., 2011. Diet and gastroin-
Adrian, T.E., Besterman, H.S., Mallinson, C.N., Greenberg, G.R., Bloom, S.R., 1979. testinal bypass-induced weight loss: the roles of ghrelin and peptide YY. Dia-
Inhibition of secretin stimulated pancreatic secretion by pancreatic poly- betes 60, 810e818.
peptide. Gut 20, 37e40. Chaudhri, O.B., Parkinson, J.R., Kuo, Y.T., Druce, M.R., Herlihy, A.H., Bell, J.D.,
Adrian, T.E., Greenberg, G.R., Fitzpatrick, M.L., Bloom, S.R., 1981. Lack of effect of Dhillo, W.S., Stanley, S.A., Ghatei, M.A., Bloom, S.R., 2006. Differential hypo-
pancreatic polypeptide in the rate of gastric emptying and gut hormone release thalamic neuronal activation following peripheral injection of GLP-1 and oxy-
during breakfast. Digestion 21, 214e218. ntomodulin in mice detected by manganese-enhanced magnetic resonance
Adrian, T.E., Ferri, G.L., Bacarese-Hamilton, A.J., Fuessl, H.S., Polak, J.M., Bloom, S.R., imaging. Biochem. Biophys. Res. Commun. 350, 298e306.
1985. Human distribution and release of a putative new gut hormone, peptide Chaudhri, O.B., Salem, V., Murphy, K.G., Bloom, S.R., 2008. Gastrointestinal satiety
YY. Gastroenterology 89, 1070e1077. signals. Annu. Rev. Physiol. 70, 239e255.
Antin, J., Gibbs, J., Holt, J., Young, R.C., Smith, G.P., 1975. Cholecystokinin elicits the Chelikani, P.K., Haver, A.C., Reidelberger, R.D., 2005. Intravenous infusion of peptide
complete behavioral sequence of satiety in rats. J. Comp. Physiol. Psychol. 89, YY(3-36) potently inhibits food intake in rats. Endocrinology 146, 879e888.
784e790. Cheng, H., Leblond, C.P., 1974. Origin, differentiation and renewal of the four main
Ariyasu, H., Takaya, K., Tagami, T., Ogawa, Y., Hosoda, K., Akamizu, T., Suda, M., epithelial cell types in the mouse small intestine. V. Unitarian theory of the
Koh, T., Natsui, K., Toyooka, S., Shirakami, G., Usui, T., Shimatsu, A., Doi, K., origin of the four epithelial cell types. Am. J. Anat. 141, 537e561.
Hosoda, H., Kojima, M., Kangawa, K., Nakao, K., 2001. Stomach is a major source Cherbut, C., Ferrier, L., Roze, C., Anini, Y., Blottiere, H., Lecannu, G., Galmiche, J.P.,
of circulating ghrelin, and feeding state determines plasma ghrelin-like 1998. Short-chain fatty acids modify colonic motility through nerves and
immunoreactivity levels in humans. J. Clin. Endocrinol. Metab. 86, 4753e4758. polypeptide YY release in the rat. Am. J. Physiol. 275, G1415eG1422.
Asakawa, A., Inui, A., Yuzuriha, H., Ueno, N., Katsuura, G., Fujimiya, M., Fujino, M.A., Christensen, R., Kristensen, P.K., Bartels, E.M., Bliddal, H., Astrup, A., 2007. Efficacy
Niijima, A., Meguid, M.M., Kasuga, M., 2003. Characterization of the effects of and Safety of the weight loss drug Rimonabant: a meta-analysis of randomised
pancreatic polypeptide in the regulation of energy balance. Gastroenterology trials. The Lancet 370, 1706e1713.
124, 1325e1336. Chu, Z.L., Jones, R.M., He, H., Carroll, C., Gutierrez, V., Lucman, A., Moloney, M.,
Astrup, A., Carraro, R., Finer, N., Harper, A., Kunesova, M., Lean, M.E., Niskanen, L., Gao, H., Mondala, H., Bagnol, D., Unett, D., Liang, Y., Demarest, K., Semple, G.,
Rasmussen, M.F., Rissanen, A., Rossner, S., Savolainen, M.J., Van Gaal, L., 2011. Behan, D.P., Leonard, J., 2007. A role for beta-cell-expressed G protein-coupled
Safety, tolerability and sustained weight loss over 2 years with the once-daily receptor 119 in glycemic control by enhancing glucose-dependent insulin
human GLP-1 analog, liraglutide. Int. J. Obes. (Lond). release. Endocrinology 148, 2601e2609.
Badman, M.K., Flier, J.S., 2005. The gut and energy balance: visceral allies in the Chu, Z.L., Carroll, C., Alfonso, J., Gutierrez, V., He, H., Lucman, A., Pedraza, M.,
obesity wars. Science 307, 1909e1914. Mondala, H., Gao, H., Bagnol, D., Chen, R., Jones, R.M., Behan, D.P., Leonard, J.,
Baggio, L.L., Huang, Q., Brown, T.J., Drucker, D.J., 2004. Oxyntomodulin and 2008. A role for intestinal endocrine cell-expressed g protein-coupled receptor
glucagon-like peptide-1 differentially regulate murine food intake and energy 119 in glycemic control by enhancing glucagon-like Peptide-1 and glucose-
expenditure. Gastroenterology 127, 546e558. dependent insulinotropic Peptide release. Endocrinology 149, 2038e2047.
Baraboi, E.D., Michel, C., Smith, P., Thibaudeau, K., Ferguson, A.V., Richard, D., 2010. Chuang, J.C., Perello, M., Sakata, I., Osborne-Lawrence, S., Savitt, J.M., Lutter, M.,
Effects of albumin-conjugated PYY on food intake: the respective roles of the Zigman, J.M., 2011. Ghrelin mediates stress-induced food-reward behavior in
circumventricular organs and vagus nerve. Eur. J. Neurosci. 32, 826e839. mice. J. Clin. Invest. 121, 2684e2692.
Batterham, R.L., Cowley, M.A., Small, C.J., Herzog, H., Cohen, M.A., Dakin, C.L., Cohen, M.A., Ellis, S.M., Le Roux, C.W., Batterham, R.L., Park, A., Patterson, M.,
Wren, A.M., Brynes, A.E., Low, M.J., Ghatei, M.A., Cone, R.D., Bloom, S.R., 2002. Gut Frost, G.S., Ghatei, M.A., Bloom, S.R., 2003. Oxyntomodulin suppresses appetite
hormone PYY(3-36) physiologically inhibits food intake. Nature 418, 650e654. and reduces food intake in humans. J. Clin. Endocrinol. Metab. 88, 4696e4701.
Batterham, R.L., Cohen, M.A., Ellis, S.M., Le Roux, C.W., Withers, D.J., Frost, G.S., Cooke, J.H., Patterson, M., Patel, S.R., Smith, K.L., Ghatei, M.A., Bloom, S.R.,
Ghatei, M.A., Bloom, S.R., 2003a. Inhibition of food intake in obese subjects by Murphy, K.G., 2009. Peripheral and central administration of xenin and neu-
peptide YY3-36. N. Engl. J. Med. 349, 941e948. rotensin suppress food intake in rodents. Obesity (Silver Spring) 17, 1135e1143.
Batterham, R.L., Le Roux, C.W., Cohen, M.A., Park, A.J., Ellis, S.M., Patterson, M., Covasa, M., Marcuson, J.K., Ritter, R.C., 2001. Diminished satiation in rats exposed to
Frost, G.S., Ghatei, M.A., Bloom, S.R., 2003b. Pancreatic polypeptide reduces elevated levels of endogenous or exogenous cholecystokinin. Am. J. Physiol.
appetite and food intake in humans. J. Clin. Endocrinol. Metab. 88, 3989e3992. Regul. Integr. Comp. Physiol. 280, R331eR337.
Batterham, R.L., Heffron, H., Kapoor, S., Chivers, J.E., Chandarana, K., Herzog, H., Le Cummings, D.E., Overduin, J., 2007. Gastrointestinal regulation of food intake. J. Clin.
Roux, C.W., Thomas, E.L., Bell, J.D., Withers, D.J., 2006. Critical role for peptide Invest. 117, 13e23.
YY in protein-mediated satiation and body-weight regulation. Cell Metab. 4, Cummings, D.E., Weigle, D.S., Frayo, R.S., Breen, P.A., Ma, M.K., Dellinger, E.P.,
223e233. Purnell, J.Q., 2002. Plasma ghrelin levels after diet-induced weight loss or
Berntson, G.G., Zipf, W.B., O’Dorisio, T.M., Hoffman, J.A., Chance, R.E., 1993. gastric bypass surgery. N. Engl. J. Med. 346, 1623e1630.
Pancreatic polypeptide infusions reduce food intake in PradereWilli syndrome. Dakin, C.L., Gunn, I., Small, C.J., Edwards, C.M., Hay, D.L., Smith, D.M., Ghatei, M.A.,
Peptides 14, 497e503. Bloom, S.R., 2001. Oxyntomodulin inhibits food intake in the rat. Endocrinology
Bewick, G.A., Gardiner, J.V., Dhillo, W.S., Kent, A.S., White, N.E., Webster, Z., 142, 4244e4250.
Ghatei, M.A., Bloom, S.R., 2005. Post-embryonic ablation of AgRP neurons in Dakin, C.L., Small, C.J., Park, A.J., Seth, A., Ghatei, M.A., Bloom, S.R., 2002. Repeated
mice leads to a lean, hypophagic phenotype. FASEB J. 19, 1680e1682. ICV administration of oxyntomodulin causes a greater reduction in body weight
Bewick, G.A., Kent, A., Campbell, D., Patterson, M., Ghatei, M.A., Bloom, S.R., gain than in pair-fed rats. Am. J. Physiol. Endocrinol. Metab. 283, E1173eE1177.
Gardiner, J.V., 2009. Mice with hyperghrelinemia are hyperphagic and glucose Dakin, C.L., Small, C.J., Batterham, R.L., Neary, N.M., Cohen, M.A., Patterson, M.,
intolerant and have reduced leptin sensitivity. Diabetes 58, 840e846. Ghatei, M.A., Bloom, S.R., 2004. Peripheral oxyntomodulin reduces food intake
Bi, S., Moran, T.H., 2002. Actions of CCK in the controls of food intake and body and body weight gain in rats. Endocrinology 145, 2687e2695.
weight: lessons from the CCK-A receptor deficient OLETF rat. Neuropeptides 36, Date, Y., Murakami, N., Toshinai, K., Matsukura, S., Niijima, A., Matsuo, H.,
171e181. Kangawa, K., Nakazato, M., 2002. The role of the gastric afferent vagal nerve in
Briggs, D.I., Enriori, P.J., Lemus, M.B., Cowley, M.A., Andrews, Z.B., 2010. Diet-induced ghrelin-induced feeding and growth hormone secretion in rats. Gastroenter-
obesity causes ghrelin resistance in arcuate NPY/AgRP neurons. Endocrinology ology 123, 1120e1128.
151, 4745e4755. Daugaard, J.R., Meier, E., Riber, D., Bæk, C. Æ, Larsen, K.S., June 2010. The New Dual
Broberger, C., Landry, M., Wong, H., Walsh, J.N., Hokfelt, T., 1997. Subtypes Y1 and Y2 Glucagon-GLP-1 Agonist ZP2929 Improves Glycemic Control and Reduces Body
of the neuropeptide Y receptor are respectively expressed in pro- Weight in Murine Models of Obesity and Type 2 diabetes. Poster at ADA, Orlando.
opiomelanocortin- and neuropeptide-Y-containing neurons of the rat hypo- Davidson, I.W., Salter, J.M., Best, C.H., 1960. The effect of glucagon on the metabolic
thalamic arcuate nucleus. Neuroendocrinology 66, 393e408. rate of rats. Am. J. Clin. Nutr. 8, 540e546.
Broberger, C., Johansen, J., Johansson, C., Schalling, M., Hokfelt, T., 1998. The Day, J.W., Ottaway, N., Patterson, J.T., Gelfanov, V., Smiley, D., Gidda, J., Findeisen, H.,
neuropeptide Y/agouti gene-related protein (AGRP) brain circuitry in normal, Bruemmer, D., Drucker, D.J., Chaudhary, N., Holland, J., Hembree, J.,
anorectic, and monosodium glutamate-treated mice. Proc. Natl. Acad. Sci. U S A Abplanalp, W., Grant, E., Ruehl, J., Wilson, H., Kirchner, H., Lockie, S.H.,
95, 15043e15048. Hofmann, S., Woods, S.C., Nogueiras, R., Pfluger, P.T., Perez-Tilve, D.,
Buse, J.B., Henry, R.R., Han, J., Kim, D.D., Fineman, M.S., Baron, A.D., 2004. Effects of DiMarchi, R., Tschöp, M.H., 2009. A new glucagon and GLP-1 co-agonist elimi-
exenatide (exendin-4) on glycemic control over 30 weeks in sulfonylurea- nates obesity in rodents. Nat. Chem. Biol. 5, 749e757.
treated patients with type 2 diabetes. Diabetes Care 27, 2628e2635. DeFronzo, R.A., Ratner, R.E., Han, J., Kim, D.D., Fineman, M.S., Baron, A.D., 2005. Effects
Carraway, R., Leeman, S.E., 1976. Characterization of radioimmunoassayable neu- of exenatide (exendin-4) on glycemic control and weight over 30 weeks in
rotensin in the rat. Its differential distribution in the central nervous system, metformin-treated patients with type 2 diabetes. Diabetes Care 28, 1092e1100.
small intestine, and stomach. J. Biol. Chem. 251, 7045e7052. Degen, L., Drewe, J., Piccoli, F., Grani, K., Oesch, S., Bunea, R., D’Amato, M.,
Challis, B.G., Pinnock, S.B., Coll, A.P., Carter, R.N., Dickson, S.L., O’Rahilly, S., 2003. Beglinger, C., 2007. Effect of CCK-1 receptor blockade on ghrelin and PYY
Acute effects of PYY3-36 on food intake and hypothalamic neuropeptide secretion in men. Am. J. Physiol. Regul. Integr. Comp. Physiol. 292,
expression in the mouse. Biochem. Biophys. Res. Commun. 311, 915e919. R1391eR1399.
Chan, J.L., Heist, K., DePaoli, A.M., Veldhuis, J.D., Mantzoros, C.S., 2003. The role of Dhanvantari, S., Seidah, N.G., Brubaker, P.L., 1996. Role of prohormone convertases
falling leptin levels in the neuroendocrine and metabolic adaptation to short- in the tissue-specific processing of proglucagon. Mol. Endocrinol. 10, 342e355.
term starvation in healthy men. J. Clin. Invest. 111, 1409e1421. Dixon, A.F., Dixon, J.B., O’Brien, P.E., 2005. Laparoscopic adjustable gastric banding
Chance, W.T., Balasubramaniam, A., Stallion, A., Fischer, J.E., 1993. Anorexia induces prolonged satiety: a randomized blind crossover study. J. Clin. Endo-
following the systemic injection of amylin. Brain Res. 607, 185e188. crinol. Metab. 90, 813e819.
54 A.H. Sam et al. / Neuropharmacology 63 (2012) 46e56

Drucker, D.J., Nauck, M.A., 2006. The incretin system: glucagon-like peptide-1 Kamegai, J., Tamura, H., Shimizu, T., Ishii, S., Sugihara, H., Wakabayashi, I., 2001. Chronic
receptor agonists and dipeptidyl peptidase-4 inhibitors in type 2 diabetes. central infusion of ghrelin increases hypothalamic neuropeptide Y and Agouti-
Lancet 368, 1696e1705. related protein mRNA levels and body weight in rats. Diabetes 50, 2438e2443.
Drucker, D.J., 2006. The biology of incretin hormones. Cell Metab. 3, 153e165. Karaki, S., Mitsui, R., Hayashi, H., Kato, I., Sugiya, H., Iwanaga, T., Furness, J.B.,
Elias, C.F., Lee, C., Kelly, J., Aschkenasi, C., Ahima, R.S., Couceyro, P.R., Kuhar, M.J., Kuwahara, A., 2006. Short-chain fatty acid receptor, GPR43, is expressed by
Saper, C.B., Elmquist, J.K., 1998. Leptin activates hypothalamic CART neurons enteroendocrine cells and mucosal mast cells in rat intestine. Cell Tissue Res.
projecting to the spinal cord. Neuron 21, 1375e1385. 324, 353e360.
Eng, J., Kleinman, W.A., Singh, L., Singh, G., Raufman, J.P., 1992. Isolation and char- Kellum, J.M., Kuemmerle, J.F., O’Dorisio, T.M., Rayford, P., Martin, D., Engle, K.,
acterization of exendin-4, an exendin-3 analogue, from Heloderma suspectum Wolf, L., Sugerman, H.J., 1990. Gastrointestinal hormone responses to meals
venom. Further evidence for an exendin receptor on dispersed acini from before and after gastric bypass and vertical banded gastroplasty. Ann. Surg. 211,
guinea pig pancreas. J. Biol. Chem. 267, 7402e7405. 763e770. discussion 770e761.
Feurle, G.E., Muller, B., Rix, E., 1987. Neurotensin induces hyperplasia of the pancreas Kendall, D.M., Riddle, M.C., Rosenstock, J., Zhuang, D., Kim, D.D., Fineman, M.S.,
and growth of the gastric antrum in rats. Gut, 19e23. 28 Suppl. Baron, A.D., 2005. Effects of exenatide (exendin-4) on glycemic control over 30
Field, B.C., Wren, A.M., Peters, V., Baynes, K.C., Martin, N.M., Patterson, M., Alsaraf, S., weeks in patients with type 2 diabetes treated with metformin and a sulfo-
Amber, V., Wynne, K., Ghatei, M.A., Bloom, S.R., 2010. PYY3-36 and oxy- nylurea. Diabetes Care 28, 1083e1091.
ntomodulin can be additive in their effect on food intake in overweight and Kenler, H.A., Brolin, R.E., Cody, R.P., 1990. Changes in eating behavior after horizontal
obese humans. Diabetes 59, 1635e1639. gastroplasty and Roux-en-Y gastric bypass. Am. J. Clin. Nutr. 52, 87e92.
Fried, M., Erlacher, U., Schwizer, W., Lochner, C., Koerfer, J., Beglinger, C., Jansen, J.B., Kitabgi, P., 2006. Prohormone convertases differentially process pro-neurotensin/
Lamers, C.B., Harder, F., Bischof-Delaloye, A., et al., 1991. Role of cholecystokinin neuromedin N in tissues and cell lines. J. Mol. Med. 84, 628e634.
in the regulation of gastric emptying and pancreatic enzyme secretion in Koda, S., Date, Y., Murakami, N., Shimbara, T., Hanada, T., Toshinai, K., Niijima, A.,
humans. Studies with the cholecystokinin-receptor antagonist loxiglumide. Furuya, M., Inomata, N., Osuye, K., Nakazato, M., 2005. The role of the vagal
Gastroenterology 101, 503e511. nerve in peripheral PYY3-36-induced feeding reduction in rats. Endocrinology
Fu-Cheng, X., Anini, Y., Chariot, J., Voisin, T., Galmiche, J.P., Roze, C., 1995. Peptide YY 146, 2369e2375.
release after intraduodenal, intraileal, and intracolonic administration of Kojima, M., Hosoda, H., Date, Y., Nakazato, M., Matsuo, H., Kangawa, K., 1999.
nutrients in rats. Pflugers Arch. 431, 66e75. Ghrelin is a growth-hormone-releasing acylated peptide from stomach. Nature
Gan, S.S., Talbot, M.L., Jorgensen, J.O., 2007. Efficacy of surgery in the management of 402, 656e660.
obesity-related type 2 diabetes mellitus. ANZ J. Surg. 77, 958e962. Kopin, A.S., Mathes, W.F., McBride, E.W., Nguyen, M., Al-Haider, W., Schmitz, F.,
Gardiner, J.V., Campbell, D., Patterson, M., Kent, A., Ghatei, M.A., Bloom, S.R., Bonner-Weir, S., Kanarek, R., Beinborn, M., 1999. The cholecystokinin-A receptor
Bewick, G.A., 2010. The hyperphagic effect of ghrelin is inhibited in mice by mediates inhibition of food intake yet is not essential for the maintenance of
a diet high in fat. Gastroenterology 138, 2468e2476. 2476 e2461. body weight. J. Clin. Invest. 103, 383e391.
Geary, N., Le Sauter, J., Noh, U., 1993. Glucagon acts in the liver to control sponta- Larhammar, D., 1996. Structural diversity of receptors for neuropeptide Y, peptide
neous meal size in rats. Am. J. Physiol. 264, R116eR122. YY and pancreatic polypeptide. Regul. Pept. 65, 165e174.
Ghamari-Langroudi, M., Colmers, W.F., Cone, R.D., 2005. PYY3-36 inhibits the action Larsen, P.J., Kristensen, P., 1997. The neuropeptide Y (Y4) receptor is highly expressed
potential firing activity of POMC neurons of arcuate nucleus through post- in neurones of the rat dorsal vagal complex. Brain Res. Mol. Brain Res. 48, 1e6.
synaptic Y2 receptors. Cell Metab. 2, 191e199. Larsson, L.I., Sundler, F., Hakanson, R., 1975. Immunohistochemical localization of
Ghourab, S., Beale, K.E., Semjonous, N.M., Simpson, K.A., Martin, N.M., human pancreatic polypeptide (HPP) to a population of islet cells. Cell Tissue
Ghatei, M.A., Bloom, S.R., Smith, K.L., 2011. Intracerebroventricular adminis- Res. 156, 167e171.
tration of vasoactive intestinal peptide inhibits food intake. Regul. Pept. (Epub Lauffer, L.M., Iakoubov, R., Brubaker, P.L., 2009. GPR119 is essential for
ahead of print). oleoylethanolamide-induced glucagon-like peptide-1 secretion from the
Grandt, D., Teyssen, S., Schimiczek, M., Reeve Jr., J.R., Feth, F., Rascher, W., Hirche, H., intestinal enteroendocrine L-cell. Diabetes 58, 1058e1066.
Singer, M.V., Layer, P., Goebell, H., et al., 1992. Novel generation of hormone Lawrence, C.B., Snape, A.C., Baudoin, F.M., Luckman, S.M., 2002. Acute central
receptor specificity by amino terminal processing of peptide YY. Biochem. ghrelin and GH secretagogues induce feeding and activate brain appetite
Biophys. Res. Commun. 186, 1299e1306. centers. Endocrinology 143, 155e162.
Grandt, D., Schimiczek, M., Struk, K., Shively, J., Eysselein, V.E., Goebell, H., Le Quellec, A., Kervran, A., Blache, P., Ciurana, A.J., Bataille, D., 1992. Oxyntomodulin-
Reeve Jr., J.R., 1994. Characterization of two forms of peptide YY, PYY(1-36) and like immunoreactivity: diurnal profile of a new potential enterogastrone. J. Clin.
PYY(3-36), in the rabbit. Peptides 15, 815e820. Endocrinol. Metab. 74, 1405e1409.
Greenberg, G.R., McCloy, R.F., Chadwick, V.S., Adrian, T.E., Baron, J.H., Bloom, S.R., le Roux, C.W., Aylwin, S.J., Batterham, R.L., Borg, C.M., Coyle, F., Prasad, V., Shurey, S.,
1979. Effect of bovine pancreatic polypeptide on basal pancreatic and biliary Ghatei, M.A., Patel, A.G., Bloom, S.R., 2006. Gut hormone profiles following
outputs in man. Dig. Dis. Sci. 24, 11e14. bariatric surgery favor an anorectic state, facilitate weight loss, and improve
Hahn, T.M., Breininger, J.F., Baskin, D.G., Schwartz, M.W., 1998. Coexpression of Agrp metabolic parameters. Ann. Surg. 243, 108e114.
and NPY in fasting-activated hypothalamic neurons. Nat. Neurosci. 1, 271e272. le Roux, C.W., Welbourn, R., Werling, M., Osborne, A., Kokkinos, A., Laurenius, A.,
Halatchev, I.G., Ellacott, K.L., Fan, W., Cone, R.D., 2004. Peptide YY3-36 inhibits food Lonroth, H., Fandriks, L., Ghatei, M.A., Bloom, S.R., Olbers, T., 2007. Gut
intake in mice through a melanocortin-4 receptor-independent mechanism. hormones as mediators of appetite and weight loss after Roux-en-Y gastric
Endocrinology 145, 2585e2590. bypass. Ann. Surg. 246, 780e785.
Hankir, M.K., Parkinson, J.R., Minnion, J.S., Addison, M.L., Bloom, S.R., Bell, J.D., 2011. le Roux, C.W., Borg, C., Wallis, K., Vincent, R.P., Bueter, M., Goodlad, R., Ghatei, M.A.,
Peptide YY 3-36 and pancreatic polypeptide differentially regulate hypotha- Patel, A., Bloom, S.R., Aylwin, S.J., 2010. Gut hypertrophy after gastric bypass is
lamic neuronal activity in mice in vivo as measured by manganese-enhanced associated with increased glucagon-like peptide 2 and intestinal crypt cell
magnetic resonance imaging. J. Neuroendocrinol 23, 371e380. proliferation. Ann. Surg. 252, 50e56.
Herrmann, C., Goke, R., Richter, G., Fehmann, H.C., Arnold, R., Goke, B., 1995. Le Sauter, J., Noh, U., Geary, N., 1991. Hepatic portal infusion of glucagon antibodies
Glucagon-like peptide-1 and glucose-dependent insulin-releasing polypeptide increases spontaneous meal size in rats. Am. J. Physiol. 261, R162eR165.
plasma levels in response to nutrients. Digestion 56, 117e126. Liddle, R.A., Goldfine, I.D., Rosen, M.S., Taplitz, R.A., Williams, J.A., 1985. Cholecys-
Hewson, A.K., Dickson, S.L., 2000. Systemic administration of ghrelin induces Fos tokinin bioactivity in human plasma. Molecular forms, responses to feeding,
and Egr-1 proteins in the hypothalamic arcuate nucleus of fasted and fed rats. and relationship to gallbladder contraction. J. Clin. Invest. 75, 1144e1152.
J. Neuroendocrinol. 12, 1047e1049. Lieverse, R.J., Jansen, J.B., Masclee, A.A., Lamers, C.B., 1995. Satiety effects of
Imeryuz, N., Yegen, B.C., Bozkurt, A., Coskun, T., Villanueva-Penacarrillo, M.L., a physiological dose of cholecystokinin in humans. Gut 36, 176e179.
Ulusoy, N.B., 1997. Glucagon-like peptide-1 inhibits gastric emptying via vagal Lin, H.C., Zhao, X.T., Wang, L., Wong, H., 1996. Fat-induced ileal brake in the dog
afferent-mediated central mechanisms. Am. J. Physiol. 273, G920eG927. depends on peptide YY. Gastroenterology 110, 1491e1495.
James, W.P.T., Caterson, I.D., Coutinho, W., Finer, N., Van Gaal, L.F., Maggioni, A.P., Liu, Y.L., Semjonous, N.M., Murphy, K.G., Ghatei, M.A., Bloom, S.R., 2008. The effects
Torp-Pedersen, C., Sharma, A.M., Shepherd, G.M., Rode, R.A., Renz, C.L., 2010. of pancreatic polypeptide on locomotor activity and food intake in mice. Int. J.
Effect of Sibutramine on Cardiovascular outcomes in overweight and obese Obes. (Lond) 32, 1712e1715.
subjects. N. Engl. J. Med. 363, 905e917. Lundberg, J.M., Tatemoto, K., Terenius, L., Hellstrom, P.M., Mutt, V., Hokfelt, T.,
Jang, H.J., Kokrashvili, Z., Theodorakis, M.J., Carlson, O.D., Kim, B.J., Zhou, J., Hamberger, B., 1982. Localization of peptide YY (PYY) in gastrointestinal
Kim, H.H., Xu, X., Chan, S.L., Juhaszova, M., Bernier, M., Mosinger, B., endocrine cells and effects on intestinal blood flow and motility. Proc. Natl.
Margolskee, R.F., Egan, J.M., 2007. Gut-expressed gustducin and taste receptors Acad. Sci. U S A 79, 4471e4475.
regulate secretion of glucagon-like peptide-1. Proc. Natl. Acad. Sci. U S A 104, Lutz, T.A., Del Prete, E., Scharrer, E., 1994. Reduction of food intake in rats by
15069e15074. intraperitoneal injection of low doses of amylin. Physiol. Behav. 55, 891e895.
Jesudason, D.R., Monteiro, M.P., McGowan, B.M., Neary, N.M., Park, A.J., Philippou, E., MacDonald, P.E., El-Kholy, W., Riedel, M.J., Salapatek, A.M., Light, P.E., Wheeler, M.B.,
Small, C.J., Frost, G.S., Ghatei, M.A., Bloom, S.R., 2007. Low-dose pancreatic 2002. The multiple actions of GLP-1 on the process of glucose-stimulated
polypeptide inhibits food intake in man. Br. J. Nutr. 97, 426e429. insulin secretion. Diabetes 51 (Suppl. 3), S434eS442.
Jobst, E.E., Enriori, P.J., Cowley, M.A., 2004. The electrophysiology of feeding circuits. Masuda, Y., Tanaka, T., Inomata, N., Ohnuma, N., Tanaka, S., Itoh, Z., Hosoda, H.,
Trends Endocrinol. Metab. 15, 488e499. Kojima, M., Kangawa, K., 2000. Ghrelin stimulates gastric acid secretion and
Jones, R.M., Leonard, J.N., Buzard, D.J., Lehmann, J., 2009. GPR119 agonists for the motility in rats. Biochem. Biophys. Res. Commun. 276, 905e908.
treatment of type 2 diabetes. Expert Opin. Ther. Pat. 19, 1339e1359. Medeiros, M.D., Turner, A.J., 1994. Processing and metabolism of peptide-YY: pivotal
Joy, S.V., Rodgers, P.T., Scates, A.C., 2005. Incretin mimetics as emerging treatments roles of dipeptidylpeptidase-IV, aminopeptidase-P, and endopeptidase-24.11.
for type 2 diabetes. Ann. Pharmacother. 39, 110e118. Endocrinology 134, 2088e2094.
A.H. Sam et al. / Neuropharmacology 63 (2012) 46e56 55

Meeran, K., O’Shea, D., Edwards, C.M., Turton, M.D., Heath, M.M., Gunn, I., Roth, K.A., Kim, S., Gordon, J.I., 1992. Immunocytochemical studies suggest two
Abusnana, S., Rossi, M., Small, C.J., Goldstone, A.P., Taylor, G.M., Sunter, D., pathways for enteroendocrine cell differentiation in the colon. Am. J. Physiol.
Steere, J., Choi, S.J., Ghatei, M.A., Bloom, S.R., 1999. Repeated intra- 263, G174eG180.
cerebroventricular administration of glucagon-like peptide-1-(7e36) amide Rozengurt, N., Wu, S.V., Chen, M.C., Huang, C., Sternini, C., Rozengurt, E., 2006.
or exendin-(9e39) alters body weight in the rat. Endocrinology 140, Colocalization of the alpha-subunit of gustducin with PYY and GLP-1 in L cells of
244e250. human colon. Am. J. Physiol. Gastrointest. Liver Physiol. 291, G792eG802.
Michel, M.C., Beck-Sickinger, A., Cox, H., Doods, H.N., Herzog, H., Larhammar, D., Rucker, D., Padwal, R., Li, S.K., Curioni, C., Lau, D.C.W., 2007. Long term pharmaco-
Quirion, R., Schwartz, T., Westfall, T., 1998. XVI. International Union of Phar- therapy for obesity and overweight: updated meta-analysis. Br. Med. J. 335,
macology recommendations for the nomenclature of neuropeptide Y, peptide 1194e1199.
YY, and pancreatic polypeptide receptors. Pharmacol. Rev. 50, 143e150. Rushing, P.A., Hagan, M.M., Seeley, R.J., Lutz, T.A., Woods, S.C., 2000. Amylin: a novel
Moran, T.H., Kinzig, K.P., 2004. Gastrointestinal satiety signals II. Cholecystokinin. action in the brain to reduce body weight. Endocrinology 141, 850e853.
Am. J. Physiol. Gastrointest. Liver Physiol. 286, G183eG188. Sandoval, S.L., Kulkosky, P.J., 1992. Effects of peripheral neurotensin on behavior of
Moran, T.H., Robinson, P.H., Goldrich, M.S., McHugh, P.R., 1986. Two brain chole- the rat. Pharmacol. Biochem. Behav. 41, 385e390.
cystokinin receptors: implications for behavioral actions. Brain Res. 362, Sato, N., Kanai, S., Takano, S., Kurosawa, M., Funakoshi, A., Miyasaka, K., 2003.
175e179. Central administration of ghrelin stimulates pancreatic exocrine secretion via
Moran, T.H., 2000. Cholecystokinin and satiety: current perspectives. Nutrition 16, the vagus in conscious rats. Jpn. J. Physiol. 53, 443e449.
858e865. Savage, A.P., Adrian, T.E., Carolan, G., Chatterjee, V.K., Bloom, S.R., 1987. Effects of
Murphy, K.G., Bloom, S.R., 2006. Gut hormones and the regulation of energy peptide YY (PYY) on mouth to caecum intestinal transit time and on the rate of
homeostasis. Nature 444, 854e859. gastric emptying in healthy volunteers. Gut 28, 166e170.
Nair, K.S., 1987. Hyperglucagonaemia increases resting metabolic rate in man during Schirra, J., Nicolaus, M., Roggel, R., Katschinski, M., Storr, M., Woerle, H.J., Goke, B.,
insulin deficiency. J. Clin. Endocrinol. Metab. 64, 896e901. 2006. Endogenous glucagon-like peptide-1 controls endocrine pancreatic
Naslund, E., Bogefors, J., Skogar, S., Gryback, P., Jacobsson, H., Holst, J.J., secretion and antro-pyloro-duodenal motility in humans. Gut 55, 243e251.
Hellstrom, P.M., 1999. GLP-1 slows solid gastric emptying and inhibits insulin, Schjoldager, B., Mortensen, P.E., Myhre, J., Christiansen, J., Holst, J.J., 1989. Oxy-
glucagon, and PYY release in humans. Am. J. Physiol. 277, R910eR916. ntomodulin from distal gut. Role in regulation of gastric and pancreatic func-
Nauck, M.A., Niedereichholz, U., Ettler, R., Holst, J.J., Orskov, C., Ritzel, R., tions. Dig. Dis. Sci. 34, 1411e1419.
Schmiegel, W.H., 1997. Glucagon-like peptide 1 inhibition of gastric emptying Schmidt, P.T., Naslund, E., Gryback, P., Jacobsson, H., Hartmann, B., Holst, J.J.,
outweighs its insulinotropic effects in healthy humans. Am. J. Physiol. 273, Hellstrom, P.M., 2003. Peripheral administration of GLP-2 to humans has no
E981eE988. effect on gastric emptying or satiety. Regul. Pept. 116, 21e25.
Neary, N.M., Small, C.J., Druce, M.R., Park, A.J., Ellis, S.M., Semjonous, N.M., Schmidt, P.T., Naslund, E., Gryback, P., Jacobsson, H., Holst, J.J., Hilsted, L.,
Dakin, C.L., Filipsson, K., Wang, F., Kent, A.S., Frost, G.S., Ghatei, M.A., Bloom, S.R., Hellstrom, P.M., 2005. A role for pancreatic polypeptide in the regulation of
2005. Peptide YY3-36 and glucagon-like peptide-17-36 inhibit food intake gastric emptying and short-term metabolic control. J. Clin. Endocrinol. Metab.
additively. Endocrinology 146, 5120e5127. 90, 5241e5246.
Overton, H.A., Babbs, A.J., Doel, S.M., Fyfe, M.C., Gardner, L.S., Griffin, G., Schulman, J.L., Carleton, J.L., Whitney, G., Whitehorn, J.C., 1957. Effect of glucagon on
Jackson, H.C., Procter, M.J., Rasamison, C.M., Tang-Christensen, M., food intake and body weight in man. J. Appl. Physiol. 11, 419e421.
Widdowson, P.S., Williams, G.M., Reynet, C., 2006. Deorphanization of a G Schwartz, T.W., Holst, J.J., Fahrenkrug, J., Jensen, S.L., Nielsen, O.V., Rehfeld, J.F., de
protein-coupled receptor for oleoylethanolamide and its use in the discovery of Muckadell, O.B., Stadil, F., 1978. Vagal, cholinergic regulation of pancreatic
small-molecule hypophagic agents. Cell Metab. 3, 167e175. polypeptide secretion. J. Clin. Invest. 61, 781e789.
Page, A.J., Slattery, J.A., Milte, C., Laker, R., O’Donnell, T., Dorian, C., Brierley, S.M., Schwartz, M.W., Woods, S.C., Porte Jr., D., Seeley, R.J., Baskin, D.G., 2000. Central
Blackshaw, L.A., 2007. Ghrelin selectively reduces mechanosensitivity of upper nervous system control of food intake. Nature 404, 661e671.
gastrointestinal vagal afferents. Am. J. Physiol. Gastrointest. Liver Physiol. 292, Scott, R.B., Kirk, D., MacNaughton, W.K., Meddings, J.B., 1998. GLP-2 augments the
G1376eG1384. adaptive response to massive intestinal resection in rat. Am. J. Physiol. 275,
Parker, R.M., Herzog, H., 1999. Regional distribution of Y-receptor subtype mRNAs in G911eG921.
rat brain. Eur. J. Neurosci. 11, 1431e1448. Shah, U., Kowalski, T.J., 2010. GPR119 agonists for the potential treatment of type 2
Parkinson, J.R.C., Chaudhri, O.B., Kuo, Y.-T., Field, B.C.T., Herlihy, A.H., Dhillo, W.S., diabetes and related metabolic disorders. Vitam. Horm. 84, 415e448.
Ghatei, M.A., Bloom, S.R., Bell, J.D., 2009. Differential patterns of neuronal Shiiya, T., Nakazato, M., Mizuta, M., Date, Y., Mondal, M.S., Tanaka, M., Nozoe, S.,
activation in the brainstem and hypothalamus following peripheral injection of Hosoda, H., Kangawa, K., Matsukura, S., 2002. Plasma ghrelin levels in lean and
GLP-1, oxyntomodulin and lithium chloride in mice detected by manganese obese humans and the effect of glucose on ghrelin secretion. J. Clin. Endocrinol.
enhanced magnetic resonance imaging (MEMRI). Neuroimage 44, 1022e1031. Metab. 87, 240e244.
Pedersen-Bjergaard, U., Host, U., Kelbaek, H., Schifter, S., Rehfeld, J.F., Faber, J., Shughrue, P.J., Lane, M.V., Merchenthaler, I., 1996. Glucagon-like peptide-1 receptor
Christensen, N.J., 1996. Influence of meal composition on postprandial periph- (GLP1-R) mRNA in the rat hypothalamus. Endocrinology 137, 5159e5162.
eral plasma concentrations of vasoactive peptides in man. Scand. J. Clin. Lab. Sjolund, K., Sanden, G., Hakanson, R., Sundler, F., 1983. Endocrine cells in human
Invest. 56, 497e503. intestine: an immunocytochemical study. Gastroenterology 85, 1120e1130.
Perez-Tilve, D., Heppner, K., Kirchner, H., Lockie, S.H., Woods, S.C., Smiley, D.L., Sjostrom, L., Narbro, K., Sjostrom, C.D., Karason, K., Larsson, B., Wedel, H., Lystig, T.,
Tschop, M., Pfluger, P., 2011. Ghrelin-induced adiposity is independent of Sullivan, M., Bouchard, C., Carlsson, B., Bengtsson, C., Dahlgren, S., Gummesson, A.,
orexigenic effects. FASEB J. Aug 25 (8), 2814e2822. Jacobson, P., Karlsson, J., Lindroos, A.K., Lonroth, H., Naslund, I., Olbers, T., Stenlof, K.,
Peruzzo, B., Pastor, F.E., Blazquez, J.L., Schobitz, K., Pelaez, B., Amat, P., Torgerson, J., Agren, G., Carlsson, L.M., 2007. Effects of bariatric surgery on
Rodriguez, E.M., 2000. A second look at the barriers of the medial basal mortality in Swedish obese subjects. N. Engl. J. Med. 357, 741e752.
hypothalamus. Exp. Brain Res. 132, 10e26. Stevenson, R.W., Steiner, K.E., Davis, M.A., Hendrick, G.K., Williams, P.E., Lacy, W.W.,
Pocai, A., Carrington, P.E., Adams, J.R., Wright, M., Eiermann, G., Zhu, L., Du, X., Brown, L., Donahue, P., Lacy, D.B., Cherrington, A.D., 1987. Similar dose
Petrov, A., Lassman, M.E., Jiang, G., Liu, F., Miller, C., Tota, L.M., Zhou, G., Zhang, X., responsiveness to hepatic glycogenolysis and gluconeogenesis to glucagon
Sountis, M.M., Santoprete, A., Capito, E., Chicchi, G.G., Thornberry, N., Bianchi, E., in vivo. Diabetes 36, 382e389.
Pessi, A., Marsh, D.J., SinhaRoy, R., 2009. Glucagon-like peptide 1/Glucagon Striffler, J., Cardell, E.L., Cardell, R.R.J., 1981. Effects of glucagon on hepatic glycogen
receptor dual agonism reverses obesity in mice. Diabetes 58, 2258e2266. and smooth endoplasmic reticulum. Am. J. Anat. 160, 363e379.
Porte Jr., D., Baskin, D.G., Schwartz, M.W., 2002. Leptin and insulin action in the Studer, R.K., Snowdowne, K.W., Borle, A.B., 1984. Regulation of hepatic glycogen-
central nervous system. Nutr. Rev. 60, S20e29. discussion S68-84, 85e27. olysis by glucagon in Male and Female rats. J. Biol. Chem. 259, 3596e3604.
Prospective_Studies_Collaboration, 2009. Body-mass index and cause-specific Svoboda, M., Tastenoy, M., Vertongen, P., Robberecht, P., 1994. Relative quantitative
mortality in 900,000 adults: collaborative analyses of 57 prospective studies. analysis of glucagon receptor mRNA in rat tissues. Mol. Cell. Endocrinol. 105,
The Lancet 373, 1083e1096. 131e137.
Punjabi, M., Arnold, M., Geary, N., Langhans, W., Pacheco-Lopez, G., 2011. Peripheral Symersky, T., Biemond, I., Frolich, M., Masclee, A.A., 2005. Effect of peptide YY on
Glucagon-like peptide-1 (GLP-1) and Satiation. Physiol. Behav.. doi:10.1016/ pancreatico-biliary secretion in humans. Scand. J. Gastroenterol. 40, 944e949.
j.physbeh.2011.2,038. Szecowka, J., Tatemoto, K., Rajamaki, G., Efendic, S., 1983. Effects of PYY and PP on
Ratner, R., Whitehouse, F., Fineman, M.S., Strobel, S., Shen, L., Maggs, D.G., endocrine pancreas. Acta Physiol. Scand. 119, 123e126.
Kolterman, O.G., Weyer, C., 2005. Adjunctive therapy with pramlintide lowers Tamura, H., Kamegai, J., Shimizu, T., Ishii, S., Sugihara, H., Oikawa, S., 2002. Ghrelin
HbA1c without concomitant weight gain and increased risk of severe hypo- stimulates GH but not food intake in arcuate nucleus ablated rats. Endocri-
glycemia in patients with type 1 diabetes approaching glycemic targets. Exp. nology 143, 3268e3275.
Clin. Endocrinol. Diabetes 113, 199e204. Tan, T.M., Field, B.C., Minnion, J.S., Cuenco-Shillito, J., Chambers, E.S., Zac-Varghese, S.,
Rehfeld, J.F., Bungaard, J.R., Friis-Hansen, L., Goetze, J.P., 2003. On the tissue-specific Brindley, C.J., Mt-Isa, S., Fiorentino, F., Ashby, D., Ward, I., Ghatei, M.A., Bloom, S.R.,
processing of procholecystokinin in the brain and gutea short review. J. Physiol. 2011. Pharmacokinetics, adverse effects and tolerability of a novel analogue of
Pharmacol. 54 (4), 73e79. human pancreatic polypeptide. Br. J. Clin. Pharmacol., 1420.
Ringback Weitoft, G., Eliasson, M., Rosen, M., 2008. Underweight, overweight and Tang-Christensen, M., Vrang, N., Larsen, P.J., 2001. Glucagon-like peptide containing
obesity as risk factors for mortality and hospitalization. Scand. J. Public Health pathways in the regulation of feeding behaviour. Int. J. Obes. Relat. Metab.
36, 169e176. Disord. 25 (Suppl. 5), S42eS47.
Rosell, S., Rokaeus, A., 1979. The effect of ingestion of amino acids, glucose and fat Tazoe, H., Otomo, Y., Karaki, S., Kato, I., Fukami, Y., Terasaki, M., Kuwahara, A., 2009.
on circulating neurotensin-like immunoreactivity (NTLI) in man. Acta Physiol. Expression of short-chain fatty acid receptor GPR41 in the human colon.
Scand. 107, 263e267. Biomed. Res. 30, 149e156.
56 A.H. Sam et al. / Neuropharmacology 63 (2012) 46e56

Toshinai, K., Mondal, M.S., Nakazato, M., Date, Y., Murakami, N., Kojima, M., glycogenolysis and gluconeogenesis during muscular work. Am. J. Physiol. 257,
Kangawa, K., Matsukura, S., 2001. Upregulation of Ghrelin expression in the E108eE117.
stomach upon fasting, insulin-induced hypoglycemia, and leptin administra- Wen, J., Phillips, S.F., Sarr, M.G., Kost, L.J., Holst, J.J., 1995. PYY and GLP-1 contribute
tion. Biochem. Biophys. Res. Commun. 281, 1220e1225. to feedback inhibition from the canine ileum and colon. Am. J. Physiol. 269,
Tschop, M., Smiley, D.L., Heiman, M.L., 2000. Ghrelin induces adiposity in rodents. G945eG952.
Nature 407, 908e913. Whitehouse, F., Kruger, D.F., Fineman, M., Shen, L., Ruggles, J.A., Maggs, D.G.,
Tschop, M., Wawarta, R., Riepl, R.L., Friedrich, S., Bidlingmaier, M., Landgraf, R., Weyer, C., Kolterman, O.G., 2002. A randomized study and open-label extension
Folwaczny, C., 2001. Post-prandial decrease of circulating human ghrelin levels. evaluating the long-term efficacy of pramlintide as an adjunct to insulin
J. Endocrinol. Invest. 24, RC19eRC21. therapy in type 1 diabetes. Diabetes Care 25, 724e730.
Tschop, M., Castaneda, T.R., Joost, H.G., Thone-Reineke, C., Ortmann, S., Klaus, S., Willms, B., Werner, J., Holst, J.J., Orskov, C., Creutzfeldt, W., Nauck, M.A., 1996. Gastric
Hagan, M.M., Chandler, P.C., Oswald, K.D., Benoit, S.C., Seeley, R.J., Kinzig, K.P., emptying, glucose responses, and insulin secretion after a liquid test meal: effects
Moran, T.H., Beck-Sickinger, A.G., Koglin, N., Rodgers, R.J., Blundell, J.E., Ishii, Y., of exogenous glucagon-like peptide-1 (GLP-1)-(7-36) amide in type 2 (non-
Beattie, A.H., Holch, P., Allison, D.B., Raun, K., Madsen, K., Wulff, B.S., insulin-dependent) diabetic patients. J. Clin. Endocrinol. Metab. 81, 327e332.
Stidsen, C.E., Birringer, M., Kreuzer, O.J., Schindler, M., Arndt, K., Rudolf, K., Wood, J.G., Hoang, H.D., Bussjaeger, L.J., Solomon, T.E., 1988a. Effect of neurotensin
Mark, M., Deng, X.Y., Whitcomb, D.C., Halem, H., Taylor, J., Dong, J., Datta, R., on pancreatic and gastric secretion and growth in rats. Pancreas 3, 332e339.
Culler, M., Craney, S., Flora, D., Smiley, D., Heiman, M.L., 2004. Physiology: does Wood, J.G., Hoang, H.D., Bussjaeger, L.J., Solomon, T.E., 1988b. Neurotensin stimu-
gut hormone PYY3-36 decrease food intake in rodents? Nature 430, 1 pp. lates growth of small intestine in rats. Am. J. Physiol. 255, G813eG817.
following 165; discussion 162 pp. following 165. Wren, A.M., Small, C.J., Ward, H.L., Murphy, K.G., Dakin, C.L., Taheri, S., Kennedy, A.R.,
Tsubouchi, S., Leblond, C.P., 1979. Migration and turnover of entero-endocrine and Roberts, G.H., Morgan, D.G., Ghatei, M.A., Bloom, S.R., 2000. The novel hypo-
caveolated cells in the epithelium of the descending colon, as shown by thalamic peptide ghrelin stimulates food intake and growth hormone secretion.
radioautography after continuous infusion of 3H-thymidine into mice. Am. J. Endocrinology 141, 4325e4328.
Anat. 156, 431e451. Wynne, K., Park, A.J., Small, C.J., Patterson, M., Ellis, S.M., Murphy, K.G., Wren, A.M.,
Turton, M.D., O’Shea, D., Gunn, I., Beak, S.A., Edwards, C.M., Meeran, K., Choi, S.J., Frost, G.S., Meeran, K., Ghatei, M.A., Bloom, S.R., 2005. Subcutaneous oxy-
Taylor, G.M., Heath, M.M., Lambert, P.D., Wilding, J.P., Smith, D.M., Ghatei, M.A., ntomodulin reduces body weight in overweight and obese subjects: a double-
Herbert, J., Bloom, S.R., 1996. A role for glucagon-like peptide-1 in the central blind, randomized, controlled trial. Diabetes 54, 2390e2395.
regulation of feeding. Nature 379, 69e72. Wynne, K., Park, A.J., Small, C.J., Meeran, K., Ghatei, M.A., Frost, G.S., Bloom, S.R.,
Ueno, N., Inui, A., Iwamoto, M., Kaga, T., Asakawa, A., Okita, M., Fujimiya, M., 2006. Oxyntomodulin increases energy expenditure in addition to decreasing
Nakajima, Y., Ohmoto, Y., Ohnaka, M., Nakaya, Y., Miyazaki, J.I., Kasuga, M., 1999. energy intake in overweight and obese humans: a randomised controlled trial.
Decreased food intake and body weight in pancreatic polypeptide- Int. J. Obes. (Lond) 30, 1729e1736.
overexpressing mice. Gastroenterology 117, 1427e1432. Zheng, W., McLerran, D.F., Rolland, B., Zhang, X., Inoue, M., Matsuo, K., He, J.,
Verdich, C., Flint, A., Gutzwiller, J.P., Naslund, E., Beglinger, C., Hellstrom, P.M., Gupta, P.C., Ramadas, Kunnambath, Tsugane, Shoichiro, Irie, Fujiko,
Long, S.J., Morgan, L.M., Holst, J.J., Astrup, A., 2001. A meta-analysis of the effect Tamakoshi, Akiko, Gao, Yu-Tang, Wang, Renwei, Shu, Xiao-Ou, Tsuji, Ichiro,
of glucagon-like peptide-1 (7-36) amide on ad libitum energy intake in Kuriyama, Shinichi, Tanaka, Hideo, Satoh, Hiroshi, Chen, Chien-Jen, Yuan, Jian-
humans. J. Clin. Endocrinol. Metab. 86, 4382e4389. Min, Yoo, Keun-Young, Ahsan, Habibul, Pan, Wen-Harn, Gu, Dongfeng,
Vrang, N., Madsen, A.N., Tang-Christensen, M., Hansen, G., Larsen, P.J., 2006. PYY(3-36) Pednekar, Mangesh Suryakant, Sauvaget, Catherine, Sasazuki, Shizuka,
reduces food intake and body weight and improves insulin sensitivity in rodent Sairenchi, Toshimi, Yang, Gong, Xiang, Yong-Bing, Nagai, Masato,
models of diet-induced obesity. Am. J. Physiol. Regul. Integr. Comp. Physiol. 291, Suzuki, Takeshi, Nishino, Yoshikazu, You, San-Lin, Koh, Woon-Puay, Park, Sue K.,
R367eR375. Chen, Yu, Shen, Chen-Yang, Thornquist, Mark, Feng, Ziding, Kang, Daehee,
Wasserman, D.H., Spalding, J.A., Lacy, D.B., Colburn, C.A., Goldstein, R.E., Boffetta, Paolo, Potter, John D., 2011. Association between body-mass index and
Cherrington, A.D., 1989. Glucagon is a primary controller of hepatic risk of death in more than 1 million Asians. N. Engl. J. Med. 364, 719e729.

You might also like