ArticlePDF AvailableLiterature Review

Abstract and Figures

GLP-1 receptors are expressed in the brain, especially in the regions responsible for the regulation of food intake, and intracerebroventricular injection of GLP-1 results in inhibition of food intake. Peripheral administration of GLP-1 dose-dependently enhances satiety and reduces food intake in normal and obese subjects as well as in type 2 diabetic patients. So far, the mechanisms by which GLP-1 exerts its effects are not completely clear. Interactions with neurons in the gastrointestinal tract or possibly direct access to the brain through the blood-brain barrier as observed in rats are possible and discussed in this chapter as well as a novel hypothesis based on the finding that GLP-1 is also expressed in taste cells. Finally, the role of GLP-1 receptor agonists as a possible treatment option in obesity is discussed as well as the role of GLP-1 in the effects of bariatric surgery on adiposity and glucose homeostasis.
Content may be subject to copyright.
Anorexigenic Effects of GLP-1 and Its Analogues
Baptist Gallwitz
Contents
1 Incretin Hormones................... ....................................................... 186
2 Biosynthesis of GLP-1 and Overview on Its Pleiotropic Actions ........................ 186
3 Central Effects of GLP-1 . . . . . . ............................................................. 189
3.1 Direct Dentral Effects of GLP-1 in the Hypothalamus ............................. 189
3.2 GLP-1 in Taste Cells . . . . . . . . . . . . . . . .. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .. . . . . . . . . . . . 192
3.3 Central Effects of GLP-1 Influencing Gastrointestinal Functions .................. 192
4 Peripheral Effects of GLP-1 Affecting Satiety . . . ......................................... 193
5 Effects of GLP-1 Receptor Agonists on Body Weight in Humans . . . .................... 196
6 GLP-1 and Bariatric Surgery . . . ............................................................ 199
References ....................................... ................................................ 201
Abstract GLP-1 receptors are expressed in the brain, especially in the regions
responsible for the regulation of food intake, and intracerebroventricular injection
of GLP-1 results in inhibition of food intake. Peripheral administration of GLP-1
dose-dependently enhances satiety and reduces food intake in normal and obese
subjects as well as in type 2 diabetic patients. So far, the mechanisms by which
GLP-1 exerts its effects are not completely clear. Interactions with neurons in the
gastrointestinal tract or possibly direct access to the brain through the blood–brain
barrier as observed in rats are possible and discussed in this chapter as well as
a novel hypothesis based on the finding that GLP-1 is also expressed in taste cells.
Finally, the role of GLP-1 receptor agonists as a possible treatment option in obesity
is discussed as well as the role of GLP-1 in the effects of bariatric surgery on
adiposity and glucose homeostasis.
B. Gallwitz (*)
Medizinische Klinik IV, Otfried-M
uller-Str. 10, 72076 T
ubingen, Germany
e-mail: baptist.gallwitz@med.uni-tuebingen.de
H.-G. Joost (ed.), Appetite Control, Handbook of Experimental Pharmacology 209,
DOI 10.1007/978-3-642-24716-3_8, #Springer-Verlag Berlin Heidelberg 2012
185
Keywords Bariatric surgery • GLP-1 • GLP-1 receptor agonists • Gut-brain axis •
Incretins • Obesity
1 Incretin Hormones
Upon stimulation by nutrients after a meal, the intestinal mucosa secretes the
gastrointestinal hormones GLP-1 and GIP (gastric inhibitory polypeptide or glu-
cose-dependent insulinotropic polypeptide) from the endocrine L and K cells,
respectively (Wellendorph et al. 2009). Both hormones are responsible for approxi-
mately 60% of the postprandial insulin secretion and contribute to the so-called
incretin effect. This effect describes the phenomenon that orally ingested glucose
leads to a much larger insulin response than an isoglycemic, intravenous glucose
load (Creutzfeldt 1979; Nauck et al. 1986).
Exogenous GLP-1 application either by subcutaneous or intravenous injection
resulting in supraphysiological GLP-1 plasma concentrations restores the incretin
effect with an adequate insulin response under hyperglycemic conditions (Nauck
et al. 1993).
2 Biosynthesis of GLP-1 and Overview on Its Pleiotropic
Actions
The glucagon gene encodes a large peptide sequence of 158 amino acids that
contains not only the sequence of glucagon but also that of other peptides that are
formed posttranslationally by organ-specific and cell-specific processing (Bell et al.
1983; Holst et al. 2007; Ørskov et al. 1987). In the neuroendocrine L cells of the
intestinal mucosa and in the central nervous system, preproglucagon is cleaved
mainly to generate GLP-1. In the pancreatic alpha cells of the islets, glucagon is the
major biologically active peptide generated from preproglucagon. Figure 1shows
the schematic tissue-specific posttranslational processing of GLP-1.
GLP-1 binds to highly specific GLP-1 receptors that belong to the G protein
coupled seven-transmembrane-spanning (7TM) receptors (Drucker and Nauck
2006). After binding to its receptor, adenylate cyclase is activated, and GLP-1
effects are mediated mainly via the cAMP and protein kinase A pathways
(Gromada et al. 1996; Reimer 2006). GLP-1 shows numerous physiological actions
in various tissues and a broad therapeutic potential (see Fig. 2for details).
The two most important metabolic actions of GLP-1 are that it stimulates insulin
secretion of the pancreatic beta cells and additionally inhibits glucagon secretion
from the alpha cells. These two actions on the islet occur in a strictly glucose-
dependent manner and lead to a normalization of hyperglycemia. Under hypogly-
cemic conditions, the counter-regulation by glucagon is not affected, and insulin
secretion is not stimulated. GLP-1 is therefore not able to elicit hypoglycemia by
186 B. Gallwitz
itself. These physiological properties of GLP-1 later translated into the pharmaco-
therapy of type 2 diabetes with incretin-based therapies (Drucker and Nauck 2006).
Like other gastrointestinal regulatory peptides, GLP-1 has multiple further
actions. In the gastrointestinal tract, GLP-1 slows gastric emptying after a meal.
This effect also contributes to a normalization of postprandial hyperglycemia,
promotes a feeling of fullness and possibly a secondary reduction of appetite
(Meier et al. 2003a; Holst et al. 2008). In addition, GLP-1 binds to its receptor on
hypothalamic neurons and stimulates satiety by direct actions described in detail in
this chapter. These two satiety-promoting effects explain that long-term treatment
with GLP-1 receptor agonists leads to long-term weight loss that persists as long as
GLP-1 is given (Drucker and Nauck 2006).
Animal studies in different rodent species and studies in isolated human islets
showed beneficial long-term actions of GLP-1: insulin synthesis is stimulated, and
beta-cell mass is restored in rodent models of type 2 diabetes (Brubaker and
Drucker 2004; Drucker and Nauck 2006; Fehmann and Habener 1992). Presently,
it is not known whether these findings reflect an additional benefit in type 2 diabetes
therapy in that GLP-1 slows or even stops disease progression. Long-term study
data from clinical studies or clinical use of GLP-1 receptor agonists (GLP-1 RA) in
type 2 diabetes with a sufficient observation time are still not yet available.
delays gastri c em ptyin g
reduces di gestive secretions
delays gastri c em ptyin g
reduces di gestive secretions
stimulates muco sal growth
stimulates hepatic
glucose pruduction
functi ons
see Fig. 2
Fig. 1 Posttranslational processing of proglucagon in different tissues. Differential posttransla-
tional processing of proglucagon in the pancreas and in the gut and brain. The numbers indicate
amino acid positions in the 160-amino acid proglucagon sequence. The vertical lines indicate
positions of basic amino acid residues, typical cleavage sites. GRPP glicentin-related pancreatic
polypeptide, IP-1 intervening peptide-1, IP-2 intervening peptide-2. The major known biological
actions of the peptides resulting from proglucagon processing are also shown (adapted from Holst
2007 and Pellissier et al. 2004)
Anorexigenic Effects of GLP-1 and Its Analogues 187
Furthermore, there are presently no reliable, validated methods to quantify beta cell
mass in humans in a clinical setting.
Additionally, recent studies demonstrated that therapeutic application of GLP-1
or GLP-1 RA improved cardiovascular parameters. Systolic blood pressure is
lowered by GLP-1 RA treatment for type 2 diabetes, and beneficial effects of
GLP-1 on myocardial ischemia were observed in animal models as well as positive
effects on left ventricular function in heart failure. These promising effects may
also have important clinical implications for type 2 diabetes therapy with GLP-1
RA (Courreges et al. 2008; Klonoff et al. 2008; Sokos et al. 2006).
GLP-1 receptors are also expressed in the brain, especially in the regions
responsible for the regulation of food intake (G
oke et al. 1995), and intracerebro-
ventricular injection of GLP-1 results in inhibition of food intake (Tang-Christensen
Heart
Cardioprotection
Neuroprotection
Appetite
Gastric emptying
Glucose production
Insulin secretion
Glucagon secretion
Insulin biosynthesis
β-cell proliferation
β-cell apoptosis
Glucose uptake
and stora
g
e
Cardiac function
Brain
Intestine
Liver
Adipose
tissue Muscle
Insulin
sensitivity
Stomach
Pancreas
Fig. 2 GLP-1 actions in peripheral tissues. The majority of the effects of GLP-1 are mediated by
direct interaction with GLP-1Rs on specific tissues. However, the actions of GLP-1 in liver, fat,
and muscle most likely occur through indirect mechanisms (adapted from Baggio and Drucker
2007)
188 B. Gallwitz
et al. 1996; Turton et al. 1996). Peripheral administration of GLP-1 dose-depen-
dently enhances satiety and reduces food intake in normal subjects (Flint et al. 1998;
Verdich et al. 2001), obese subjects (N
aslund et al. 1999), and type 2 diabetic
patients (Gutzwiller et al. 1999; Zander et al. 2002). The mechanisms by which
GLP-1 exerts its effects are not completely clear yet. Interactions with neurons in the
gastrointestinal tract or possibly direct access to the brain through as observed in rats
(Ørskov et al. 1996) are possible and discussed in this chapter as well as other novel
hypothesis based on the finding that GLP-1 is also expressed in taste cells. Finally,
the role of GLP-1 receptor agonists as possible treatment options in obesity is
discussed as well as the role of GLP-1 in the weight losing and metabolic effects
after various methods of bariatric surgery.
3 Central Effects of GLP-1
3.1 Direct Dentral Effects of GLP-1 in the Hypothalamus
As early as in 1988, it was shown that GLP-1 is also synthesized in the CNS in the
caudal part of the nucleus of the solitary tract (Jin et al. 1988) in addition to its
peripheral synthesis in the intestinal L cell. Receptors for GLP-1 are expressed
throughout the brain widely, with highest levels in the paraventricular nucleus
(Larsen et al. 1997b; Van Dijk et al. 1996; Turton et al. 1996). The presence of
both, the peptide GLP-1 and the GLP-1 receptor, in the CNS points toward
important physiological actions of GLP-1 in the CNS in addition to its actions on
the peripheral system.
The first report that GLP-1 exerted effects in the central nervous system (CNS)
came from Turton and colleagues. This group gave intracerebroventricular (icv)
injections of GLP-1 to rats. The injections reduced the food intake of the animals
compared to saline-treated control rats (Turton et al. 1996). The group also
demonstrated the presence of GLP-1-containing neurons in the rat brain in hypo-
thalamic areas that are known to be responsible for regulating satiety and food
intake.
Since then, there has been a great interest in understanding the role of GLP-1 in
the regulation of food intake and satiety. In the rat, other groups confirmed the
findings of Turton by using either GLP-1 or exendin-4, a naturally occurring GLP-1
RA (Meeran et al. 1999; Turton et al. 1996; Tang-Christensen et al. 1996). In
humans, subcutaneous or intravenous application of GLP-1 also reduced hunger
and food intake, prandial injections in obese subjects led to a reduction in food
intake (N
aslund et al. 2004). Blocking GLP-1 action in the CNS by using the GLP-1
receptor antagonist exendin(9–39) increased food intake in rats that had had icv
injections with exendin(9–39), and additionally facilitated weight gain in these
animals after long-term administration (Turton et al. 1996).
Anorexigenic Effects of GLP-1 and Its Analogues 189
Icv injections of GLP-1 receptor agonists inhibit food intake in rodents (Turton
et al. 1996; Meeran et al. 1999). Repeated icv administration of GLP-1 in rats leads
to weight loss (see Fig. 3). Conversely, icv injection of the GLP-1 receptor antago-
nist exendin(9–39) promoted weight gain in the animals, and exendin(9–39)
administered simultaneously with the central orexigenic agent neuropeptide Y
(NPY) resulted in an increased food intake and weight gain compared with that
observed with neuropeptide Y alone (Meeran et al. 1999; Abu-Hamdah et al. 2009).
In this respect, it is important that the intestinal L cells cosecrete GLP-1 and peptide
YY (PYY). Immunohistological studies demonstrated that these peptides are
Fig. 3 Effect of multiple icv injections of GLP-1 on food intake and body weight. Food intake (a)
and body weight (b) after daily icv injection of GLP-1 or saline as control are shown. The hatched
bars and filled circles represent animals given 3 nmol GLP-1, and the open bars and open circles
represent control animals that received saline. Food intake and body weight were significantly
decreased through the study period in animals receiving GLP-1 (P<0.05 for both groups). Body
weight was similar in noninjected controls (open triangles) and those given icv normal saline
(from Meeran et al. 1999 with permission)
190 B. Gallwitz
colocalized and coreleased from these cells. The truncated PYY(3–36), comprising
the major circulating form of PYY, has been reported to be a potent anorexigenic
agent in rats as well as in man (Batterham et al. 2002,2003). Although these effects
have not been reproduced by others (Tsch
op et al. 2004; Boggiano et al. 2005),
it was suggested that the corelease of GLP-1 and PYY has an important roles in the
mediation of satiety (Abu-Hamdah et al. 2009).
It has further been demonstrated that GLP-1 may play a role in the regulation of
the hypothalamic pituitary axis via effects on CRH, LH, TSH, oxytocin, and
vasopressin secretion (Beak et al. 1996,1998). The available evidence suggests
that taste and/or food aversion induced by GLP-1 is mediated by different CNS
pathways (Kinzig et al. 2002; Seeley et al. 2000; Tang-Christensen et al. 1996).
The GLP-1 receptor is widely expressed in the rodent brain in the hypothalamic
arcuate nucleus (ARC), the paraventricular nucleus (PVN), and supraoptic nuclei
(Shughrue et al. 1996). Furthermore, GLP-1 neurons of the solitary tract predomi-
nantly project into the PVN (Larsen et al. 1997a). The food-intake decreasing effect
of GLP-1 in rodents is associated with an increase in c-Fos expression in the ARC
(Larsen et al. 1997b). In rats treated with monosodium glutamate, the inhibitory
effect of GLP-1 on hunger-induced feeding was completely abolished (Tang-
Christensen et al. 1998). Additionally, GLP-1 stimulates the electrical activity of
proopiomelanocortin (POMC) neurons via the protein kinase A pathway and
a consecutive increase of L-type calcium currents (Ma et al. 2007). These findings
suggest that the hypothalamic ARC may play a role in GLP-1-induced inhibition of
food intake.
The regulation of energy balance involves the interaction of numerous regu-
latory peptides and neurotransmitters in the hypothalamus. The orexigenic peptides
neuropeptide Y (NPY) and agouti-related peptide (AgRP) as well as the anorexi-
genic peptides POMC and cocaine- and amphetamine-related transcripts (CART)
are produced in the ARC of the hypothalamus and play an important role in the
regulation of energy intake and energy expenditure (Schwartz et al. 2000). The
hypothalamic neurons are sensitive to satiety and hunger signals such as cholecys-
tokinin (CCK) and ghrelin. These hypothalamic neurons are also sensitive to
signals of long-term energy stores such as insulin and leptin (Schwartz et al.
2000). However, the effect of GLP-1 on the expression of these hypothalamic
mediators is not completely known. NPY and AgRP expression as measured by
mRNA concentrations is increased during fasting. These hunger-induced increases
are significantly diminished by icv injections of GLP-1. Conversely, the
expressions of POMC and CART are decreased during fasting, and again, these
changes are attenuated by the icv injection of GLP-1. Additionally, when deter-
mining mRNA concentrations of AMP-activated kinase (AMPK), a stimulation of
hypothalamic AMPKa2 could be observed during fasting that was also inhibited by
GLP-1 application. In summary, these findings suggest that the decreased food
intake mediated by GLP-1 is facilitated by the above mentioned changes of
orexigenic and anorexigenic hypothalamic neurotransmitter expression changes
(Seo et al. 2008).
Anorexigenic Effects of GLP-1 and Its Analogues 191
Another regulatory system most likely to be involved is a GLP-1-mediated
activation of the hypothalamo–pituitary–adrenocortical (HPA) axis (Larsen et al.
1997a; Larsen et al. 1997b). This mechanism primarily involves the stimulation of
corticotropin-releasing factor (CRF) neurons by GLP-1, and this activation may
also be responsible for the inhibition of feeding behavior. There seem to be species
differences regarding this regulatory mechanism: in rats, plasma concentrations of
corticosterone were rapidly increased after central administration of GLP-1,
whereas icv injections of GLP-1 did not alter plasma corticosterone concentrations
in the neonatal chick (Furuse et al. 1997).
In neonatal chicks, a noradrenergic mechanism was shown to contribute to the
anorexigenic effect of GLP-1 (Bungo et al. 2001a). Icv administration of norepi-
nephrine (NE) suppressed food intake and produced narcolepsy comparable to the
effect of GLP-1 in chicks. Although dopamine (DA) did not alter food intake, the
coadministration of inhibitors of dopamine-b-hydroxylase (DBH) or fusaric acid
(FA) attenuated the suppressive effect of GLP-1 on feeding behavior. Thus, it is
suggested that there may be interactive relationships between GLP-1 and noradren-
ergic regulatory systems in chicks (Bungo et al. 2001b), and that additionally, there
may be species differences in GLP-1-mediated appetite control.
3.2 GLP-1 in Taste Cells
GLP-1 and PYY are secreted not only from L cells in the small intestine but also
from mammalian taste cells. Both cell types, human duodenal L cells and taste cells
of the tongue, express the sweet taste receptor G protein gustducin that may
additionally be involved in the regulation of GLP-1 release (Jang et al. 2007).
In many L cells, GLP-1, gustducin, and PYY are colocalized (Jang et al. 2007).
Furthermore, GLP-1 is produced in two subsets of mammalian taste cells (type
2 and type 3).The corresponding GLP-1 receptors are present on adjacent
intragemmal afferent nerve fibers (Shin et al. 2008). It is therefore hypothesized
that GLP-1 (and PYY) activates anorexigenic CNS events prior to stimulating islet
hormones (Egan and Margolskee 2008).
3.3 Central Effects of GLP-1 Influencing Gastrointestinal
Functions
It has also been demonstrated already in 1997 that icv injections of GLP-1 cause
a retardation of liquid gastric emptying (Imery
uz et al. 1997). Nakade and his group
showed that the peripheral sympathetic nervous system and the central CRF
receptors are involved in the central GLP-1-mediated delay of solid gastric empty-
ing in rats (Nakade et al. 2006).
192 B. Gallwitz
4 Peripheral Effects of GLP-1 Affecting Satiety
GLP-1 exerts potent and important inhibitory effects on gastric emptying and
gastric acid secretion. It is primarily responsible for the “ileal break,” a tightly
regulated process under neural and hormonal control that regulates the passage of
nutrients through the digestive tract. GLP-1 enhances satiety and reduces food
intake (Pitomobo 2008). GLP-1 inhibits these proximal events of the gastrointesti-
nal tract in a negative feedback manner (Ahren 2004). Nauck and colleagues were
able to inhibit gastric emptying after a liquid meal by icv administration of GLP-1
in healthy, normoglycemic volunteers (Nauck et al. 1997). The observed effect of
GLP-1 on gastric emptying was dose dependent and highly significant with physio-
logical GLP-1 plasma concentrations (Nauck et al. 1997; Meier et al. 2002,2003a)
(see Fig. 4). Another study in healthy volunteers investigated the effect of two
different doses of GLP-1 (0.125-nmol/kg or 0.25-nmol/kg body weight)
administered subcutaneously 5 min prior to a mixed test meal (Schirra et al.
1997). The pattern of gastric emptying of the mixed meal as well as pancreatic
secretion, antroduodenal motility, and the glycemic response and the release of
insulin, C-peptide, and glucagon were quantified. The lag period or the time to
reach maximal velocity of gastric emptying was dose-dependently prolonged in
response to the subcutaneous application of GLP-1. However, the maximal empty-
ing velocity, the total emptying rate, and the exponential emptying rate were
unaltered (Schirra et al. 1997). The subcutaneous infusion of GLP-1 resulted in
a dose-dependent inhibition of antral and duodenal motility, and both doses of
GLP-1 led to coordinated antroduodenal contractions. GLP-1 initially reduced
and then transiently stimulated the secretion of pancreatic enzymes. Both doses
of GLP-1 delayed the postprandial insulin peak and enhanced total insulin release.
The postprandial response of pancreatic polypeptide and glucagon was diminished
(Schirra et al. 1997).
In another study, the same group investigated the antropyloroduodenal motility
in humans and the actions of endogenously released GLP-1 on endocrine pancreas
secretion (Schirra et al. 2006). In this study, the GLP-1 receptor antagonist exendin
(9–39) was used to test whether GLP-1 acts as an incretin and/or as an enterogas-
trone in humans. The endogenously secreted GLP-1 significantly enhanced post-
prandial insulin secretion and suppressed the secretion of glucagon (Schirra et al.
2006). During the fasting and postprandial state, antroduodenal motility was
inhibited by GLP-1, which qualifies GLP-1 as an enterogastrone. The stimulation
of pyloric motility that is induced by intestinal glucose was mediated by GLP-1.
The presence of nutrients in the small intestine stimulates the L cells to release
GLP-1 into the circulation. The rise in GLP-1 concentrations not only stimulates the
beta cells to produce insulin but also slows gastric emptying and may lead to
a decrease in appetite and a sensation of fullness (N
aslund et al. 1999; Flint et al.
2001; Meier et al. 2003b; Silvestre et al. 2003; Ling et al. 2001; Nagai et al. 2004).
The mechanisms by which GLP-1 inhibits gastric emptying appear to be com-
plex and to involve communication with the central and peripheral nervous systems
Anorexigenic Effects of GLP-1 and Its Analogues 193
(D’Alessio 2008; Drucker 2006). Gastric distension increases the expression of
c-Fos in brain stem neurons that produce GLP-1 (Vrang et al. 2003). Furthermore,
icv administration of GLP-1 resulted in reduction of food intake (Kinzig et al.
2002), which is accompanied with increased expression of c-Fos in the brain stem
of the rat (Larsen et al. 1997b; Dakin et al. 2004). The denervation of afferent vagal
060 120 180 240
-5
15
35
55
75
95
1.2 pmol .kg
-1
.min
0.8 pmol.kg
-1
.min
0.4 pmol.kg
-1
.min
Placebo
A: p < 0.0001
B: p < 0.0001
AB: p < 0.0001
0.0
0.5
1.0
1.5
2.0
2.5
3.0
3.5
4.0
0.0 0.4 0.8 1.2
p < 0.0001
*
*
*
Gatric content
[% of initial value]
coefficient
Gatric emptying
time [min]
GLP-1 dose [pmolkg-1min]
Fig. 4 Time and dose dependency of GLP-1 on gastric emptying in humans. Panel A: Time
pattern of gastric emptying of a solid meal (250 kcal) during icv administration of different doses
of GLP-1 (0.4, 0.8, and 1.2 pmol/kg/min; filled symbols) or placebo (open symbols) in patients with
type 2 diabetes (n¼12). Gastric emptying was determined from the measurement of
13
CO
2
in
breath samples collected after the ingestion of the test meal labeled with [
13
C] octanoic acid using
infrared absorptiometry. Data are expressed as the mean + SE. Pvalues were calculated using
repeated measures ANOVA and denote: A, differences between the doses tested; B, differences
over time; and AB, differences due to the interaction of experiment and time. Panel B: Gastric
emptying coefficients. Data are expressed as the mean + SE. Asterisks indicate significant
differences (P<0.05) versus placebo. (from Meier et al. 2003a with permission)
194 B. Gallwitz
fibers abolishes the effects of GLP-1 on gastric emptying in the rat (Imery
uz et al.
1997). The stimulation to the CNS is most likely responsible for the reduction in
food intake, inhibition of gastric emptying, as well as inhibitory action on gastric
motor function (Kinzig et al. 2002; Imery
uz et al. 1997). These actions are most
likely mediated by increased action potential and calcium influx in neurons of the
nodose ganglion (Kakei et al. 2002).
Although small peptides such as GLP-1 and exendin-4 are capable of rapidly
crossing the blood–brain barrier and directly accessing the CNS, GLP-1 receptor
agonists with a larger molecule size and a higher molecular weight, such as
albumin-bound GLP-1, that do not cross the blood–brain barrier, are still capable
of inhibiting gastric emptying and food intake (Baggio et al. 2004). These findings
underline the importance of ascending vagal afferents for the GLP-1 receptor-
dependent control of gastrointestinal motility. Interestingly, studies by Meier and
his group (Meier et al. 2005) showed that antagonizing the delaying effects of GLP-
1 on gastric emptying by a prokinetic agent such as erythromycin resulted in an
augmentation of the insulin secretory response after meal ingestion. GLP-1
receptors are also directly expressed in the stomach on gastric parietal cells,
where GLP-1 may directly regulate gastric acid secretion (Schmidtler et al.
1994). However, the effects of GLP-1 on gastric acid secretion were found to be
absent in vagotomized human subjects (Wettergren et al. 1997). Hence, consider-
able evidence supports the importance of vagal innervation for GLP-1 regulation of
gastric secretion and motility.
These observed effects of delayed gastric emptying have been generally
demonstrated with at least physiological or, as in most studies, supraphysiological
doses of exogenously administered GLP-1 (Delgado-Aros et al. 2002; Schirra et al.
1996). Therefore, it remains unclear whether endogenously released GLP-1 has
a significant effect on gastric emptying. Studies in healthy baboons have shown
that with intragastric infusion of glucose and D-xylose (a marker for rate of
emptying of glucose from stomach), plasma levels of D-xylose were similar when
the effects of GLP-1 were blocked with exendin(9–36) amide or with a specific
monoclonal antibody to GLP-1 (D’Alessio et al. 1996; D’Alessio 2008). These
findings suggest that gastric emptying is not increased when the effects of GLP-1
are blocked, at least in the baboon. The use of a DPP-4 inhibitor, which increases
plasma concentrations of endogenous GLP-1, might be expected to delay gastric
emptying, but a study in patients with type 2 diabetes and DPP-4 inhibitor treatment
did not reveal any changes in the gastric emptying of a solid meal (Vella et al.
2007). Most recently, an iv-oral hyperglycemic clamp study in humans was
reported during which 75-g glucose-containing D-xylose was ingested. During the
entire clamp, plasma glucose levels were held at a steady level despite the ingestion
of glucose. Two studies were conducted, with blockade of GLP-1 receptor in one.
The rate of appearance of ingested D-xylose was not different between the two
studies, indicating that endogenously released GLP-1 has at best only a modest
effect on gastric emptying (Salehi et al. 2008).
In a variety of endocrine regulatory systems, a negative feedback mechanism
regulates the secretion of the hormone, e.g., the reproductive hormone regulation by
Anorexigenic Effects of GLP-1 and Its Analogues 195
the hypothalamus. Exogenous infusion of a hormone may also exert negative
feedback regulation of the endogenously released hormone. An example of this is
the documented suppression of C-peptide plasma concentrations when insulin is
infused (Elahi et al. 1982). In this context, it is presently still not known whether
exogenously administered GLP-1 really has a significant impact on the regulation
of endogenously released GLP-1.
5 Effects of GLP-1 Receptor Agonists on Body Weight
in Humans
Native GLP-1 cannot be used in a feasible way for treatment of type 2 diabetes or
obesity due to its very short biological half-life of 1–2 min. For this reason, long-
acting GLP-1 receptor agonists were developed. In 1992 exendin-4, a reptilian
peptide isolated from the lizard Heloderma suspectum was identified as a long-
acting GLP-1 receptor agonist (Raufman et al. 1992;G
oke et al. 1993). Exendin-4
has a 53% sequence homology with GLP-1 and has a biological half-life of
approximately 3.5 h. The synthetic form of exendin-4, exenatide, was the first
GLP-1 receptor agonist that was approved for type 2 diabetes therapy in patients
not sufficiently controlled on a therapy with metformin or sulfonylureas or
a combination of both (Klonoff et al. 2008). Exenatide (Byetta
®
, Eli Lilly
Pharmaceuticals, Indianapolis, USA and Amylin Pharmaceuticals, San Diego,
USA) is given subcutaneously twice daily.
Liraglutide (Victoza
®
, Novo Nordisk Pharmaceuticals, Copenhagen, Denmark)
was the first human GLP-1 analogue that was developed for once daily subcutane-
ous application. Liraglutide has a half-life of 13.5 h. It has a fatty acid side chain
that allows heptamer formation of the molecule that prevents direct DPP-4 action as
well as fast dissociation from the subcutaneous tissue into the circulation. Further-
more, the fatty acid side chain allows albumin binding that further protracts
degradation and prolongs biological availability (Garber et al. 2009; Madsbad
et al. 2011).
Presently, even longer-acting GLP-1 receptor analogues are being developed in
order to reduce the frequency of injections, to reduce fasting glucose more effi-
ciently, and to reduce the gastrointestinal side effects (mainly fullness and nausea)
that are associated with the fluctuation of GLP-1 receptor agonist plasma
concentrations. Compounds with an exendin-4/exenatide backbone are exenatide
once weekly (Bydureon
®
, Eli Lilly Pharmaceuticals, Indianapolis, USA and
Amylin Pharmaceuticals, San Diego, USA) that has just received a positive opinion
by the regulatory agencies in the United States and Europe and is expected to be
marketed later in 2011 (Madsbad et al. 2011).
Lixisenatide (Sanofi-Aventis Pharmaceuticals, Paris, France) is an exendin-4
analogue for once daily application, presently in phase III of the clinical study
program (Christensen et al. 2011; Madsbad et al. 2011).
196 B. Gallwitz
Albiglutide, an albumin-bound-human GLP-1 fusion protein (GlaxoSmithKline
Pharmaceuticals, London, UK) is feasible for once weekly dosing and is also
presently in phase III of the clinical study program (Madsbad et al. 2011;St
Onge and Miller 2011).
Another human GLP-1 receptor agonist in earlier development for once weekly
dosing is LY2189265 (Dulaglutide, Eli Lilly Pharmaceuticals, Indianapolis, USA)
(Glaesner et al. 2010; Madsbad et al. 2011). Table 1gives an overview on the GLP-1
receptor agonists available and in development and their respective characteristics.
Chronic peripheral administration of GLP-1 RA agonists (exendin-4/exenatide
and liraglutide) has consistently been associated with reductions in food intake and
weight loss in animal studies and in humans (Szayna et al. 2000; Young et al. 1999;
Schnabel et al. 2006; Garber et al. 2009). Also, weight loss was documented in
a pivotal study in which a continuous 6-week infusion of GLP-1 was given to obese
type 2 diabetic patients (Zander et al. 2002). Conversely, a continuous subcutaneous
administration study of a lower dose of GLP-1 (1.5 pmolkg
1
min
1
) for 12 weeks
Table 1 GLP-1 receptor agonists and their characteristics. The available GLP-1 receptor
agonists as well as substances advanced in clinical development are shown
Substance Chemical backbone Dosing interval Approval/
developmental status
Exenatide (Byetta
®
, Eli
Lilly and Amylin)
Exendin-4 Twice daily Approved 2005
Liraglutide (Victoza
®
,
NovoNordisk)
Human GLP-1 with two
amino acid exchanges
and a c-16 fatty acid side
chain
Once daily Approved 2009
Exenatide QW
(Bydureon
®
, Eli
Lilly and Amylin)
Exendin-4, incorporated into
a matrix of poly(D,L-
lactide-co-glycolide)
(PLG) to prolong action
Once weekly Approved 2011
Lixisenatide
(Sanofi-Aventis)
Exendin-4, 44 amino acids
with C-terminal
extension (C-terminal
with six Lys residues and
one Pro deleted)
Once daily Phase III clinical
investigation
Albiglutide (Syncrea
®
,
GlaxoSmithKline)
Human GLP-1 receptor
agonist consisting of two
copies of a 30-amino acid
sequence of a dipeptyl
peptidase-4-resistant
human GLP-1 (as a
tandem repeat) coupled
to serum human albumin
Once weekly
(planned)
Phase III clinical
investigation
LY2189265
(Dulaglutide
®
,
Eli Lilly)
DPP-4-protected GLP-1
analogue is fused to a
modified
immunoglobulin G4
(IgG4) Fc fragment
Once weekly
(planned)
Phase III clinical
investigation
Anorexigenic Effects of GLP-1 and Its Analogues 197
produced no significant weight loss (Meneilly et al. 2003). This study most likely
demonstrates that weight loss with exogenous GLP-1 administration is only possi-
ble, when much larger doses are given.
In most phase 3 studies with exenatide and liraglutide, the weight loss was in the
range of 2–3 kg after 26 weeks of treatment compared with placebo, and greatest
when added to metformin (Madsbad 2009). In studies with a longer duration, a
plateau of the weight loss is seen in the same range (Garber et al. 2009,2011).
Presently, long-acting GLP-1 receptor agonists that require a single weekly dose
only, or other long-range dosing regimens, are in clinical development (Madsbad
et al. 2011). Summarizing the known effects on body weight, there does not seem to
be a difference between the short-acting, established GLP-1 RA and the novel long-
acting ones. The weight loss in a study comparing the novel, once weekly GLP-1
RA albiglutide with exenatide did not reveal a significant difference in the body
weight development. The weight loss amounted from 1.1 to 1.7 kg in the
albiglutide groups compared with 0.7 kg in the placebo group and 2.4 kg in
the exenatide group (Rosenstock et al. 2009).
Exenatide once weekly (exenatide QW) is a new dosage form of the active drug
exenatide. Exenatide QW’s microsphere technology enables very slow release due
to the use of a slowly biodegradable polymer as the exenatide carrier. This changes
both the effect and adverse effect profiles versus the short-acting receptor stimula-
tion produced by the established, unretarded exenatide for twice daily application.
Long-term stimulation of GLP-1 receptors results in superior lowering of fasting
blood glucose levels and HbA1c (Kim et al. 2007). In subjects on first-line treatment
with metformin, exenatide QW produced a superior HbA1c reduction. Because of
the weaker inhibition of gastric emptying, gastrointestinal side effects are reduced
by about one-third (20% with exenatide QW versus 35% with exenatide)
(Linnebjerg et al. 2008; Wang et al. 2008). In addition, a relevant loss in mean
weight (ranging from 2.3 kg to 3.7 kg) was seen in all studies (Drucker et al. 2008;
Buse et al. 2010; Bergenstal et al. 2010; Diamant et al. 2010; Kim et al. 2007).
Another GLP-1 RA in development is CJC-1134-PC (ConjuChem, Montreal,
Quebec, Canada), which consists of an exendin-4 molecule covalently linked to
human recombinant albumin. Its half-life of approximately 8 days corresponds to
that of circulating albumin (Thibaudeau et al. 2006; Baggio et al. 2008). At present,
it is unclear whether the modest effect on body weight is explained by a reduced
efficacy in engaging the central nervous system regions regulating appetite and
body weight, because large proteins like albumin are not expected to cross the
blood–brain barrier (Chuang et al. 2002). Alternatively, the compound can still
regulate feeding and body weight via the vagus nerve (Abbott et al. 2005; Imery
uz
et al. 1997).
With respect to weight control, no clinically significant differences seem to exist
within the entire group of GLP-1 receptor agonists, although it remains possible
that the CJC-1134-PC is less effective (Chuang et al. 2002; Wang et al. 2009).
With the short-acting GLP-1 RA liraglutide, a clinical study was performed
in obese, nondiabetic subjects to investigate the efficacy and safety as a weight-
loss-promoting drug. In a placebo-controlled 20-week trial, with an open-label
198 B. Gallwitz
orlistat (120 mg t.i.d.) comparator arm, study participants were assigned to arms of
four liraglutide doses (1.2 mg/d, 1.8 mg/d, 2.4 mg/d, or 3.0 mg/d) or to placebo.
Participants on liraglutide lost significantly more weight than did those on placebo
and orlistat. The mean weight loss caused by liraglutide was dose dependent and
amounted to 4.8 kg, 5.5 kg, 6.3 kg, and 7.2 kg for the respective liraglutide doses
(1.2 mg/d, 1.8 mg/d, 2.4 mg/d, or 3.0 mg/d) and was 2.1 kg greater than that in the
placebo group. The weight loss with placebo amounted to 2.8 kg and with orlistat
4.1 kg (Astrup et al. 2009) (see Fig. 5).
6 GLP-1 and Bariatric Surgery
The mechanisms involved in weight loss and metabolic improvements after bariat-
ric surgery are dependent on the type of surgery performed and are not yet
completely understood. They are presently under thorough investigation. Changes
in the size of the gastric pouch and the length and parts of the intestinal surfaces
after bariatric surgery determine the contact of food with enteroendocrine cells and
consequently also the gut hormonal response. Each type of bariatric surgery has
Fig. 5 Dose dependent effects of the GLP-1 RA liraglutide on body weight in obese subjects. Five
hundred sixty-four individuals (age range 18–65 years, BMI 30–40 kg/m) were randomized to one
of four liraglutide doses (1·2 mg, 1·8 mg, 2·4 mg, or 3·0 mg, n¼90–95) or to placebo (n¼98)
administered once a day subcutaneously, or orlistat (120 mg, n¼95) three times a day orally.
Weight change was analyzed by intention to treat. Data are mean (95% CI) (ANCOVA estimate)
for the intention-to-treat population with the last observation carried forward (from Astrup et al.
2009 with permission)
Anorexigenic Effects of GLP-1 and Its Analogues 199
a different effect on hormonal secretion and thus may play a significant role in the
mechanism of weight loss (Vetter et al. 2009).
Rubino and colleagues evaluated the early effects of Roux-Y gastric bypass
(RYGB) on glucose, insulin, glucagon, insulin-like growth factor-1, GIP, GLP-1,
CCK, adrenocorticotropic hormone (ACTH), corticosterone, and neuropeptide Y
(Rubino et al. 2004; Thomas and Schauer 2010). RYGB led to a decrease in BMI
paralleled by a significant decrease in glucose, insulin, leptin, and an increase in
ACTH levels 3 weeks after surgery. The other hormones, especially GLP-1, did not
change significantly. However, of the six diabetic patients in the study, all had
normal glucose and insulin levels after surgery and did not require any diabetic
medications.
It has been observed that the initial weight loss observed with either laparoscopic
sleeve gastrectomy (LSG) as a restrictive surgical method or RYGB as a diversion
method of surgery leads to similar results (Thomas and Schauer 2010). There are
metabolic differences however, demonstrating that patients with RYGB show
a rapid normalization of fasting glucose and an improvement of insulin clearance
and sensitivity, but these changes do not occur in patients with LSG. Testing the
different cohorts of patients after RYGB or LSG with a mixed meal tolerance test,
the dramatic increase in insulin secretion and an increase in GLP-1 are only
observed in the RYGB group (Thomas and Schauer 2010).
In obese patients with coexisting type 2 diabetes, both types of bariatric
procedures were associated with an improvement of hyperglycemia. RYGB was
associated with an insulinotropic response with an oral mixed meal but not with
intravenous glucose, consistent with an incretin effect. These data suggest a differ-
ent effect of the two procedures on pancreatic beta-cell function. The improvement
may be due to insulin sensitivity and therefore a reduced insulin response following
gastric restriction only (Thomas and Schauer 2010). It is still not known, however,
whether the portion that is bypassed causes this effect or whether this effect is due
to nutrients that rapidly reach the distal ileum and release insulinotropic and beta-
cell-enhancing hormones (Kashyap et al. 2010). The limitations of the study are the
small sample size and the sample being a nonrandomized convenience sample.
To evaluate the potential role of the exclusion of the proximal small intestine in
the improvement of diabetes mellitus after gastric bypass surgery, Peterli and his
group conducted a prospective, randomized, controlled trial comparing RYGB and
LSG. Patients were evaluated 1 week and 3 months after surgery before and after
a standard test meal. At 3 months, body weight and BMI decreased significantly and
comparably in both groups with markedly increased postprandial plasma insulin
and GLP-1 levels (Dirksen et al. 2010). RYGB patients had increased insulin
responses as early as 1 week after the surgery; however, no significant differences
were seen at 3 months in insulin or GLP-1 levels. Thus, both procedures improved
glucose homeostasis, insulin, and GLP-1 and PYY levels (Peterli et al. 2009).
Two different hypotheses have been proposed to explain these conflicting data.
One offered is the “hindgut explanation,” suggesting that the rapid transit of
nutrients to the distal intestine improves glucose metabolism by stimulating secre-
tion of GLP-1 and other appetite-suppressing gut peptides such as PYY. Insulin
200 B. Gallwitz
secretion is increased and glucose tolerance improves, affecting body weight and
food intake (Cummings et al. 2007; Patrita et al. 2007). On the other hand, Rubino
and his group proposed the “foregut hypothesis.” They propose that there is a yet-
unknown factor that promotes insulin resistance and type 2 diabetes. When food
bypasses the duodenum and proximal jejunum after bariatric surgery, this so-called
anti-incretin or decretin factor is inhibited, and thus insulin resistance is decreased
and glucose tolerance improves. Other factors may help to explain the differences
seen among the various types of procedures (Vetter et al. 2009; Cummings et al.
2008). Likewise, because GLP-1, PYY, and GIP are secreted by the small intestine,
differences in the length of the roux limb may contribute to the secretion of these
gut hormones and thus the results seen. Finally, as this is a relatively new, evolving
field of research, there are most likely unknown factors to be considered (Thomas
and Schauer 2010).
References
Abbott CR, Monteiro M, Small CJ, Sajedi A, Smith KL, Parkinson JR, Ghatei MA, Bloom SR
(2005) The inhibitory effects of peripheral administration of peptide YY(3–36) and glucagon-
like peptide-1 on food intake are attenuated by ablation of the vagal-brainstem-hypothalamic
pathway. Brain Res 1044:127–131
Abu-Hamdah R, Rabiee A, Meneilly GS, Shannon RP, Andersen DK, Elahi D (2009) Clinical
review: the extrapancreatic effects of glucagon-like peptide-1 and related peptides. J Clin
Endocrinol Metab 94:1843–1852
Ahren B (2004) GLP-1 and extra-islet effects. Horm Metab Res 36:842–845
Astrup A, R
ossner S, Van Gaal L, Rissanen A, Niskanen L, Al Hakim M, Madsen J, Rasmussen
MF, Lean ME, NN8022-1807 Study Group (2009) Effects of liraglutide in the treatment of
obesity: a randomised, double-blind, placebo-controlled study. Lancet 374:1606–1616
Baggio LL, Drucker DJ (2007) Biology of incretins: GLP-1 and GIP. Gastroenterology
132:2131–2157
Baggio LL, Huang Q, Brown TJ, Drucker DJ (2004) A recombinant human glucagon-like peptide
(GLP)-1-albumin protein (albugon) mimics peptidergic activation of GLP-1R-dependent
pathways coupled with satiety, gastrointestinal motility, and glucose homeostasis. Diabetes
53:2492–2500
Baggio LL, Huang Q, Cao X, Drucker DJ (2008) An albumin-exendin-4 conjugate engages central
and peripheral circuits regulating murine energy and glucose homeostasis. Gastroenterology
134:1137–1147
Batterham RL, Cowley MA, Small CJ, Herzog H, Cohen MA, Dakin CL, Wren AM, Brynes AE,
Low MJ, Ghatei MA, Cone RD, Bloom SR (2002) Guthormone PYY(3–36) physiologically
inhibits food intake. Nature 418:650–654
Batterham RL, Cohen MA, Ellis SM, Le Roux CW, Withers DJ, Frost GS, Ghatei MA, Bloom SR
(2003) Inhibition of food intake in obese subjects by peptide YY3-36. N Engl J Med
349:941–948
Beak SA, Small CJ, Ilovaiskaia I, Hurley JD, Ghatei MA, Bloom SR, Smith DM (1996) Glucagon-
like peptide-1 (GLP-1) releases thyrotropin (TSH): characterization of binding sites for GLP-1
on a-TSH cells. Endocrinology 137:4130–4138
Beak SA, Heath MM, Small CJ, Morgan DG, Ghatei MA, Taylor AD, Buckingham JC, Bloom SR,
Smith DM (1998) Glucagon-like peptide-1 stimulates luteinizing hormone-releasing hormone
secretion in a rodent hypothalamic neuronal cell line. J Clin Invest 101:1334–1341
Anorexigenic Effects of GLP-1 and Its Analogues 201
Bell GI, Sanchez-Pescador R, Laybourn PJ, Najarian RC (1983) Exon duplication and divergence
in the human preproglucagon gene. Nature 304:368–371
Bergenstal RM, Wysham C, Macconell L, Malloy J, Walsh B, Yan P, Wilhelm K, Malone J, Porter
LE (2010) Efficacy and safety of exenatide once weekly versus sitagliptin or pioglitazone as an
adjunct to metformin for treatment of type 2 diabetes (DURATION-2): a randomised trial.
Lancet 376:431–439
Boggiano MM, Chandler PC, Oswald KD, Rodgers RJ, Blundell JE, Ishii Y, Beattie AH, Holch P,
Allison DB, Schindler M, Arndt K, Rudolf K, Mark M, Schoelch C, Joost HG, Klaus S, Thone-
Reineke C, Benoit SC, Seeley RJ, Beck-Sickinger AG, Koglin N, Raun K, Madsen K, Wulff
BS, Stidsen CE, Birringer M, Kreuzer OJ, Deng XY, Whitcomb DC, Halem H, Taylor J, Dong
J, Datta R, Culler M, Ortmann S, Castaneda TR, Tschop M (2005) PYY3-36 as an anti-obesity
drug target. Obesity Rev 6:307–322
Brubaker PL, Drucker DJ (2004) Minireview: glucagon-like peptides regulate cell proliferation
and apoptosis in the pancreas, gut, and central nervous system. Endocrinology 145:2653–2659
Bungo T, Ando R, Kawakami S-I, Ohgushi A, Furuse M (2001a) The role of central catechol-
aminergic systems in regulation of food intake of chicks. J Poult Sci 38:35–40
Bungo T, Kawakami S-I, Ohgushi A, Sashihara K, Saito N, Sugahara K, Hasegawa S, Denbow
DM, Furuse M (2001b) Intracerebroventricular injection of fusaric acid attenuates the anorexia
by glucagon-like peptide-1 in the neonatal chick. Pharmacol Biochem Behav 70:251–255
Buse JB, Drucker DJ, Taylor KL, Kim T, Walsh B, Hu H, Wilhelm K, Trautmann M, Shen LZ,
Porter LE (2010) DURATION-1: exenatide once weekly produces sustained glycemic control
and weight loss over 52 weeks. Diabetes Care 33:1255–1261
Christensen M, Knop FK, Vilsbøll T, Holst JJ (2011) Lixisenatide for type 2 diabetes mellitus.
Expert Opin Investig Drugs 20:549–557
Chuang VT, Kragh-Hansen U, Otagiri M (2002) Pharmaceutical strategies utilizing recombinant
human serum albumin. Pharm Res 19:569–577
Courreges JP, Vilsboll T, Zdravkovic M, Le-Thi T, Krarup T, Schmitz O, Verhoeven R, Buganova
I, Madsbad S (2008) Beneficial effects of once-daily liraglutide, a human glucagon-like
peptide-1 analogue, on cardiovascular risk biomarkers in patients with Type 2 diabetes. Diabet
Med 25:1129–1131
Creutzfeldt W (1979) The incretin concept today. Diabetologia 16:75–85
Cummings D, Overduin J, Foster-Schubert K, Carlson M (2007) Role of the bypassed proximal
intestine in the anti-diabetic effects of bariatric surgery. Surg Obes Relat Dis 3:109–115
Cummings D, Foster-Schubert K, Carlson M, Shannon M, Overduin J (2008) Possible hormonal
mechanisms mediating the effects of bariatric surgery. In: Pitomobo C, Jones K, Higa K, Pareja
J (eds) Obesity surgery: principles and practice. McGraw-Hill, New York, NY, pp 137–147
D’Alessio D (2008) Intestinal hormones and regulation of satiety: the case for CCK, GLP-1, PYY,
and Apo A-IV. JPEN J Parenter Enteral Nutr 32:567–568
D’Alessio DA, Vogel R, Prigeon R, Laschansky E, Koerker D, Eng J, Ensinck JW (1996)
Elimination of the action of glucagon-like peptide 1 causes an impairment of glucose tolerance
after nutrient ingestion by healthy baboons. J Clin Invest 97:133–138
Dakin CL, Small CJ, Batterham RL, Neary NM, Cohen MA, Patterson M, Ghatei MA, Bloom SR
(2004) Peripheral oxyntomodulin reduces food intake and body weight gain in rats. Endocri-
nology 145:2687–2695
Delgado-Aros S, Kim DY, Burton DD, Thomforde GM, Stephens D, Brinkmann BH, Vella A,
Camilleri M (2002) Effect of GLP-1 on gastric volume, emptying, maximum volume ingested,
and postprandial symptoms in humans. Am J Physiol Gastrointest Liver Physiol 282:
G424–G431
Diamant M, Van GL, Stranks S, Northrup J, Cao D, Taylor K, Trautmann M (2010) Once weekly
exenatide compared with insulin glargine titrated to target in patients with type 2 diabetes
(DURATION-3): an open-label randomised trial. Lancet 375:2234–2243
Dirksen C, Hansen DL, Madsbad S, Hvolris LE, Naver LS, Holst JJ, Worm D (2010) Postprandial
diabetic glucose tolerance is normalized by gastric bypass feeding as opposed to gastric
202 B. Gallwitz
feeding and is associated with exaggerated GLP-1 secretion: a case report. Diabetes Care
33:375–377
Drucker DJ (2006) The biology of incretin hormones. Cell Metab 3:153–165
Drucker DJ, Nauck MA (2006) The incretin system: glucagon-like peptide-1 receptor agonists and
dipeptidyl peptidase-4 inhibitors in type 2 diabetes. Lancet 368:1696–1705
Drucker DJ, Buse JB, Taylor K, Kendall DM, Trautmann M, Zhuang D, Porter L (2008) Exenatide
once weekly versus twice daily for the treatment of type 2 diabetes: a randomised, open-label,
non-inferiority study. Lancet 372:1240–1250
Egan JM, Margolskee RF (2008) Taste cells of the gut and gastrointestinal chemosensation.
Mol Interv 8:78–81
Elahi D, Nagulesparan M, Hershcopf RJ, Muller DC, Tobin JD, Blix PM, Rubenstein AH, Unger
RH, Andres R (1982) Feedback inhibition of insulin secretion by insulin: relation to the
hyperinsulinemia of obesity. N Engl J Med 306:1196–1202
Fehmann HC, Habener JF (1992) Insulinotropic hormone glucagon-like peptide-I(7–37) stimula-
tion of proinsulin gene expression and proinsulin biosynthesis in insulinoma beta TC-1 cells.
Endocrinology 130:159–166
Flint A, Raben A, Astrup A, Holst JJ (1998) Glucagon-like peptide 1 promotes satiety and
suppresses energy intake in humans. J Clin Invest 101:515–520
Flint A, Raben A, Ersbøll AK, Holst JJ, Astrup A (2001) The effect of physiological levels of
glucagon-like peptide-1 on appetite, gastric emptying, energy and substrate metabolism in
obesity. Int J Obes Relat Metab Disord 25:781–792
Furuse M, Matsumoto M, Saito N, Sugahara K, Hasegawa S (1997) The central corticotropin-
releasing factor and glucagon-like peptide-1 in food intake of the neonatal chick. Eur J
Pharmacol 339:211–213
Garber A, Henry R, Ratner R, Garcia-Hernandez PA, Rodriguez-Pattzi H, Olvera-Alvarez I, Hale
PM, Zdravkovic M, Bode B (2009) Liraglutide versus glimepiride monotherapy for type
2 diabetes (LEAD-3 Mono): a randomised,52-week, phase III, double-blind, parallel-treatment
trial. Lancet 373:473–481
Garber A, Henry RR, Ratner R, Hale P, Chang CT, Bode B, LEAD-3 (Mono) Study Group (2011)
Liraglutide, a once-daily human glucagon-like peptide 1 analogue, provides sustained
improvements in glycaemic control and weight for 2 years as monotherapy compared with
glimepiride in patients with type 2 diabetes. Diabetes Obes Metab 13:348–356
Glaesner W, Vick AM, Millican R, Ellis B, Tschang SH, Tian Y, Bokvist K, Brenner M, Koester
A, Porksen N, Etgen G, Bumol T (2010) Engineering and characterization of the long-acting
glucagon-like peptide-1 analogue LY2189265, an Fc fusion protein. Diabetes Metab Res Rev
26:287–296
G
oke R, Fehmann HC, Linn T, Schmidt H, Krause M, Eng J, G
oke B (1993) Exendin-4 is a high
potency agonist and truncated exendin-(9–39)-amide an antagonist at the glucagon-like peptide
1-(7–36)-amide receptor of insulin-secreting beta-cells. J Biol Chem 268:19650–19655
G
oke R, Larsen PJ, Mikkelsen JD, Sheikh SP (1995) Distribution of GLP-1 binding sites in the rat
brain: evidence that exendin-4 is a ligand of brain GLP-1 binding sites. Eur J Neurosci
7:2294–2300
Gromada J, Rorsman P, Dissing S, Wulff BS (1996) Stimulation of cloned human glucagon-like
peptide 1 receptor expressed in HEK 293 cells induces cAMP-dependent activation of calcium-
induced calcium release. FEBS Lett 373:182–186
Gutzwiller JP, Drewe J, Goke B, Schmidt H, Rohrer B, Lareida J, Beglinger C (1999) Glucagon-
like peptide-1 promotes satiety and reduces food intake in patients with diabetes mellitus type
2. Am J Physiol 276:R1541–R1544
Holst JJ (2007) The physiology of glucagon-like peptide 1. Physiol Rev 87:1409–1439
Holst JJ, Deacon CF, Vilsbøll T, Krarup T, Madsbad S (2008) Glucagon-like peptide-1, glucose
homeostasis and diabetes. Trends Mol Med 14:161–168
Imery
uz N, Yeden BC, Bozkurt A, Copkun T, Villanueva-Pen
˜acarrillo ML, Ulusoy NB (1997)
Glucagon-like peptide-1 inhibits gastric emptying via vagal afferent-mediated central
mechanisms. Am J Physiol 273:G920–G927
Anorexigenic Effects of GLP-1 and Its Analogues 203
Jang HJ, Kokrashvili Z, Theodorakis MJ, Carlson OD, Kim BJ, Zhou J, KimHH XuX, Chan SL,
Juhaszova M, Bernier M, Mosinger B, Margolskee RF, Egan JM (2007) Gut-expressed
gustducin and taste receptors regulate secretion of glucagon-like peptide-1. Proc Natl Acad
Sci USA 104:15069–15074
Jin SL, Han VK, Simmons JG, Towle AC, Lauder JM, Lund PK (1988) Distribution of
glucagonlike peptide I (GLP-I), glucagon, and glicentin in the rat brain: an immunocytochemi-
cal study. J Comp Neurol 271:519–532
Kakei M, Yada T, Nakagawa A, Nakabayashi H (2002) Glucagon-like peptide-1 evokes action
potentials and increases cytosolic Ca2+ in rat nodose ganglion neurons. Auton Neurosci
102:39–44
Kashyap SR, Daud S, Kelly KR, Gastaldelli A, Win H, Brethauer S, Kirwan JP, Schauer PR (2010)
Acute effects of gastric bypass versus gastric restrictive surgery on beta-cell function and
insulinotropic hormones in severely obese patients with type 2 diabetes. Int J Obes (Lond)
34:462–471
Kim D, Macconell L, Zhuang D, Kothare PA, Trautmann M, Fineman M, Taylor K (2007) Effects
of once-weekly dosing of a long-acting release formulation of exenatide on glucose control and
body weight in subjects with type 2 diabetes. Diabetes Care 30:1487–1493
Kinzig KP, D’Alessio DA, Seeley RJ (2002) The diverse roles of specific GLP-1Rs in the control
of food intake and the response to visceral illness. J Neurosci 22:10470–10476
Klonoff DC, Buse JB, Nielsen LL, Guan X, Bowlus CL, Holcombe JH, Wintle ME, Maggs DG
(2008) Exenatide effects on diabetes, obesity, cardiovascular risk factors and hepatic
biomarkers in patients with type 2 diabetes treated for at least 3 years. Curr Med Res Opin
24:275–286
Larsen PJ, Tang-Christensen M, Holst JJ, Ørskov C (1997a) Distribution of glucagon-like peptide-
1 and other preproglucagon-derived peptides in the rat hypothalamus and brainstem. Neuro-
science 77:257–270
Larsen PJ, Tang-Christensen M, Jessop DS (1997b) Central administration of glucagon-like
peptide-1 activates hypothalamic neuroendocrine neurons in the rat. Endocrinology
138:4445–4455
Ling Z, Wu D, Zambre Y, Flamez D, Drucker DJ, Pipeleers DG, Schuit FC (2001) Glucagon-like
peptide 1 receptor signaling influences topography of islet cells in mice. Virchows Arch
438:382–387
Linnebjerg H, Park S, Kothare PA, Trautmann ME, Mace K, Fineman M, Wilding I, Nauck M,
Horowitz M (2008) Effect of exenatide on gastric emptying and relationship to postprandial
glycemia in type 2 diabetes. Regul Pept 151:123–129
Ma X, Bruning J, Ashcroft FM (2007) Glucagon-like peptide 1 stimulates hypothalamic
proopiomelanocortin neurons. J Neurosci 27:7125–7129
Madsbad S (2009) Exenatide and liraglutide: different approaches to develop GLP-1 receptor
agonists (incretin mimetics) - preclinical and clinical results. Best Pract Res Clin Endocrinol
Metab 23:463–477
Madsbad S, Kielgast U, Asmar M, Deacon C, Torekov SS, Holst JJ (2011) An overview of once-
weekly GLP-1 receptor agonists - available efficacy and safety data and perspectives for the
future. Diabetes Obes Metab 13:394–407
Meeran K, O’Shea D, Edwards CM, Turton MD, Heath MM, Gunn I, Abusnana S, Rossi M, Small
CJ, Goldstone AP, Taylor GM, Sunter D, SteereJ CSJ, Ghatei MA, Bloom SR (1999) Repeated
intracerebroventricular administration of glucagon-like peptide-1-(7–36) amide or exendin-
(9–39) alters body weight in the rat. Endocrinology 140:244–250
Meier JJ, Gallwitz B, Schmidt WE, Nauck MA (2002) Glucagon-like peptide 1 as a regulator of
food intake and body weight: therapeutic perspectives. Eur J Pharmacol 440:269–279
Meier JJ, Gallwitz B, Salmen S, Goetze O, Holst JJ, Schmidt WE, Nauck MA (2003a) Normaliza-
tion of glucose concentrations and deceleration of gastric emptying after solid meals during
intravenous glucagon-like peptide 1 in patients with type 2 diabetes. J Clin Endocrinol Metab
88:2719–2725
204 B. Gallwitz
Meier S, H
ucking K, Ritzel R, Holst JJ, Schmiegel WH, Nauck MA (2003b) Absence of a memory
effect for the insulinotropic action of glucagon-like peptide 1 (GLP-1) in healthy volunteers.
Horm Metab Res 35:551–556
Meier JJ, Kemmeries G, Holst JJ, Nauck MA (2005) Erythromycin antagonizes the deceleration of
gastric emptying by glucagon-like peptide 1 and unmasks its insulinotropic effect in healthy
subjects. Diabetes 54:2212–2218
Meneilly GS, Greig N, Tildesley H, Habener JF, Egan JM, Elahi D (2003) Effects of 3 months of
continuous subcutaneous administration of glucagon-like peptide 1 in elderly patients with
type 2 diabetes. Diabetes Care 26:2835–2841
Nagai K, Tsuchiya K, Ezaki T, Tsuchiya M, Ohgawara H (2004) Effect of GLP-1 (glucagon-like
peptide 1:7–36 amide) on porcine pancreatic endocrine cell proliferation and insulin secretion.
Pancreas 28:138–145
Nakade Y, Tsukamoto K, Pappas TN, Takahashi T (2006) Central glucagon like peptide-1 delays
solid gastric emptying via central CRF and peripheral sympathetic pathway in rats. Brain Res
1111:117–121
N
aslund E, Barkeling B, King N, Gutniak M, Blundell JE, Holst JJ, R
ossner S, Hellstr
om PM
(1999) Energy intake and appetite are suppressed by glucagon-like peptide-1 (GLP-1) in obese
men. Int J Obes Relat Metab Disord 23:304–311
N
aslund E, King N, Mansten S, Adner N, Holst JJ, Gutniak M, Hellstr
om PM (2004) Prandial
subcutaneous injections of glucagon-like peptide-1 cause weight loss in obese human subjects.
Br J Nutr 91:439–446
Nauck M, St
ockmann F, Ebert R, Creutzfeldt W (1986) Reduced incretin effect in type 2 (non-
insulin-dependent) diabetes. Diabetologia 29:46–52
Nauck MA, Kleine N, Ørskov C, Holst JJ, Willms B, Creutzfeldt W (1993) Normalization of
fasting hyperglycaemia by exogenous glucagon-like peptide 1 (7–36 amide) in type 2 (non-
insulin-dependent) diabetic patients. Diabetologia 36:741–744
Nauck MA, Niedereichholz U, Ettler R, Holst JJ, Ørskov C, Ritzel R, Schmiegel WH (1997)
Glucagon-like peptide 1 inhibition of gastric emptying outweighs its insulinotropic effects in
healthy humans. Am J Physiol 273:E981–E988
Ørskov C, Holst JJ, Poulsen SS, Kirkegaard P (1987) Pancreatic and intestinal processing of
proglucagon in man. Diabetologia 30:874–881
Ørskov C, Poulsen SS, Moller M, Holst JJ (1996) Glucagon-like peptide I receptors in the
subfornical organ and the area postrema are accessible to circulating glucagon-like peptide I.
Diabetes 45:832–835
Patrita A, Aisa M, Annetti C, Sidoni A, Galli F, Ferri I, Gulla
`N, Donini A (2007) How the hindgut
can cure type 2 diabetes. Ileal transposition improves glucose metabolism and b-cell function
in Goto-kakizaki rats through enhanced proglucagon gene expression and L-cell number.
Surgery 142:74–85
Pellissier S, Sasaki K, Le-Nguyen D, Bataille D, Jarrousse C (2004) Oxyntomodulin and glicentin
are potent inhibitors of the fed motility pattern in small intestine. Neurogastroenterol Motil
16:455–463
Peterli R, W
olnerhanssen B, Peters T, Devaux N, Kern B, Christoffel-Courtin C, Drewe J, von
Fl
ue M, Beglinger C (2009) Improvement in glucose metabolism after bariatric surgery:
comparison of laparoscopic Roux-en-Y gastric bypass and laparoscopic sleeve gastrectomy:
a prospective randomized trial. Ann Surg 250:234–241
Pitomobo C (2008) Central nervous system regulation and hormonal signaling. In: Pitomobo C,
Jones K, Higa K, Pareja C (eds) Obesity surgery: principles and practice. McGraw-Hill, New
York, NY
Raufman JP, Singh L, Singh G, Eng J (1992) Truncated glucagon-like peptide-1 interacts with
exendin receptors on dispersed acini from guinea pig pancreas. Identification of a mammalian
analogue of the reptilian peptide exendin-4. J Biol Chem 267:21432–21437
Reimer RA (2006) Meat hydrolysate and essential amino acid-induced glucagon-like peptide-1
secretion, in the human NCI-H716 enteroendocrine cell line, is regulated by extracellular
Anorexigenic Effects of GLP-1 and Its Analogues 205
signal-regulated kinase1/2 and p38 mitogen-activated protein kinases. J Endocrinol
191:159–170
Rosenstock J, Reusch J, Bush M, Yang F, Stewart M (2009) Potential of albiglutide, a long-acting
GLP-1 receptor agonist, in type 2 diabetes: a randomized controlled trial exploring weekly,
biweekly, and monthly dosing. Diabetes Care 32:1880–1886
Rubino F, Gagner M, Gentileschi P, Kini S, Fukuyama S, Feng J, Diamond E (2004) The early
effect of the Roux-en-Y gastric bypass on hormones involved in body weight regulation and
glucose metabolism. Ann Surg 240:236–242
Salehi M, Vahl TP, D’Alessio DA (2008) Regulation of islet hormone release and gastric emptying
by endogenous GLP-1 following glucose ingestion. J Clin Endocrinol Metab 93:4909–4916
Schirra J, Katschinski M, Weidmann C, Sch
afer T, Wank U, Arnold R, G
oke B (1996) Gastric
emptying and release of incretin hormones after glucose ingestion in humans. J Clin Invest
97:92–103
Schirra J, Kuwert P, Wank U, Leicht P, Arnold R, G
oke B, Katschinski M (1997) Differential
effects of subcutaneous GLP-1 on gastric emptying, antroduodenal motility, and pancreatic
function in men. Proc Assoc Am Physicians 109:84–97
Schirra J, Nicolaus M, Roggel R, Katschinski M, Storr M, Woerle HJ, G
oke B (2006) Endogenous
glucagon-like peptide 1 controls endocrine pancreatic secretion and antro-pyloro-duodenal
motility in humans. Gut 55:243–251
Schmidtler J, Dehne K, Allescher HD, Schusdziarra V, Classen M, Holst JJ, Polack A, Schepp W
(1994) Rat parietal cell receptors for GLP-1-(7–36) amide: Northern blot, cross-linking, and
radioligand binding. Am J Physiol 267:G423–G432
Schnabel CA, Wintle M, Kolterman O (2006) Metabolic effects of the incretin mimetic exenatide
in the treatment of type 2 diabetes. Vasc Health Risk Manag 2:69–77
Schwartz MW, Woods SC, Porte D Jr, Seeley RJ, Baskin DG (2000) Central nervous system
control of food intake. Nature 404:661–671
Seeley RJ, Blake K, Rushing PA, Benoit S, Eng J, Woods SC, D’Alessio D (2000) The role of CNS
glucagon-like peptide-1 (7–36) amide receptors in mediating the visceral illness effects of
lithium chloride. J Neurosci 20:1616–1621
Seo S, Ju S, Chung H, Lee D, Park S (2008) Acute effects of glucagon-like peptide-1 on
hypothalamic neuropeptide and AMP activated kinase expression in fasted rats. Endocr J
55:867–874
Shin YK, Martin B, Golden E, Dotson CD, Maudsley S, Kim W, Jang HJ, Mattson MP, Drucker
DJ, Egan JM, Munger SD (2008) Modulation of taste sensitivity by GLP-1 signaling.
J Neurochem 106:455–463
Shughrue PJ, Lane MV, Merchenthaler I (1996) Glucagon-like peptide-1 receptor (GLP1-R)
mRNA in the rat hypothalamus. Endocrinology 137:5159–5162
Silvestre RA, Rodrı
´guez-Gallardo J, Egido EM, Marco J (2003) Interrelationship among insulin,
glucagon and somatostatin secretory responses to exendin-4 in the perfused rat pancreas. Eur J
Pharmacol 469:195–200
Sokos GG, Nikolaidis LA, Mankad S, Elahi D, Shannon RP (2006) Glucagon-like peptide-1
infusion improves left ventricular ejection fraction and functional status in patients with
chronic heart failure. J Card Fail 12:694–699
St Onge EL, Miller SA (2011) Albiglutide: a new GLP-1 analog for the treatment of type
2 diabetes. Expert Opin Biol Ther 10:801–806
Szayna M, Doyle ME, Betkey JA, Holloway HW, Spencer RG, Greig NH, Egan JM (2000)
Exendin-4 decelerates food intake, weight gain, and fat deposition in Zucker rats. Endocrinol-
ogy 141:1936–1941
Tang-Christensen M, Larsen PJ, G
oke R, Fink-Jensen A, Jessop DS, Møller M, Sheikh SP (1996)
Central administration of GLP-1-(7–36) amide inhibits food and water intake in rats. Am J
Physiol 271:R848–R856
Tang-Christensen M, Vrang N, Larsen PJ (1998) Glucagon-like peptide 1(7–36) amide’s central
inhibition of feeding and peripheral inhibition of drinking are abolished by neonatal
monosodium glutamate treatment. Diabetes 47:530–537
206 B. Gallwitz
Thibaudeau K, Robitaille M, Wen S et al (2006) CJC-1134-PC: an exendin-4 conjugate with
extended pharmacodynamic profiles in rodents. Diabetes 55(suppl 1):A103
Thomas S, Schauer P (2010) Bariatric surgery and the gut hormone response. Nutr Clin Pract
25:175–182
Tsch
op M, Castaneda T, Joost HG, Th
one-Reineke C, Ortmann S, Klaus S, Hagan HH, Chandler
PC, Oswald KD, Benoit SC, Seeley RJ, Kinzig KP, Moran TH, Beck-Sickinger AG, Koglin N,
Rodgers RJ, Blundell JE, Ishii Y, Beattie AH, Holch P, Allison DB, Raun K, Madsen K, Wulff
BS, Stidsen CE, Birringer M, Kreuzer O, Schindler M, Arndt K, Rudolf K, Mark M, Deng XY,
Withcomb DC, Halem H, Taylor J, Dong J, Datta R, Culler M, Craney S, Flora D, Smiley D,
Heiman ML (2004) Does gut hormone PYY3-36 decrease food intake in rodents? Nature
430:165–166
Turton MD, O’Shea D, Gunn I, Beak SA, Edwards CM, Meeran K, Choi SJ, Taylor GM, Heath
MM, Lambert PD, Wilding JP, Smith DM, Ghatei MA, Herbert J, Bloom SR (1996) A role for
glucagon-like peptide-1 in the central regulation of feeding. Nature 379:69–72
Van Dijk G, Thiele TE, Donahey JC, Campfield LA, Smith FJ, Burn P, Bernstein IL, Woods SC,
Seeley RJ (1996) Central infusions of leptin and GLP-1-(7–36) amide differentially stimulate
c-FLI in the rat brain. Am J Physiol 271:R1096–R1100
Vella A, Bock G, Giesler PD, Burton DB, Serra DB, Saylan ML, Dunning BE, Foley JE, Rizza
RA, Camilleri M (2007) Effects of dipeptidyl peptidase-4 inhibition on gastrointestinal func-
tion, meal appearance, and glucose metabolism in type 2 diabetes. Diabetes 56:1475–1480
Verdich C, Flint A, Gutzwiller JP, N
aslund E, Beglinger C, Hellstr
om PM, Long SJ, Morgan LM,
Holst JJ, Astrup A (2001) A meta-analysis of the effect of glucagon-like peptide-1 (7–36)
amide on ad libitum energy intake in humans. J Clin Endocrinol Metab 86:4382–4389
Vetter ML, Cardillo S, Rickels MR, Iqbal N (2009) Narrative review: effect of bariatric surgery on
type 2 diabetes mellitus. Ann Intern Med 150:94–103
Vrang N, Phifer CB, Corkern MM, Berthoud HR (2003) Gastric distension induces c-Fos in
medullary GLP-1/2-containing neurons. Am J Physiol Regul Integr Comp Physiol 285:
R470–R478
Wang GJ, Tomasi D, Backus W, Wang R, Telang F, Geliebter A, Korner J, Bauman A, Fowler JS,
Thanos PK, Volkow ND (2008) Gastric distention activates satiety circuitry in the human
brain. Neuroimage 39:1824–1831
Wang M, Matheson S, Picard J, Pezzullo J, Ulich T (2009) PC-DAC (TM): exendin-4 (CJC-1134-
PC) significantly reduces HbA1c and body weight as an adjunct therapy to metformin: two
randomized, double-blind, placebo-controlled, 12 week, phase II studies in patients with type
2 diabetes mellitus. Diabetes 58(suppl 1):A148
Wellendorph P, Johansen LD, Brauner-Osborne H (2009) Molecular pharmacology of promiscu-
ous seven transmembrane receptors sensing organic nutrients. Mol Pharmacol 76:453–465
Wettergren A, Wøjdemann M, Meisner S, Stadil F, Holst JJ (1997) The inhibitory effect of
glucagon-like peptide-1 (GLP-1) 7–36 amide on gastric acid secretion in humans depends on
an intact vagal innervation. Gut 40:597–601
Young AA, Gedulin BR, Bhavsar S, Bodkin N, Jodka C, Hansen B, Denaro M (1999) Glucose-
lowering and insulin-sensitizing actions of exendin-4: studies in obese diabetic (ob/ob, db/db)
mice, diabetic fatty Zucker rats, and diabetic rhesus monkeys (Macacamulatta). Diabetes
48:1026–1034
Zander M, Madsbad S, Madsen JL, Holst JJ (2002) Effect of 6-week course of glucagon-like
peptide 1 on glycaemic control, insulin sensitivity, and b-cell function in type 2 diabetes:
a parallel-group study. Lancet 359:824–830
Anorexigenic Effects of GLP-1 and Its Analogues 207
... In humans, both GLP-1 and GLP-2 enhance satiety and reduce food intake. GLP-1 stimulates insulin secretion from the pancreas and inhibits glucagon secretion, lowering blood glucose levels, and GLP-2 stimulates growth and repair of the intestinal lining and increases the absorption of nutrients from the intestines [106,111,112]. Therefore, we would have expected the precursor gcga levels to be higher in the postprandial fish, which was not a clear pattern in this trial either per diet or in general; the expression of gcga was in the same ranges in feeding and postprandial fish with few significant differences both in midgut and hindgut. ...
... It may be that the anorexic signals already were sent and gcg production was reduced because of the finalization of the meal. It may also be that the active role of Glp-1 and Glp-2 in the enhancement of satiety and reduction of food intake is not the same in salmon as in humans [106,111,112]. The upregulation of several NPY neurons from GHRL and low glucose is counteracted by leptin and insulin downregulating the same neurons [85]. ...
Article
Full-text available
Whitefish fisheries’ side-stream biomass is an abundant underutilized resource that can be valorized to benefit future aquaculture sustainability. Four novel ingredients based on side-streams from Atlantic cod (Gadus morhua) fileting were produced. FM-hb, a fish meal (FM), and FPH-hb, a fish protein hydrolysate based on heads (h) and backbones (b); FM-hbg, a FM based on heads, backbones, and viscera/guts (g); and FPC-g, a fish protein concentrate based on viscera preserved in formic acid. Four diets were prepared containing one of the ingredients replacing 50% of the dietary FM protein, in addition to a positive (FM10) and a negative (FM5) control. The six diets were fed to triplicate tanks with Atlantic salmon (Salmo salar L.; 113 ± 1 g) over 8 weeks. Besides general performance, gut and brain gene expression for selected hormones and key neuropeptides involved in the control of appetite and digestive processes were studied during feeding and postprandial, and possible reference levels for Atlantic salmon were established. All side-stream-added diets performed well, with no significant differences in performance and biometrics between the treatments. Some gene expression differences were observed, but no well-defined patterns emerged supporting clear dietary effects related to digestive performance or appetite. However, in the brain, a short-time upregulation of agouti-related protein-1 (agrp1), corresponded to higher cumulative feed intake (FI) for the FM10 diet supporting notions that this may be a candidate biomarker for appetite in salmon. Expression of stomach ghrelin-1 (ghrl1) was higher than ghrelin-2 (ghrl2) and membrane-bound O-acyltransferase domain-containing 4 (mboat4), and midgut peptide YYa-2 (pyya2) and glucagon-a (gcga) were higher than peptide YYb-1 (pyyb1). A comparison showed that midgut peptide YYa-1 (pyya1), pyya2, and gcga expressions were higher than in the hindgut, which is opposite of what is found in mammals. In conclusion, this study shows that sustainable side-stream raw materials with different characteristics can partly replace high-quality commercial FMs giving similar performance.
... 44 Pro-opiomelanocortin (POMC) neurons are stimulated electrically by GLP-1 via the protein kinase A pathway and a gradual increase in L-type calcium currents, so the role of GLP-1 in anorexia may be related to the arcuate nucleus (ARC) of the hypothalamus. 45 Following thylakoid consumption, an increase in leptin levels is also observed. 18 Leptin affects the transcription of POMC, whose alpha melanocyte stimulating hormone product is released into the synapse to activate neurons via binding to the melanocortin receptor (MCR), which suppresses appetite; also, leptin inhibits the synthesis and release of NPY in the ARC, which regulates appetite and metabolism. ...
Article
Context It has been suggested that thylakoid supplementation could be associated with favorable changes in appetite. However, the present trials are inconsistent. Objective This systematic review and meta-analysis aimed to investigate whether thylakoid supplementation is effective in hunger/fullness changes. Data Sources A comprehensive literature search was performed before conducting a systematic search in PubMed, Scopus, and ISI Web of Sciences databases up to May 2023. Data Extraction Of the 262 articles found, 5 articles that measured hunger fullness expressed as ratings on the visual analog scale (VAS) for appetite analyses were selected and thoroughly examined. Relevant information, such as the authors' names, year of publication, country of origin, study design, number and status of participants, intervention dosage and type, study duration, assay used, and key findings, was extracted from these articles. The Jadad scale was used to evaluate the quality of studies. Data Analysis The data were obtained using STATA version 13 (StataCorp). Effect sizes were reported as weighted mean differences (WMDs) and 95% confidence intervals (CIs). Subgroup analyses were performed to estimate whether the effects of thylakoid supplementation on hunger and fullness varied according to dose, intervention duration, baseline population age and body mass index, baseline hunger and fullness, total sample size, and study quality. Fractional polynomial modeling was carried out to estimate time- and dose-response meta-analysis of the association between thylakoid and satiety and fullness. The meta-analysis also suggests a significant association between the combination of thylakoids with meals and reduced hunger (WMD: –2.415 mm; 95% CI: –3.544, –1.287; P < 0.001) and increased fullness (WMD: +4.602 mm; 95% CI: 2.356, 6.848; P < 0.001). Further analysis showed that thylakoid supplementation with 2 doses of 5 g/day (P = 0.002) and 7.4 g/day (P = 0.021) is more effective in reducing the feeling of hunger during an intervention of less than 56 days (P < 0.001) and with a lower level of hunger (VAS <45 mm, P < 0.001). Furthermore, a significant increase in the perception of fullness was found at a duration of less than 84 days (P < 0.001) and baseline fullness of more than 45 mm (P < 0.001). Conclusions Supplementation with thylakoid shows favorable effects on reducing hunger and increasing fullness during a shorter intervention duration. Further trials are required to shed light on this relationship. Systematic Review Registration PROSPERO registration no. CRD42023432429.
... Its plasma concentrations increase after food intake, acting as a hormone with different metabolic effects such as insulin secretion and decrease of gastric emptying (63,64). GLP-1 in the gastrointestinal tract slows gastric emptying after a meal, promoting a feeling of fullness and a reduction of appetite; also its receptors on hypothalamic neurons stimulate satiety (65). It has been seen that the mechanism of satiety of GPL-1 is due to its effects on the central nervous system; GLP-1 receptors are present in the nucleus of the solitary tract and arcuate region, where they receive afferent input from the vagus and glossopharyngeal nerves and integrate both neural and humoral factors (66). ...
Article
Full-text available
Non-alcoholic fatty liver disease (NAFLD) is a highly prevalent metabolic disease, although prevalence could change according to region, nowadays is considered a public health problem whose real impact on the health system is unknown. NAFLD has a multifactorial and complex pathophysiology, due to this, developing a unique and effective pharmacological treatment has not been successful in reverting or avoiding the progression of this liver disease. Even though NAFLD pathophysiology is known, all actual treatments are focused on modifying or regulating the metabolic pathways, some of which interplay with obesity. It has been known that impairments in hunger and satiety signals are associated with obesity, however, abnormalities in these signals in patients with NAFLD and obesity are not fully elucidated. To describe these mechanisms opens an additional option as a therapeutic target sharing metabolic pathways with NAFLD, therefore, this review aims to describe the hormones and peptides implicated in both hunger-satiety in NAFLD. It has been established that NAFLD pharmacological treatment cannot be focused on a single purpose; hence, identifying interplays that lead to adding or modifying current treatment options could also have an impact on another related outcome such as hunger or satiety signals.
... This incretin peptide also controls glucagon, gastric emptying, and glucose-dependent insulin secretion. Studies have demonstrated that GLP-1 receptors in the brainstem and hypothalamus were necessary for the hormone's anorexigenic and glucoregulatory actions (Gallwitz, 2012). GLP-1 has functions other than regulating glucose and food intake, and that these functions likely include reward control.GLP-1 receptors are found throughout the mesolimbic system and that neurons expressing GLP-1 directly project to the VTA and NAc. ...
Article
Full-text available
Background: Substance abuse has become a serious issue that is faced by the world. The effects of drug abuse can be detrimental to corporeal, psychological and societal well-being of a person. Drug misuse is actually unrestrained habit of drugs leading to inspiration of reward-system in brain Substance abuse also influences the food preferences and dietary habits of the users, therefore affecting the person’s nutrition. Instead of how frequently a person engages in the behavior, substance abuse and substance addiction are distinguished by how challenging it is for the person to manage without the behavior or quit it for a specific period of time. Methodology: Electronic databases were used to conduct a thorough, systematic analysis of research, which was restricted to those with English/English abstracts and used words like "addiction," "nutrition," "lifestyle," "alcohol," "reward system," "recreational substances," etc., Conclusion: An effort has been made in this research article to highlight the effects of various addictive substances on different systems in human body with the role of adequate nutrition to fight back this addiction for the overall betterment of the society.
... Among pleiotropic effects of gastrointestinal hormones and especially GLP-1, it is important to report the emerging role of GLP-1 as anorexigenic and neuroprotective hormone [43][44][45]. Currently, other novel peptide hormones have been discovered. ...
Article
Full-text available
Glycemic homeostasis is an essential mechanism for the proper working of an organism. However, balance in blood lipid and protein levels also plays an important role. The discovery of the hormone insulin and the description of its function for glycemic control made fundamental scientific progress in this field. However, since then our view of the problem has been deeply influenced only in terms of glucose and insulin (in an insulin-centric and glucose-centric way). Based on recent scientific discoveries, a fine and sophisticated network of hormonal and metabolic interactions, involving almost every apparatus and tissue of the human body, has been theorized. Efficient metabolic homeostasis is founded on these intricate interactions. Although it is still not fully defined, this complex network can undergo alterations that lead to metabolic disorders such as diabetes mellitus (DM). The endocrine pancreas plays a crucial role in the metabolic balance of an organism, but insulin is just one of the elements involved and each single pancreatic islet hormone is worthy of our concern. Moreover, pancreatic hormones need to be considered in a general view, concerning both their systemic function as direct mediators and as hormones, which, in turn, are regulated by other hormones or other substances. This more complex scenario should be taken into account for a better understanding of the pathophysiology and the therapeutic algorithms of DM. As a consequence, improvements in modern medicine could help to contemplate this new perspective. This review is focused on some aspects of gut-pancreas interaction, aiming to integrate this synergy into a wider context involving other organs and tissues.
... These findings are interesting and do not exclude that chronic ingestion of these sweeteners may play a role in the regulation of glucose homeostasis. Also, the increase in GLP-1 levels may have relevant clinical implications beyond the insulinotropic action of GLP-1, considering the well-known role exerted by GLP-1 receptor agonists in the reduction of cardiovascular risk among diabetic patients, along with the anorexigenic properties of GLP-1 and its analogs (75)(76)(77). Remarkably, CCK has also been shown to induce short-term satiety and to play a role in the regulation of insulin secretion and overall β-cell function and survival, displaying complementary biological actions with those exerted by GLP-1 (78). ...
Article
Full-text available
Since excessive sugar consumption has been related to the development of chronic metabolic diseases prevalent in the western world, the use of sweeteners has gradually increased worldwide over the last few years. Although low-and non-calorie sweeteners may represent a valuable tool to reduce calorie intake and prevent weight gain, studies investigating the safety and efficacy of these compounds in the short-and long-term period are scarce and controversial. Therefore, future studies will need to elucidate the potential beneficial and/or detrimental effects of different types of sweeteners on metabolic health (energy balance, appetite, body weight, cardiometabolic risk factors) in healthy subjects and patients with diabetes, obesity and metabolic syndrome. In this regard, the impact of different sweeteners on central nervous system, gut hormones and gut microbiota is important, given the strong implications that changes in such systems may have for human health. The aim of this narrative review is to summarize the current evidence for the neuroendocrine and metabolic effects of sweeteners, as well as their impact on gut microbiota. Finally, we briefly discuss the advantages of the use of sweeteners in the context of very-low calorie ketogenic diets.
Article
Background Glucagon-like peptide-1 (GLP-1) agonists, such as exenatide, liraglutide, dulaglutide, semaglutide, and tirzepatide, effectively manage type 2 diabetes by promoting insulin release, suppressing glucagon secretion, and enhancing glucose metabolism. They also aid weight reduction and cardiovascular health, potentially broadening their therapeutic scope. In plastic surgery, they hold promise for perioperative weight management and glycemic control, potentially impacting surgical outcomes. Methods A comprehensive review was conducted to assess GLP-1 agonists' utilization in plastic surgery. We analyzed relevant studies, meta-analyses, and trials to evaluate their benefits and limitations across surgical contexts, focusing on weight reduction, glycemic control, cardiovascular risk factors, and potential complications. Results Studies demonstrate GLP-1 agonists' versatility, spanning weight management, cardiovascular health, neurological disorders, and metabolic dysfunction-associated liver diseases. Comparative analyses highlight variations in glycemic control, weight loss, and cardiometabolic risk. Meta-analyses reveal significant reductions in hemoglobin A1C levels, especially with high-dose semaglutide (2 mg) and tirzepatide (15 mg). However, increased dosing may lead to gastrointestinal side effects and serious complications like pancreatitis and bowel obstruction. Notably, GLP-1 agonists' efficacy in weight reduction and glycemic control may impact perioperative management in plastic surgery, potentially expanding surgical candidacy for procedures like autologous flap-based breast reconstruction and influencing outcomes related to lymphedema. Concerns persist regarding venous thromboembolism and delayed gastric emptying, necessitating further investigation into bleeding and aspiration risk with anesthesia. Conclusions GLP-1 agonists offer advantages in perioperative weight management and glycemic control in plastic surgery patients. They may broaden surgical candidacy and mitigate lymphedema risk but require careful consideration of complications, particularly perioperative aspiration risk. Future research should focus on their specific impacts on surgical outcomes to optimize their integration into perioperative protocols effectively. Despite challenges, GLP-1 agonists promise to enhance surgical outcomes and patient care in plastic surgery.
Article
Physical activity contributes up to 30% of the total daily energy expenditure of domestic cats (Felis catus) and, therefore, can play an important role in the prevention and reduction of obesity and may impact macronutrient digestibility. An age-related decline in physical activity in domestic cats has been observed, however, scientific data is scarce. The aim of this study was to examine the relationship between physical activity, cat age and body condition score. A secondary aim was to determine whether physical activity level influenced apparent macronutrient digestibility. Using accelerometers, the physical activity of 138 domestic shorthair cats grouped-housed in identical outdoor pens was measured. Cats were divided into five age groups: kitten (n=10), junior (n=37), prime (n=30), mature (n=33) or senior (n=27), and physical activity data was collected from each cat for seven consecutive days. One cat was randomly chosen from each age group as a control and its physical activity was measured for the entire 11-week period. Body condition score of the cats was assessed in the same week that physical activity data was collected. A decline in physical activity with age was found, although this decline was not linear. Cats in the kitten and junior age groups had the highest physical activity. Compared with the younger age groups cats in the prime age group showed a significant reduction physical activity, which then remained constant in the mature and senior age groups. An overall decline in physical activity with an increasing body condition score was found. To investigate whether physical activity influenced apparent macronutrient digestibility, a subset of six cats from the junior and prime age groups were selected at the end of the activity assessment period. Cats were individually housed, fed a commercially available wet canned diet, and had ad libitum access to water. No effect of physical activity on apparent macronutrient digestibility was found.
Article
Aims We investigated if individuals with higher emotional eating scores are less sensitive to the effects of a GLP-1RA on central responses to food cues. Additionally, we investigated the associations of higher external and restraint eating scores with the sensitivity to the central effects of GLP-1RA. Methods This secondary analysis of a randomized crossover study in people with obesity and type 2 diabetes, consisted of two periods of 12-week treatment with liraglutide or insulin glargine. Using functional MRI, we assessed the relation between baseline eating behavior and the effects of the GLP-1RA liraglutide compared with insulin after 10 days and 12 weeks of treatment on brain responses to food cues. Results After 10 days, higher emotional eating scores were associated with less pronounced GLP-1RA induced reductions in brain responses to food pictures in the amygdala, insula and caudate nucleus. In addition, higher emotional eating scores tended to be associated with less pronounced GLP-1RA increases in brain responses to chocolate milk receipt in the caudate nucleus and insula. After 12 weeks, there were no significant associations between emotional eating scores and liraglutide-induced changes in brain responses to food cues. After 10 days, baseline external eating scores were associated with less pronounced GLP-1RA induced reductions in brain responses to food pictures in the insula, amygdala and orbitofrontal cortex. After 12 weeks, baseline restraint eating scores were associated with more GLP-1RA induced reductions in brain responses to food pictures in the insula and caudate nucleus, and with more GLP-1RA induced reductions in brain responses to the anticipation of chocolate milk in the caudate nucleus. Conclusions Our findings indicate that individuals with higher baseline emotional eating scores are less sensitive to the central effect of GLP-1RA treatment. Additionally, external eating may also decrease, whereas restraint eating may increase the sensitivity to the treatment effects of GLP-1RAs. These insights may help to optimize treatment strategies for obesity and to select patient groups with better efficacy of GLP-1RA treatment.
Article
Full-text available
To examine the influence of the putative satiety factor (GLP-1) on the hypothalamo-pituitary-gonadal axis, we used GT1-7 cells as a model of neuronal luteinizing hormone- releasing hormone (LHRH) release. GLP-1 caused a concentration-dependent increase in LHRH release from GT1-7 cells. Specific, saturable GLP-1 binding sites were demonstrated on these cells. The binding of [125I]GLP-1 was time-dependent and consistent with a single binding site (Kd = 0.07+/-0.016 nM; binding capacity = 160+/-11 fmol/mg protein). The specific GLP-1 receptor agonists, exendin-3 and exendin-4, also showed high affinity (Ki = 0.3+/-0.05 and 0.32+/-0.06 nM, respectively) as did the antagonist exendin-(9-39) (Ki = 0.98+/-0.24 nM). At concentrations that increased LHRH release, GLP-1 (0.5-10 nM) also caused an increase in intracellular cAMP in GT1-7 cells (10 nM GLP-1: 7.66+/-0.4 vs. control: 0.23+/-0.02 nmol/mg protein; P < 0.001). Intracerebroventricular injection of GLP-1 at a single concentration (10 microg) produced a prompt increase in the plasma luteinizing hormone concentration in male rats (GLP-1: 1.09+/-0.11 vs. saline: 0.69+/-0.06 ng/ml; P < 0.005). GLP-1 levels in the hypothalami of 48-h-fasted male rats showed a decrease, indicating a possible association of the satiety factor with the low luteinizing hormone levels in animals with a negative energy balance.
Article
Glucagon-like peptide-I(7-37) [GLP-I(7-37)] is an intestinal peptide hormone that is released in response to oral nutrients and that potently augments glucose-mediated insulin secretion. GLP-I(7-37) has potent insulin-releasing activities in vivo in response to oral nutrients, in situ in the isolated perfused pancreas, and in vitro in cultured pancreatic B-cells. As such GLP-I(7-37) is a potent hormonal mediator in the enteroinsular axis involved in the regulation of glucose homeostasis. We now show that in addition to stimulating the release of insulin, GLP-I(7-37) stimulates proinsulin gene expression at the levels of gene transcription and cellular levels of proinsulin messenger RNA as well as the translational biosynthesis of proinsulin. These findings of the positive anabolic actions of GLP-I(7-37) on the synthesis of insulin in B-cells support the notion that GLP-I(7-37) may be of therapeutic use in stimulating the production of insulin in patients with noninsulin-dependent diabetes mellitus and tha...
Article
To clarify catecholaminergic systems on the regulation of food intake in the neonatal chick, we examined the effects of intracerebroventricular (ICV) injection of prazosin (α1-adrenoceptor antagonist), yohimbine (α2-adrenoceptor antagonist) and benserazide (an inhibitor of L-aromatic amino acid decarboxylase). We found that food intake was significantly suppressed by ICV injection of yohimbine (25 and 50μg) over 60min (P<0.05). Any doses of prazosin (1.25, 2.5 and 5.0μg) did not alter food intake of chicks (P>0.05). ICV administration of benserazide induced a hypophagia after 60min postinjection (P<0.05). It is suggested that catecholaminergic systems play an important role in the neural regulation of food intake in chicks, especially through α2-adrenoceptor.
Article
Introduction: Type 2 diabetes mellitus (T2DM) is an increasing health problem worldwide. Glucagon-like peptide-1 (GLP-1) receptor agonists are an expanding drug class that target several of the pathophysiological traits of T2DM. Lixisenatide is a GLP-1 receptor agonist in development for once-daily treatment of T2DM. Areas covered: Pharmacological, preclinical and clinical evidence demonstrating the applicability of lixisenatide for the treatment of T2DM are reviewed. Available data and pending clinical development are summarized, critically appraised and compared to competitor drugs. The most relevant papers and meeting abstracts published up to November 2010 are used as sources for this review. Expert opinion: Efficacy and safety in T2DM are demonstrated with lixisenatide in monotherapy and in combination with metformin. However, limited data with the intended once-daily 20 μg subcutaneous dosing necessitate further evaluation of lixisenatide as add-on to various antidiabetic treatments. It remains to be established whether the slightly differing chemical properties compared to other GLP-1 receptor agonists including a rather short duration of action will be a disadvantage or maybe even an advantage, for example, when combined with long-acting insulin.